05.02.2013 Views

plant surface microbiology.pdf

plant surface microbiology.pdf

plant surface microbiology.pdf

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

PLANT SURFACE MICROBIOLOGY


Ajit Varma<br />

Lynette Abbott<br />

Dietrich Werner<br />

Rüdiger Hampp (Eds.)<br />

Plant Surface<br />

Microbiology<br />

With 138 Figures, 2 in Color<br />

1 23


Professor Dr. Ajit Varma<br />

Director<br />

Amity Institiute of Microbial Sciences<br />

Amity University<br />

Noida 201303<br />

UP, India<br />

email: ajitvarma@aihmr.amity.edu<br />

Professor Dr. Lynette Abbott<br />

School of Earth and Geographical Sciences<br />

The University of Western Australia<br />

Nedlands WA 6009<br />

Australia<br />

email: labbott@cyllene.uwa.edu.au<br />

ISBN 978-3-540-74050-6<br />

Library of Congress Control Number: 2007934913<br />

Springer-Verlag Berlin Heidelberg New York<br />

Professor Dr. Dietrich Werner<br />

FG Zellbiologie und Angewandte Botanik<br />

Philipps Universität Marburg<br />

35032 Marburg<br />

Germany<br />

email: djg.werner@gmx.de<br />

Professor Dr. Rüdiger Hampp<br />

Physiological Ecology of Plants<br />

University of Tübingen<br />

72116 Tübingen<br />

Germany<br />

email: ruediger.hampp@uni-tuebingen.de<br />

This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned,<br />

specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on<br />

microfilm or in any other way, and storage in data banks. Duplication of this publication or parts thereof is permitted<br />

only under the provisions of the German Copyright Law of September 9, 1965, in its current version, and permissions<br />

for use must always be obtained from Springer-Verlag. Violations are liable for prosecution under the<br />

German Copyright Law.<br />

Springer-Verlag is a part of Springer Science+Business Media<br />

springer.com<br />

© Springer-Verlag Berlin Heidelberg 2004, 2008<br />

The use of general descriptive names, registered names, trademarks, etc. in this publication does not imply, even in<br />

the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations<br />

and therefore free for general use.<br />

5 4 3 2 1 0 – Printed on acid free paper


Preface<br />

The complexity of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> is based on combinations.A large<br />

number of microbial species and genera interact with several hundred thousand<br />

species of higher <strong>plant</strong>s. At the same time, they interact with each other.<br />

Therefore, this book describes only some very important model interactions<br />

which have been studied intensively over the last years.The methods developed<br />

for some important groups of microorganisms can be used for a large number<br />

of other less studied interactions and combinations. The pace of discovery has<br />

been particularly fast at two poles of biological complexity,the molecular events<br />

leading to changes in growth and differentiation, as well as the factors regulating<br />

the structure and diversity of natural populations and communities.<br />

The area of <strong>plant</strong> <strong>surface</strong>s is enormous. A single maize <strong>plant</strong> has a leaf<br />

<strong>surface</strong> of up to 8000 cm 2 , a single beech tree has a leaf <strong>surface</strong> of around<br />

4.5 million cm 2 . The leaf area index (LAI) varies from 0.45 in tundra areas<br />

up to 14 in areas with a dense vegetation. Calculated for all <strong>plant</strong> <strong>surface</strong>s<br />

above ground, the <strong>surface</strong> area is more than 200 million km 2 . This area is still<br />

surpassed by the below ground <strong>surface</strong> areas of <strong>plant</strong>s, especially those with<br />

an extensive root hair system. For a single rye <strong>plant</strong>, a root hair <strong>surface</strong> of<br />

around 400 m 2 has been calculated. Even if this is an exceptional case, it can<br />

be assumed that in many <strong>plant</strong>s the root and root hair <strong>surface</strong> is ten times<br />

larger than the <strong>surface</strong>s of the above ground <strong>plant</strong> parts. This means that<br />

more than 2000 million km 2 of <strong>plant</strong> <strong>surface</strong> is present underground. Taking<br />

both figures together, it exceeds the land <strong>surface</strong> area of the planet Earth of<br />

149 million km 2 by more than a factor of 10.<br />

This volume summarizes and updates both the state of knowledge and theories<br />

and their possible biotechnological applications. It will thus be of interest<br />

to a diverse audience of researchers and instructors, especially biologists,<br />

biochemists, agronomists, foresters, horticulturists, mycologists, soil scientists,<br />

ecologists, <strong>plant</strong> physiologists, <strong>plant</strong> molecular biologists, geneticists,<br />

and microbiologists.<br />

In the planning of the book, invitations for contributions were extended to<br />

leading international scientists working in the field of <strong>plant</strong> <strong>surface</strong> microbi-


VI<br />

ology. The basic concepts in <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> are discussed at<br />

length in 30 chapters including a few specialized and innovative methodologies<br />

and novel techniques. The editors would like to express deep appreciation<br />

to each contributor for his/her work, patience and attention to detail during<br />

the entire production process. It is hoped that their reviews, interpretations,<br />

and basic concepts will stimulate further research. We are confident that the<br />

joint efforts of the authors and editors will contribute to a better understanding<br />

of the advances in the study of the challenging area of <strong>surface</strong> <strong>microbiology</strong><br />

and will further stimulate progress in this field.<br />

It has been a pleasure to edit this book, primarily due to the stimulating<br />

cooperation of the contributors.We would like to express sincere thanks to all<br />

the staff members of Springer-Verlag, Heidelberg, especially, Drs. Dieter<br />

Czeschlik and Jutta Lindenborn for their help and active cooperation during<br />

the preparation of the book.<br />

New Delhi, India Ajit Varma<br />

Nedlands, Australia Lynette Abbott<br />

Marburg, Germany Dietrich Werner<br />

Tübingen, Germany Rüdiger Hampp<br />

July 2003<br />

Preface


Contents<br />

1 The State of the Art . . . . . . . . . . . . . . . . . . . . . . . 1<br />

Ajit Varma, Lynette K. Abbott, Dietrich Werner<br />

and Rüdiger Hampp<br />

Section A<br />

2 Root Colonisation Following Seed Inoculation . . . . . . . 13<br />

Thomas F.C. Chin-A-Woeng and Ben J.J. Lugtenberg<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 13<br />

2 Bacterial Root Colonisation . . . . . . . . . . . . . . . . . . 13<br />

3 Analysis of Tomato Root Tip Colonisation<br />

After Seed Inoculation Using a Gnotobiotic Assay . . . . . . 14<br />

3.1 Description of the Gnotobiotic System . . . . . . . . . . . . 14<br />

3.2 Seed Disinfection . . . . . . . . . . . . . . . . . . . . . . . . 15<br />

3.3 Growth and Preparation of Bacteria . . . . . . . . . . . . . . 16<br />

3.4 Seed Inoculation . . . . . . . . . . . . . . . . . . . . . . . . 17<br />

3.5 Analysis of the Tomato Root Tip . . . . . . . . . . . . . . . . 17<br />

3.6 Confocal Laser Scanning Microscopy . . . . . . . . . . . . . 18<br />

4 Genetic Tools for Studying Root Colonisation . . . . . . . . 18<br />

4.1 Marking and Selecting Bacteria . . . . . . . . . . . . . . . . 18<br />

4.2 Rhizosphere-Stable Plasmids . . . . . . . . . . . . . . . . . 21<br />

4.3 Genetic and Metabolic Burdens . . . . . . . . . . . . . . . . 21<br />

5 Behaviour of Root-Colonising<br />

Pseudomonas Bacteria in a Gnotobiotic System . . . . . . . 22<br />

5.1 Colonisation Strategies of Bacteria . . . . . . . . . . . . . . 22<br />

5.2 Competitive Colonisation Studies . . . . . . . . . . . . . . . 23<br />

5.3 Monocots versus Dicots . . . . . . . . . . . . . . . . . . . . . 25<br />

6 Influence of Abiotic and Biotic Factors . . . . . . . . . . . . 25


VIII<br />

Contents<br />

6.1 Abiotic Factors . . . . . . . . . . . . . . . . . . . . . . . . . 25<br />

6.2 Biotic Factors . . . . . . . . . . . . . . . . . . . . . . . . . . 27<br />

7 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 28<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 28<br />

3 Methanogenic Microbial Communities Associated<br />

with Aquatic Plants . . . . . . . . . . . . . . . . . . . . . . . 35<br />

Ralf Conrad<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 35<br />

2 Role of Plants in Emission of CH 4 to the Atmosphere . . . . 35<br />

3 Role of Photosynthates and Plant Debris for CH 4 Production 38<br />

4 Methanogenic Microbial Communities on Plant Debris . . . 40<br />

5 Methanogenic Microbial Communities on Roots . . . . . . . 42<br />

6 Interaction of Methanogens and Methanotrophs . . . . . . . 44<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 45<br />

4 Role of Functional Groups of Microorganisms<br />

on the Rhizosphere Microcosm Dynamics . . . . . . . . . . 51<br />

Galdino Andrade<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 51<br />

2 General Aspects of Functional Groups<br />

of Soil Microorganisms . . . . . . . . . . . . . . . . . . . . . 52<br />

3 Carbon Cycle Functional Groups . . . . . . . . . . . . . . . 53<br />

4 Functional Groups of Microorganisms of the Nitrogen Cycle 55<br />

5 Functional Groups of Microorganisms of the Sulphur Cycle 57<br />

6 Functional Groups of Microorganisms<br />

of the Phosphorus Cycle . . . . . . . . . . . . . . . . . . . . 59<br />

7 Dynamics of the Rhizosphere Functional Groups<br />

of Microorganisms . . . . . . . . . . . . . . . . . . . . . . . 60<br />

8 Relationship Among r and k Strategist Functional Groups . 61<br />

9 Arbuscular Mycorrhizal Fungi Dynamics<br />

in the Rhizosphere . . . . . . . . . . . . . . . . . . . . . . . 61<br />

10 Dynamics Among the Functional Micro-Organism Groups<br />

of the Carbon, Nitrogen, Phosphorus and Sulphur Cycles . . 65<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 68


Contents IX<br />

5 Diversity and Functions of Soil Microflora<br />

in Development of Plants . . . . . . . . . . . . . . . . . . . . 71<br />

Ramesh Chander Kuhad, David Manohar Kothamasi,<br />

K.K. Tripathi and Ajay Singh<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 71<br />

2 Functional Diversity of Soil Microflora . . . . . . . . . . . . 72<br />

3 Role of Soil Microflora in Plant Development . . . . . . . . 76<br />

3.1 Mycorrhiza . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76<br />

3.2 Actinorhiza . . . . . . . . . . . . . . . . . . . . . . . . . . . 80<br />

3.3 Plant Growth-Promoting Rhizobacteria . . . . . . . . . . . 82<br />

3.4 Phosphate-Solubilizing Microorganisms . . . . . . . . . . . 84<br />

3.5 Lignocellulolytic Microorganisms . . . . . . . . . . . . . . . 85<br />

4 Plant Growth Promoting Substances Produced<br />

by Soil Microbes . . . . . . . . . . . . . . . . . . . . . . . . . 88<br />

5 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 90<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 91<br />

6 Signalling in the Rhizobia–Legumes Symbiosis . . . . . . . 99<br />

Dietrich Werner<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 99<br />

2 The Signals from the Host Plants . . . . . . . . . . . . . . . 101<br />

2.1 Phenylpropanoids: Simple Phenolics, Flavonoids<br />

and Isoflavonoids . . . . . . . . . . . . . . . . . . . . . . . . 102<br />

2.2 Metabolization of Flavonoids and Isoflavonoids . . . . . . . 104<br />

2.3 Vitamins as Growth Factors and Signal Molecules . . . . . . 106<br />

3 Signals from the Microsymbionts . . . . . . . . . . . . . . . 107<br />

3.1 Nod Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . 107<br />

3.2 Cyclic Glucans . . . . . . . . . . . . . . . . . . . . . . . . . . 109<br />

3.3 Lipopolysaccharides . . . . . . . . . . . . . . . . . . . . . . 110<br />

3.4 Exopolysaccharides . . . . . . . . . . . . . . . . . . . . . . . 110<br />

4 Signal Perception and Molecular Biology<br />

of Nodule Initiation . . . . . . . . . . . . . . . . . . . . . . . 111<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 114


X<br />

Section B<br />

Contents<br />

7 The Functional Groups of Micro-organisms Used<br />

as Bio-indicator on Soil Disturbance Caused<br />

by Biotech Products such as Bacillus thuringiensis<br />

and Bt Transgenic Plants . . . . . . . . . . . . . . . . . . . . 121<br />

Galdino Andrade<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 121<br />

2 General Aspects of Bacillus thuringiensis . . . . . . . . . . . 122<br />

3 Survival in the Soil . . . . . . . . . . . . . . . . . . . . . . . 123<br />

4 History of Bacillus thuringiensis-Transgenic Plants . . . . . 124<br />

5 Persistence of the Protein Crystal in the Soil . . . . . . . . . 125<br />

6 Effect of Bacillus thuringiensis and Its Bio-insecticide<br />

Protein on Functional Soil Microorganism Assemblage . . . 126<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 130<br />

8 The Use of ACC Deaminase-Containing<br />

Plant Growth-Promoting Bacteria to Protect Plants<br />

Against the Deleterious Effects of Ethylene . . . . . . . . . 133<br />

Bernard R. Glick and Donna M. Penrose<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 133<br />

2 Ethylene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134<br />

3 ACC Deaminase . . . . . . . . . . . . . . . . . . . . . . . . . 135<br />

3.1 Treatment of Plants with ACC Deaminase<br />

Containing Bacteria . . . . . . . . . . . . . . . . . . . . . . . 137<br />

4 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 140<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 141<br />

9 Interactions Between Epiphyllic Microorganisms<br />

and Leaf Cuticles . . . . . . . . . . . . . . . . . . . . . . . . 145<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 145<br />

2 Physical and Chemical Parameters of the Phyllosphere . . . 147<br />

3 Leaf Surface Colonisation and Species Composition . . . . . 149<br />

4 Alteration of Leaf Surface Wetting . . . . . . . . . . . . . . . 150<br />

5 Interaction of Bacteria with Isolated Plant Cuticles . . . . . 152<br />

6 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 153<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 154


Contents XI<br />

10 Developmental Interactions Between Clavicipitaleans<br />

and Their Host Plants . . . . . . . . . . . . . . . . . . . . . 157<br />

James F. White Jr., Faith Belanger, Raymond Sullivan,<br />

Elizabeth Lewis, Melinda Moy, William Meyer<br />

and Charles W. Bacon<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 157<br />

2 Endophyte/Epibiont Niche . . . . . . . . . . . . . . . . . . . 157<br />

3 Coevolution of Clavicipitalean Fungi with Grass Hosts . . . 158<br />

4 The Jump from Insects to Plants . . . . . . . . . . . . . . . . 158<br />

4.1 Trans-Kingdom Jump . . . . . . . . . . . . . . . . . . . . . . 158<br />

4.2 Intermediate Stages in the Transition to Plants . . . . . . . . 158<br />

4.3 Parasitism of Grass Meristematic Tissues . . . . . . . . . . . 160<br />

5 Developmental Differentiation of Endophytic<br />

and Epiphyllous Mycelium . . . . . . . . . . . . . . . . . . . 160<br />

5.1 Plant Cell Wall Alteration . . . . . . . . . . . . . . . . . . . . 160<br />

5.2 Endophytic Mycelial Growth . . . . . . . . . . . . . . . . . . 160<br />

5.3 Control of Endophytic Mycelial Development . . . . . . . . 163<br />

5.4 Epiphyllous Mycelial Development . . . . . . . . . . . . . . 163<br />

5.5 Expression of Fungal Secreted Hydrolytic<br />

Enzymes in Infected Plants . . . . . . . . . . . . . . . . . . . 164<br />

6 Modifications of Plant Tissues for Nutrient Acquisition . . . 165<br />

6.1 Development of the Stroma in Epichloë . . . . . . . . . . . . 165<br />

6.2 Stroma Development in Myriogenospora . . . . . . . . . . . 166<br />

6.3 Mechanisms for Modifying Plant Tissues . . . . . . . . . . . 168<br />

6.4 Evaporative-Flow Mechanism for Nutrient Acquisition . . . 169<br />

6.5 The Cytokinin Induction Hypothesis . . . . . . . . . . . . . 169<br />

7 Evolution of Asexual Derivatives of Epichloë . . . . . . . . . 171<br />

7.1 Reproduction and Loss of Sexual Reproduction . . . . . . . 171<br />

7.2 The Hypotheses . . . . . . . . . . . . . . . . . . . . . . . . . 172<br />

7.3 The Process of Stroma Development and its Loss . . . . . . 173<br />

7.4 The Shift from Pathogen to Mutualist . . . . . . . . . . . . . 174<br />

8 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 174<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 174<br />

11 Interactions of Microbes with Genetically Modified Plants . 179<br />

Michael Kaldorf, Chi Zhang, Uwe Nehls,<br />

Rüdiger Hampp and François Buscot<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 179<br />

2 Changes in Microbial Communities Induced<br />

by Genetically Modified Plants . . . . . . . . . . . . . . . . . 181


XII<br />

3 Impact of Genetically Modified Plants<br />

on Symbiotic Interactions . . . . . . . . . . . . . . . . . . . 184<br />

4 Horizontal Gene Transfer . . . . . . . . . . . . . . . . . . . . 186<br />

5 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 191<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 192<br />

Section C<br />

Contents<br />

12 Interaction Between Soil Bacteria<br />

and Ectomycorrhiza-Forming Fungi . . . . . . . . . . . . . 197<br />

Rüdiger Hampp and Andreas Maier<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 197<br />

2 Bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198<br />

3 Bacterial Community Structure . . . . . . . . . . . . . . . . 198<br />

4 Association of Bacteria with Fungal/Ectomycorrhizal<br />

Structures . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199<br />

5 Bacteria Associated with Sporocarps and Ectomycorrhiza . 200<br />

6 Benefits from Bacteria/Ectomycorrhiza Interactions . . . . 201<br />

7 Possible Mechanisms of Interaction . . . . . . . . . . . . . . 202<br />

8 Biochemical Evidence for Interaction . . . . . . . . . . . . . 203<br />

9 Impacts of Environmental Pollution . . . . . . . . . . . . . . 206<br />

10 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 206<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 207<br />

13 The Surface of Ectomycorrhizal Roots and the<br />

Interaction with Ectomycorrhizal Fungi . . . . . . . . . . . 211<br />

Ingrid Kottke<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 211<br />

2 Long and Short Roots of Ectomycorrhiza-Forming Plants . 212<br />

3 A Cuticle-Like Layer on the Surface of Short Roots . . . . . 214<br />

4 Involvement of the Cuticle-Like Layer<br />

in Mycorrhiza Formation . . . . . . . . . . . . . . . . . . . . 218<br />

5 Involvement of the Cuticle-Like Layer in Hyphal Attachment 218<br />

6 Digestion of the Suberin Layer and the Cell Wall<br />

of the Root Cap . . . . . . . . . . . . . . . . . . . . . . . . . 220<br />

7 Hartig Net Formation . . . . . . . . . . . . . . . . . . . . . . 221<br />

8 Pectins in the Cortical Cell Walls of Nonmycorrhizal<br />

Long and Mycorrhizal Short Roots . . . . . . . . . . . . . . 222<br />

9 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 223<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 224


Contents XIII<br />

14 Cellular Ustilaginomycete—Plant Interactions . . . . . . . 227<br />

Robert Bauer and Franz Oberwinkler<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 227<br />

2 The Term Smut Fungus . . . . . . . . . . . . . . . . . . . . . 227<br />

3 Life Cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228<br />

4 Hosts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228<br />

5 Cellular Interactions . . . . . . . . . . . . . . . . . . . . . . 229<br />

5.1 Local Interaction Zones . . . . . . . . . . . . . . . . . . . . . 230<br />

5.2 Enlarged Interaction Zones . . . . . . . . . . . . . . . . . . 234<br />

6 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 235<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 236<br />

15 Interaction of Piriformospora indica<br />

with Diverse Microorganisms and Plants . . . . . . . . . . . 237<br />

Giang Huong Pham, Anjana Singh, Rajani Malla,<br />

Rina Kumari, , Ram Prasad, Minu Sachdev,<br />

Karl-Heinz Rexer, Gerhard Kost, Patricia Luis,<br />

Michael Kaldorf, François Buscot, Sylvie Herrmann,<br />

Tanja Peskan, Ralf Oelmüller, Anil Kumar Saxena,<br />

Stephané Declerck, Maria Mittag, Edith Stabentheiner,<br />

Solveig Hehl and Ajit Varma<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 237<br />

2 Interaction with Microorganisms . . . . . . . . . . . . . . . 238<br />

2.1 Rhizobacteria . . . . . . . . . . . . . . . . . . . . . . . . . . 238<br />

2.2 Chlamydomonas reinhardtii . . . . . . . . . . . . . . . . . . 239<br />

2.3 Sebacina vermifera . . . . . . . . . . . . . . . . . . . . . . . 239<br />

2.4 Other Soil Fungi . . . . . . . . . . . . . . . . . . . . . . . . . 240<br />

2.5 Gaeumannomyces graminis . . . . . . . . . . . . . . . . . . 240<br />

3 Interaction with Bryophyte . . . . . . . . . . . . . . . . . . . 242<br />

4 Interaction with Higher Plants . . . . . . . . . . . . . . . . . 242<br />

4.1 Monocots . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245<br />

4.2 Legumes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245<br />

4.3 Orchids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246<br />

4.4 Medicinal Plants . . . . . . . . . . . . . . . . . . . . . . . . . 247<br />

4.5 Economically Important Plants . . . . . . . . . . . . . . . . 249<br />

4.6 Timber Plants . . . . . . . . . . . . . . . . . . . . . . . . . . 252<br />

4.7 Unexpected Interactions with Wild Type<br />

and Genetically Modified Populus Plants . . . . . . . . . . . 253<br />

4.8 Nonmycorrhizal Plants . . . . . . . . . . . . . . . . . . . . . 255


XIV<br />

4.9 Arabidopsis thaliana . . . . . . . . . . . . . . . . . . . . . . 256<br />

4.10 Root Organ Culture . . . . . . . . . . . . . . . . . . . . . . . 259<br />

5 Cell Wall Degrading Enzymes . . . . . . . . . . . . . . . . . 260<br />

6 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 263<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 264<br />

16 Cellular Basidiomycete–Fungus Interactions . . . . . . . . 267<br />

Robert Bauer and Franz Oberwinkler<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 267<br />

2 Occurrence of Mycoparasites Within the Basidiomycota . . 267<br />

3 Hosts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268<br />

4 Cellular Interactions . . . . . . . . . . . . . . . . . . . . . . 268<br />

4.1 Colacosome-Interactions . . . . . . . . . . . . . . . . . . . . 268<br />

4.2 Fusion-Interaction . . . . . . . . . . . . . . . . . . . . . . . 275<br />

5 Basidiomycetous Mycoparasitism, a Result of Convergent<br />

Evolution? . . . . . . . . . . . . . . . . . . . . . . . . . . . . 277<br />

6 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 278<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 278<br />

Section D<br />

Contents<br />

17 Fungal Endophytes . . . . . . . . . . . . . . . . . . . . . . . 281<br />

Sita R. Ghimire and Kevin D. Hyde<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 281<br />

2 Definition of a Fungal Endophyte . . . . . . . . . . . . . . . 281<br />

3 Role of Endophytes . . . . . . . . . . . . . . . . . . . . . . . 282<br />

4 Modes of Endophytic Infection and Colonization . . . . . . 283<br />

5 Isolation of Endophytes . . . . . . . . . . . . . . . . . . . . 284<br />

6 Molecular Characterization of Endophytes . . . . . . . . . . 285<br />

7 Are Endophytes Responsible for Host Exclusivity/<br />

Recurrence in Saprobic Fungi? . . . . . . . . . . . . . . . . . 286<br />

8 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 287<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 288


Contents XV<br />

18 Mycorrhizal Development and Cytoskeleton . . . . . . . . . 293<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 293<br />

2 Cytoskeletal Components . . . . . . . . . . . . . . . . . . . 293<br />

2.1 Expression of Tubulin-Encoding Genes . . . . . . . . . . . . 294<br />

2.2 Expression of Actin-Encoding Genes . . . . . . . . . . . . . 297<br />

3 Organization of Cytoskeleton in Endomycorrhiza . . . . . . 298<br />

3.1 Root Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298<br />

3.2 Fungal Hyphae . . . . . . . . . . . . . . . . . . . . . . . . . 300<br />

4 Organization of Cytoskeleton in Ectomycorrhiza . . . . . . 300<br />

4.1 Root Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300<br />

4.2 Fungal Hyphae . . . . . . . . . . . . . . . . . . . . . . . . . 304<br />

5 Regulation of Actin Cytoskeleton Organization<br />

in Fungal Hyphae and Plant Cells . . . . . . . . . . . . . . . 305<br />

6 Actin Binding-Proteins . . . . . . . . . . . . . . . . . . . . . 307<br />

7 Microtubule-Associated Proteins . . . . . . . . . . . . . . . 308<br />

7.1 Plant Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308<br />

7.2 Fungal Hyphae . . . . . . . . . . . . . . . . . . . . . . . . . 310<br />

8 Cell Cycle and Cytoskeleton in Mycorrhiza . . . . . . . . . . 313<br />

9 Cytoskeletal Research Methods . . . . . . . . . . . . . . . . 315<br />

9.1 Indirect Immunofluorescence Microscopy . . . . . . . . . . 316<br />

9.2 Microinjection Method . . . . . . . . . . . . . . . . . . . . . 317<br />

9.3 Green Fluorescence Protein Technique . . . . . . . . . . . . 317<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 318<br />

19 Functional Diversity of Arbuscular Mycorrhizal Fungi<br />

on Root Surfaces . . . . . . . . . . . . . . . . . . . . . . . . 331<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 331<br />

2 Mycorrhiza Formation and Ecological Specificity . . . . . . 332<br />

2.1 Establishment of the Symbiosis . . . . . . . . . . . . . . . . 333<br />

2.2 Spore Germination and Hyphal Growth . . . . . . . . . . . 333<br />

2.3 Role of Plant Root Exudates . . . . . . . . . . . . . . . . . . 333<br />

3 Functioning of Arbuscular Mycorrhizas<br />

in Nutrient Exchange . . . . . . . . . . . . . . . . . . . . . . 334<br />

3.1 Metabolic Activity During Mycorrhiza Formation . . . . . . 335<br />

3.2 Gene Expression During Mycorrhiza Formation . . . . . . . 336<br />

3.3 Nutrient Exchange Mechanisms in Arbuscular Mycorrhizas 336<br />

4 Functional Diversity of Arbuscular Mycorrhizal Fungi<br />

in Root and Hyphal Interactions . . . . . . . . . . . . . . . . 338


XVI<br />

Contents<br />

4.1 Diversity of Arbuscular Mycorrhizal Fungi Inside Roots . . 339<br />

4.2 Relationship Between Hyphae in the Root and in the Soil . . 340<br />

5 Role of Arbuscular Mycorrhizal Fungi Associated<br />

with Roots in Soil Aggregation . . . . . . . . . . . . . . . . . 340<br />

6 Environmental Influence on Functional Diversity<br />

of Arbuscular Mycorrhizal Fungi . . . . . . . . . . . . . . . 341<br />

7 Role of Plant Mutants in Studying the Interactions<br />

Between Arbuscular Mycorrhizal Fungi and Roots . . . . . 341<br />

8 Conclusion and Future Research Needs . . . . . . . . . . . . 343<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 343<br />

20 Mycorrhizal Fungi and Plant Growth<br />

Promoting Rhizobacteria . . . . . . . . . . . . . . . . . . . 351<br />

José-Miguel Barea, Rosario Azcón<br />

and Concepción Azcón-Aguilar<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 351<br />

2 Main Types of Rhizosphere Microorganisms . . . . . . . . . 352<br />

3 Mycorrhizal Fungi . . . . . . . . . . . . . . . . . . . . . . . . 353<br />

4 Plant Growth Promoting Rhizobacteria . . . . . . . . . . . . 354<br />

5 Reasons for Studying Arbuscular Mycorrhizal Fungi<br />

and Plant Growth Promoting Rhizobacteria Interactions<br />

and Main Scenarios . . . . . . . . . . . . . . . . . . . . . . . 356<br />

6 Effect of Plant Growth Promoting Rhizobacteria<br />

on Mycorrhiza Formation . . . . . . . . . . . . . . . . . . . 357<br />

7 Effect of Mycorrhizas on the Establishment of Plant<br />

Growth Promoting Rhizobacteria in the Rhizosphere . . . . 357<br />

8 Interactions Involved in Nutrient Cycling and Plant<br />

Growth Promotion . . . . . . . . . . . . . . . . . . . . . . . 359<br />

9 Interactions for the Biological Control of Root Pathogens . . 361<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 362<br />

21 Carbohydrates and Nitrogen: Nutrients<br />

and Signals in Ectomycorrhizas . . . . . . . . . . . . . . . . 373<br />

Uwe Nehls<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 373<br />

2 Trehalose Utilization by Ectomycorrhizal Fungi . . . . . . . 374<br />

3 Carbohydrate Uptake . . . . . . . . . . . . . . . . . . . . . . 374<br />

4 Carbohydrate Metabolism . . . . . . . . . . . . . . . . . . . 376<br />

5 Carbohydrate Storage . . . . . . . . . . . . . . . . . . . . . . 376


Contents XVII<br />

6 Carbohydrates as Signal, Regulating Fungal<br />

Gene Expression in Ectomycorrhizas . . . . . . . . . . . . . 377<br />

7 Nitrogen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380<br />

8 Utilization of Inorganic Nitrogen . . . . . . . . . . . . . . . 381<br />

9 Utilization of Organic Nitrogen . . . . . . . . . . . . . . . . 382<br />

10 Proteolytic Activities of Ectomycorrhizal Fungi . . . . . . . 383<br />

11 Uptake of Amino Acids . . . . . . . . . . . . . . . . . . . . . 383<br />

12 Regulation of Fungal Nitrogen Export in Mycorrhizas<br />

by the Nitrogen-Status of Hyphae . . . . . . . . . . . . . . . 385<br />

13 Carbohydrate and Nitrogen-Dependent Regulation<br />

of Fungal Gene Expression . . . . . . . . . . . . . . . . . . . 385<br />

14 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 385<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 386<br />

22 Nitrogen Transport and Metabolism<br />

in Mycorrhizal Fungi and Mycorrhizas . . . . . . . . . . . . 393<br />

Arnaud Javelle, Michel Chalot, Annick Brun<br />

and Bernard Botton<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 393<br />

1.1 Ecological Significance of Ectomycorrhizas . . . . . . . . . 393<br />

1.2 Nitrogen Uptake and Translocation by Ectomycorrhizas . . 394<br />

2 Nitrate and Nitrite Transport . . . . . . . . . . . . . . . . . . 395<br />

2.1 Uptake Kinetics . . . . . . . . . . . . . . . . . . . . . . . . . 395<br />

2.2 Characterization of Nitrate and Nitrite Transporters . . . . 395<br />

3 Ammonium Transport . . . . . . . . . . . . . . . . . . . . . 398<br />

3.1 Physico-Chemical Properties of Ammonium:<br />

Active Uptake Versus Diffusion . . . . . . . . . . . . . . . . 398<br />

3.2 Physiology of Ammonium Transport in Ectomycorrhizas . . 399<br />

3.3 Isolation of Ammonium Transporter Genes . . . . . . . . . 400<br />

3.4 Regulation of the Ammonium Transporters . . . . . . . . . 400<br />

3.5 Other Putative Functions of Ammonium Transporters . . . 402<br />

4 Amino Acid Transport . . . . . . . . . . . . . . . . . . . . . 403<br />

4.1 Utilization of Amino Acids by Ectomycorrhizal Partners . . 403<br />

4.2 Molecular Regulation of Amino Acid Transport . . . . . . . 404<br />

5 Reduction of Nitrate to Nitrite and Ammonium . . . . . . . 405<br />

5.1 Reduction of Nitrate to Nitrite . . . . . . . . . . . . . . . . . 405<br />

5.2 Reduction of Nitrite to Ammonium . . . . . . . . . . . . . . 406<br />

5.3 Molecular Characterization of Nitrate Reductase<br />

and Nitrite Reductase . . . . . . . . . . . . . . . . . . . . . . 406<br />

6 Assimilation of Ammonium . . . . . . . . . . . . . . . . . . 409<br />

6.1 Role and Properties of Glutamate Dehydrogenase . . . . . . 410


XVIII<br />

6.2 Role and Properties of Glutamine Synthetase . . . . . . . . 413<br />

6.3 Role and Properties of Glutamate Synthase . . . . . . . . . . 415<br />

7 Amino Acid Metabolism . . . . . . . . . . . . . . . . . . . . 417<br />

7.1 Utilization of Proteins by Ectomycorrhizal Fungi<br />

and Ectomycorrhizas . . . . . . . . . . . . . . . . . . . . . . 417<br />

7.2 Amino Acids Used as Nitrogen and Carbon Sources . . . . . 418<br />

8 Conclusion and Future Prospects . . . . . . . . . . . . . . . 419<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 421<br />

Section E<br />

Contents<br />

23 Visualisation of Rhizosphere Interactions<br />

of Pseudomonas and Bacillus Biocontrol Strains . . . . . . 431<br />

Thomas F.C. Chin-A-Woeng, Anastasia L. Lagopodi,<br />

Ine H.M. Mulders, Guido V. Bloemberg<br />

and Ben J.J. Lugtenberg<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 431<br />

2 Tomato Foot and Root Rot and the Need<br />

for Biological Control . . . . . . . . . . . . . . . . . . . . . . 431<br />

3 Selection of Antagonistic Strains . . . . . . . . . . . . . . . 432<br />

3.1 Selection of Antagonistic Pseudomonas and Bacillus sp. . . . 432<br />

3.2 In Vitro Antifungal Activity Test . . . . . . . . . . . . . . . . 434<br />

4 In Vivo Biocontrol Assays . . . . . . . . . . . . . . . . . . . . 434<br />

4.1 Fusarium oxysporum—Tomato Biocontrol Assay<br />

in a Potting Soil System . . . . . . . . . . . . . . . . . . . . . 434<br />

4.2 Gnotobiotic Fusarium oxysporum–Pythium ultimum<br />

and Rhizoctonia solani–Tomato Bioassays . . . . . . . . . . 435<br />

5 Microscope Analysis of Infection and Biocontrol . . . . . . 435<br />

5.1 Marking Fungi with Autofluorescent Proteins . . . . . . . . 437<br />

5.2 Marking Rhizosphere Bacteria with Autofluorescent<br />

Proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 438<br />

5.3 Confocal Laser Scanning Microscopy<br />

of Rhizosphere Interactions . . . . . . . . . . . . . . . . . . 442<br />

6 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 443<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 443


Contents XIX<br />

24 Microbial Community Analysis in the Rhizosphere<br />

by in Situ and ex Situ Application of Molecular Probing,<br />

Biomarker and Cultivation Techniques . . . . . . . . . . . . 449<br />

Anton Hartmann, Rüdiger Pukall,<br />

Michael Rothballer, Stephan Gantner,<br />

Sigrun Metz, Michael Schloter and Bernhard Mogge<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 449<br />

2 In Situ Studies of Microbial Communities Using<br />

Specific Fluorescence Labeling and Confocal<br />

Laser Scanning Microscopy . . . . . . . . . . . . . . . . . . 451<br />

2.1 Fluorescence in Situ Hybridization . . . . . . . . . . . . . . 451<br />

2.2 Immunofluorescence Labeling Combined with<br />

Fluorescence in Situ Hybridization . . . . . . . . . . . . . . 453<br />

2.3 Application of Fluorescence Tagging and Reporter<br />

Constructs . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456<br />

3 Ex Situ Studies of Microbial Communities<br />

After Separation of Rhizosphere Compartments . . . . . . . 457<br />

3.1 Recovery of Bacteria from Bulk Soil, Ecto- and<br />

Endorhizosphere . . . . . . . . . . . . . . . . . . . . . . . . 457<br />

3.2 Community Analysis by Cultivation and Dot Blot Studies . . 458<br />

3.3 Community Analysis by Fluorescence<br />

in Situ Hybridization on Polycarbonate Filters . . . . . . . . 460<br />

3.4 Community Analysis by (RT) PCR-Amplification<br />

of Phylogenetic Marker Genes, D/TGGE-Fingerprinting<br />

and Clone Bank Studies . . . . . . . . . . . . . . . . . . . . . 461<br />

3.5 Community Analysis by Fatty Acid Pattern<br />

and Community Level Physiological Profile Studies . . . . . 463<br />

4 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 463<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 464<br />

25 Methods for Analysing the Interactions Between Epiphyllic<br />

Microorganisms and Leaf Cuticles . . . . . . . . . . . . . . 471<br />

Daniel Knoll and Lukas Schreiber<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 471<br />

2 Physical Characterisation of Cuticle Surfaces<br />

by Contact Angle Measurements . . . . . . . . . . . . . . . . 471<br />

3 Chemical Characterisation of Cuticle Surfaces . . . . . . . . 473<br />

4 A New in Vitro System for the Study<br />

of Interactions Between Microbes and Cuticles . . . . . . . 475


XX<br />

Contents<br />

4.1 Isolated Cuticles as Model Surfaces for Phyllosphere Studies 475<br />

4.2 Enzymatic Isolation of Plant Cuticles . . . . . . . . . . . . . 476<br />

4.3 The Experimental Set-Up of the System . . . . . . . . . . . . 476<br />

4.4 Inoculation of Cuticular Membranes<br />

with Epiphytic Microorganisms . . . . . . . . . . . . . . . . 477<br />

4.5 Measurement of Changes in Cuticular Transport Properties 479<br />

4.6 Measuring Penetration of Microorganisms<br />

Through Cuticular Membranes . . . . . . . . . . . . . . . . 481<br />

4.7 Determination of the Viable Cell Number<br />

on the Cuticle Surface . . . . . . . . . . . . . . . . . . . . . . 483<br />

4.8 Microscopic Visualisation of Microorganisms on the Cuticle 483<br />

5 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 486<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 486<br />

26 Quantifying the Impact of ACC Deaminase-Containing<br />

Bacteria on Plants . . . . . . . . . . . . . . . . . . . . . . . . 489<br />

Donna M. Penrose and Bernard R. Glick<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 489<br />

2 Selection of Bacterial Strains that Contain ACC Deaminase . 489<br />

3 Culture Conditions for the Induction<br />

of Bacterial ACC Deaminase Activity . . . . . . . . . . . . . 491<br />

4 Gnotobiotic Root Elongation Assay . . . . . . . . . . . . . . 492<br />

5 Measurement of ACC Deaminase Activity . . . . . . . . . . 493<br />

5.1 Assay of ACC Deaminase Activity in Bacterial Extracts . . . 494<br />

6 Measurement of ACC in Plant Roots, Seed Tissues<br />

and Seed Exudates . . . . . . . . . . . . . . . . . . . . . . . 495<br />

6.1 Collection of Canola Seed Tissue and Exudate<br />

During Germination . . . . . . . . . . . . . . . . . . . . . . 495<br />

6.2 Preparation of Plant Extracts . . . . . . . . . . . . . . . . . 496<br />

6.3 Protein Concentration Assay . . . . . . . . . . . . . . . . . . 497<br />

6.4 Measurement of ACC by HPLC . . . . . . . . . . . . . . . . . 498<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 501


Contents XXI<br />

27 Applications of Quantitative Microscopy in Studies<br />

of Plant Surface Microbiology . . . . . . . . . . . . . . . . . 503<br />

Frank B. Dazzo<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 503<br />

2 Quantitation of Symbiotic Interactions Between<br />

Rhizobium and Legumes by Visual Counting Techniques . . 504<br />

2.1 The Modified Fåhraeus Slide Culture Technique<br />

for Studies of the Root—Nodule Symbiosis . . . . . . . . . . 504<br />

2.2 Attachment of Rhizobia to Legume Root Hairs . . . . . . . . 506<br />

2.3 Rhizobium-Induced Root Hair Deformations . . . . . . . . 508<br />

2.4 Primary Entry of Rhizobia into Legume Roots . . . . . . . . 509<br />

2.5 In Situ Molecular Interactions Between Legumes<br />

Roots and Surface-Colonizing Rhizobia . . . . . . . . . . . . 511<br />

2.6 Cross-Reactive Surface Antigens and Trifoliin A Host Lectin 511<br />

2.7 Rhizobium Acidic Heteropolysaccharides . . . . . . . . . . . 513<br />

2.8 Rhizobium Lipopolysaccharides . . . . . . . . . . . . . . . . 516<br />

2.9 Chitolipooligosaccharide Nod Factors . . . . . . . . . . . . 518<br />

2.10 Epidermal Pit Erosions . . . . . . . . . . . . . . . . . . . . . 522<br />

2.11 Elicitation of Root Hair Wall Peroxidase by Rhizobia . . . . 524<br />

2.12 In Situ Gene Expression . . . . . . . . . . . . . . . . . . . . 525<br />

3 Quantitation of Symbiotic Interactions Between<br />

Rhizobium and Legumes by Image Analysis . . . . . . . . . 526<br />

3.1 Definitive Elucidation of the Nature of Rhizobium<br />

Extracellular Microfibrils . . . . . . . . . . . . . . . . . . . . 526<br />

3.2 Rhizobial Modulation of Root Hair Cytoplasmic Streaming 527<br />

3.3 Motility of Rhizobia in the External Root Environment . . . 527<br />

3.4 Root Hair Alterations Affecting Their Dynamic<br />

Growth Extension and Primary Host Infection . . . . . . . . 528<br />

4 A Working Model for Very Early Stages of Root<br />

Hair Infection by Rhizobia . . . . . . . . . . . . . . . . . . . 529<br />

5 Improvements in Specimen Preparation and<br />

Imaging Optics for Plant Rhizoplane Microbiology . . . . . 529<br />

6 CMEIAS: A New Generation of Image Analysis<br />

Software for in Situ Studies of Microbial Ecology . . . . . . 531<br />

6.1 CMEIAS v. 1.27: Major Advancements in Bacterial<br />

Morphotype Classification . . . . . . . . . . . . . . . . . . . 531<br />

6.2 CMEIAS v. 3.0: Comprehensive Image Analysis<br />

of Microbial Communities . . . . . . . . . . . . . . . . . . . 532<br />

6.3 CMEIAS v. 3.0: Plotless and Plot-Based Spatial<br />

Distribution Analysis of Root Colonization . . . . . . . . . . 533<br />

6.4 CMEIAS v. 3.0: In Situ Analysis of Microbial<br />

Communities on Plant Phylloplanes . . . . . . . . . . . . . . 535


XXII<br />

Contents<br />

6.5 CMEIAS v. 3.0: In Situ Geostatistical Analysis<br />

of Root Colonization by Pioneer Rhizobacteria . . . . . . . 540<br />

6.6 CMEIAS v. 3.0: Quantitative Autecological Biogeography<br />

of the Rhizobium–Rice Association . . . . . . . . . . . . . . 541<br />

6.7 CMEIAS v. 3.0: Spatial Scale Analysis of in Situ<br />

Quorum Sensing by Root-Colonizing Bacteria . . . . . . . . 543<br />

7 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 544<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 544<br />

28 Analysis of Microbial Population Genetics . . . . . . . . . . 551<br />

Emanuele G. Biondi, Alessio Mengoni<br />

and Marco Bazzicalupo<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 551<br />

2 Materials for RAPD, AFLP and ITS . . . . . . . . . . . . . . 552<br />

3 RAPD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 553<br />

4 AFLP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 556<br />

5 ITS-RFLP Analysis . . . . . . . . . . . . . . . . . . . . . . . 559<br />

6 Statistical analysis . . . . . . . . . . . . . . . . . . . . . . . . 561<br />

7 Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . 563<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 564<br />

29 Functional Genomic Approaches for Studies<br />

of Mycorrhizal Symbiosis . . . . . . . . . . . . . . . . . . . 567<br />

Gopi K. Podila and Luisa Lanfranco<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 567<br />

2 Material and Methods . . . . . . . . . . . . . . . . . . . . . . 568<br />

2.1 Equipment . . . . . . . . . . . . . . . . . . . . . . . . . . . . 568<br />

2.2 Biological Material . . . . . . . . . . . . . . . . . . . . . . . 569<br />

2.3 RNA Extraction . . . . . . . . . . . . . . . . . . . . . . . . . 569<br />

3 RNA Quantification . . . . . . . . . . . . . . . . . . . . . . . 570<br />

3.1 Construction of a cDNA Library . . . . . . . . . . . . . . . . 570<br />

4 Conversion Protocol . . . . . . . . . . . . . . . . . . . . . . 577<br />

4.1 Evaluation of the Quality of the cDNA Library . . . . . . . . 577<br />

5 Troubleshooting . . . . . . . . . . . . . . . . . . . . . . . . . 578<br />

6 Sequencing Strategies . . . . . . . . . . . . . . . . . . . . . . 578<br />

6.1 Data Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . 579<br />

6.2 Sequence Homology Comparisons . . . . . . . . . . . . . . 579


Contents XXIII<br />

6.3 Examples of Expressed Sequence Tag Data Analysis . . . . . 579<br />

7 Macroarrays . . . . . . . . . . . . . . . . . . . . . . . . . . . 582<br />

7.1 PCR Amplification of cDNA Inserts . . . . . . . . . . . . . . 582<br />

7.2 Purification and Quantification of PCR Products . . . . . . 583<br />

7.3 Printing of Macroarrays . . . . . . . . . . . . . . . . . . . . 583<br />

7.4 Generation of Exponential cDNA Probes from<br />

RNA for Macroarrays and Hybridization Analysis . . . . . . 584<br />

7.5 Exponential Amplification of the sscDNAs . . . . . . . . . . 585<br />

8 Generation of Radiolabeled Probes . . . . . . . . . . . . . . 585<br />

9 Hybridization of Macroarrays to Radiolabeled Probes . . . 586<br />

10 Data Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . 586<br />

10.1 Data Analysis Autoradiography Images on X-ray Films . . . 587<br />

11 Example of Laccaria bicolor Macroarrays . . . . . . . . . . . 588<br />

12 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 590<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 591<br />

30 Axenic Culture of Symbiotic Fungus Piriformospora indica 593<br />

Giang Huong Pham, Rina Kumari, Anjana Singh,<br />

Rajani Malla, Ram Prasad, Minu Sachdev,<br />

Michael Kaldorf, Francois Buscot, Ralf Oelmuller,<br />

Rüdiger Hampp, Anil Kumar Saxena, Karl-Heinz Rexer,<br />

Gerhard Kost and Ajit Varma<br />

1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . 593<br />

2 Morphology . . . . . . . . . . . . . . . . . . . . . . . . . . . 593<br />

3 Taxonomy of the Fungus . . . . . . . . . . . . . . . . . . . . 595<br />

4 Chlamydospore Formation and Germination . . . . . . . . 597<br />

5 Cultivation . . . . . . . . . . . . . . . . . . . . . . . . . . . . 597<br />

6 Carbon and Energy Sources . . . . . . . . . . . . . . . . . . 600<br />

7 Biomass on Individual Amino Acids . . . . . . . . . . . . . . 604<br />

8 Growth on Complex Media . . . . . . . . . . . . . . . . . . . 604<br />

9 Phosphatic Nutrients . . . . . . . . . . . . . . . . . . . . . . 605<br />

10 Composition of Media . . . . . . . . . . . . . . . . . . . . . 606<br />

11 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . 612<br />

References and Selected Reading . . . . . . . . . . . . . . . . . . . . . 612<br />

Subject Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 615


Contributors<br />

Abbott, Lynette K.<br />

School of Earth and Geographical<br />

Sciences<br />

Faculty of Natural and Agricultural<br />

Sciences<br />

The University of Western Australia<br />

Crawley, WA 6009<br />

Australia<br />

(e-mail: labbott@cyllene.uwa.edu.au)<br />

Andrade, Galdino<br />

State University of Londrina, CCB<br />

Dept of Microbiology<br />

Microbial Ecology Laboratory<br />

PO Box 6001<br />

86051-990 Londrina, PR<br />

Brazil<br />

(e-mail: andradeg@uel.br)<br />

Azcón, Rosario<br />

Departamento de Microbiología del<br />

Suelo y Sistemas Simbióticos<br />

Estación Experimental del Zaidín<br />

CSIC<br />

Prof. Albareda 1<br />

18008 Granada<br />

Spain<br />

Azcón-Aguilar, Concepción<br />

Departamento de Microbiología del<br />

Suelo y Sistemas Simbióticos<br />

Estación Experimental del Zaidín<br />

CSIC<br />

Prof. Albareda 1<br />

18008 Granada<br />

Spain<br />

Bacon, Charles W.<br />

Department of Agriculture<br />

Agriculture Research Service<br />

Athens, Georgia<br />

USA<br />

Barea, José-Miguel<br />

Departamento de Microbiología del<br />

Suelo y Sistemas Simbióticos<br />

Estación Experimental del Zaidín<br />

CSIC<br />

Prof. Albareda 1<br />

18008 Granada<br />

Spain<br />

(e-mail: josemiguel.barea@eez.csic.es)<br />

Bauer, Robert<br />

Universität Tübingen<br />

Lehrstuhl Spezielle Botanik<br />

und Mykologie<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany<br />

(e mail: robert.bauer<br />

@uni-tuebingen.de)<br />

Bazzicalupo, Marco<br />

Dipartimento di Biologia Animale e<br />

Genetica ‘Leo Pardi’<br />

Via Romana 17<br />

50125 Firenze<br />

Italy<br />

(email: marcobazzi@dbag.unifi.it)


XXVI<br />

Contributors<br />

Belanger, Faith<br />

Department of Plant Biology<br />

and Pathology<br />

Cook College-Rutgers University<br />

New Brunswick, New Jersey<br />

USA<br />

Biondi, Emanuele G.<br />

Dipartimento di Biologia Animale e<br />

Genetica ‘Leo Pardi’<br />

Via Romana 17<br />

50125 Firenze<br />

Italy<br />

Bloemberg, Guido V.<br />

Leiden University<br />

Institute of Biology<br />

Wassenaarseweg 64<br />

2333 AL Leiden<br />

The Netherlands<br />

Botton, Bernard<br />

University Henri Poincaré Nancy 1<br />

Faculty of Sciences and Techniques<br />

UMR INRA-UHP no. 1136<br />

B.P. 236<br />

54506 Vandoeuvre-Les-Nancy Cedex<br />

France<br />

(e-mail: Bernard.Botton<br />

@scbiol.uhp-nancy.fr)<br />

Brun, Annick<br />

University Henri Poincaré Nancy 1<br />

Faculty of Sciences and Techniques<br />

UMR INRA-UHP no. 1136<br />

B.P. 236<br />

54506 Vandoeuvre-Les-Nancy Cedex<br />

France<br />

Buscot, François<br />

Institute of Ecology<br />

Department of Environmental Sciences<br />

University of Jena<br />

Dornburger Strasse 159<br />

07743 Jena, Germany<br />

Present address: Institute of Botany<br />

Department of Terrestrial Ecology<br />

University of Leipzig<br />

Johannisallee 21<br />

04103 Leipzig<br />

Germany<br />

Chalot, Michel<br />

University Henri Poincaré Nancy 1<br />

Faculty of Sciences and Techniques<br />

UMR INRA-UHP no. 1136<br />

B.P. 236<br />

54506 Vandoeuvre-Les-Nancy Cedex<br />

France<br />

Chin-A-Woeng, Thomas F.C.<br />

Leiden University<br />

Institute of Biology<br />

Wassenaarseweg 64<br />

2333 AL Leiden<br />

The Netherlands<br />

(email: chin@rulbim.leidenuniv.nl)<br />

Conrad, Ralf<br />

Max-Planck-Institut für Terrestrische<br />

Mikrobiologie<br />

Marburg, Germany<br />

(e-mail: conrad@staff.uni-marburg.de)<br />

Dazzo, Frank B.<br />

Center for Microbial Ecology<br />

Department of Microbiology and Molecular<br />

Genetics<br />

Michigan State University<br />

East Lansing, MI 48824,<br />

USA<br />

(e-mail: dazzo@msu.edu)<br />

Declerck, Stephané<br />

Unité de Microbiologie<br />

Mycothèque de l’Université catholique de<br />

Louvain<br />

Université catholique de Louvai<br />

3 Place Croix du Sud<br />

1348 Louvain-la-Neuve<br />

Belgium<br />

Gantner, Stephan<br />

GSF–National Research Center for Environment<br />

and Health<br />

Institute of Soil Ecology<br />

Ingolstädter Landstrasse 1<br />

85764 Neuherberg/München<br />

Germany


Ghimire, Sita R.<br />

Centre for Research in Fungal Diversity<br />

Department of Ecology and Biodiversity<br />

The University of Hong Kong<br />

Pokfulam Road, Hong Kong<br />

Hong Kong SAR<br />

PR China<br />

Glick, Bernard R.<br />

Department of Biology<br />

University of Waterloo, Waterloo<br />

Ontario, Canada N2L 3G1<br />

(e-mail: glick@sciborg.uwaterloo.ca)<br />

Hampp, Rüdiger<br />

Institute of Botany<br />

Department of Physiological Ecology of<br />

Plants<br />

University of Tübingen<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany<br />

(e-mail: ruediger.hampp<br />

@uni-tuebingen.de)<br />

Hartmann, Anton<br />

GSF–National Research Center for Environment<br />

and Health<br />

Institute of Soil Ecology<br />

Ingolstädter Landstrasse 1<br />

85764 Neuherberg/München<br />

Germany<br />

(e-mail: anton.hartmann@gsf.de)<br />

Hehl, Solveig<br />

Application Specialist<br />

Advanced Imaging Microscopy<br />

Carl Zeiss Jena GmbH<br />

Carl-Zeiss-Promenade 10<br />

07745 Jena<br />

Germany<br />

Herrmann, Sylvie<br />

Institute of Ecology<br />

Department of Environmental Sciences<br />

University of Jena<br />

Dornburger Strasse 159<br />

07743 Jena<br />

Germany<br />

Contributors XXVII<br />

Hyde, Kevin D.<br />

Centre for Research in Fungal Diversity<br />

Department of Ecology and Biodiversity<br />

The University of Hong Kong<br />

Pokfulam Road, Hong Kong<br />

Hong Kong SAR<br />

PR China<br />

(e-mail: kdhyde@hkucc.hku.hk)<br />

Javelle, Arnaud<br />

University Henri Poincaré Nancy 1<br />

Faculty of Sciences and Techniques<br />

UMR INRA-UHP no. 1136<br />

B.P. 236<br />

54506 Vandoeuvre-Les-Nancy Cedex<br />

France<br />

Kaldorf, Michael<br />

Institute of Ecology<br />

Department of Environmental Sciences<br />

University of Jena<br />

Dornburger Strasse 159<br />

07743 Jena, Germany<br />

Present address: Institute of Botany<br />

Department of Terrestrial Ecology<br />

University of Leipzig<br />

Johannisallee 21<br />

04103 Leipzig<br />

Germany<br />

(e-mail: kaldorf@rz.uni-leipzig.de)<br />

Knoll, Daniel<br />

Institut für Allgemeine Botanik<br />

Angewandte Molekularbiologie<br />

der Pflanzen<br />

Universität Hamburg<br />

Ohnhorststrasse 18<br />

22609 Hamburg<br />

Germany<br />

Kost, Gerhard<br />

FB Biologie<br />

Spezielle Botanik und Mykologie<br />

Philipps-Universität Marburg<br />

35032 Marburg<br />

Germany<br />

Kothamasi, David Manohar<br />

Department of Microbiology<br />

University of Delhi South Campus<br />

Benito Juarez Road<br />

New Delhi 110 021, India


XXVIII<br />

Contributors<br />

Kottke, Ingrid<br />

Fakultät für Biologie<br />

Botanisches Institut<br />

Spezielle Botanik<br />

Mykologie und Botanischer Garten<br />

Universität Tübingen<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany<br />

(e-mail: ingrid.kottke<br />

@uni-tuebingen.de)<br />

Krimm, Ursula<br />

Institut für Zelluläre und Molekulare<br />

Botanik (IZMB)<br />

Abteilung Ökophysiologie<br />

Universität Bonn<br />

Kirschallee 1<br />

53115 Bonn<br />

Germany<br />

Kuhad, Ramesh Chander<br />

Department of Microbiology<br />

University of Delhi South Campus<br />

Benito Juarez Road<br />

New Delhi 110 021<br />

India<br />

(e-mail: kuhad@hotmail.com)<br />

Kumari, Rina<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

Lagopodi, Anastasia L.<br />

Leiden University<br />

Institute of Biology<br />

Wassenaarseweg 64<br />

2333 AL Leiden<br />

The Netherlands<br />

Lanfranco, Luisa<br />

Dipartimento di Biologia Vegetale dell’Università<br />

Viale Mattioli 25<br />

10125 Torino<br />

Italy<br />

Lewis, Elizabeth<br />

Department of Plant Biology<br />

and Pathology<br />

Cook College-Rutgers University<br />

New Brunswick, New Jersey<br />

USA<br />

Lugtenberg, Ben J.J.<br />

Leiden University<br />

Institute of Biology<br />

Wassenaarseweg 64<br />

2333 AL Leiden<br />

The Netherlands<br />

Luis, Patricia<br />

Institute of Ecology<br />

Department of Environmental Sciences<br />

University of Jena<br />

Dornburger Strasse 159<br />

07743 Jena<br />

Germany<br />

Maier, Andreas<br />

Institute of Botany<br />

Department of Physiological<br />

Ecology of Plants<br />

University of Tübingen<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany<br />

Malla, Rajni<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

Mengoni, Alessio<br />

Dipartimento di Biologia Animale e<br />

Genetica ‘Leo Pardi’<br />

Via Romana 17<br />

50125 Firenze<br />

Italy<br />

Metz, Sigrun<br />

GSF–National Research Center for Environment<br />

and Health<br />

Institute of Soil Ecology<br />

Ingolstädter Landstrasse 1<br />

85764 Neuherberg/München<br />

Germany


Meyer, William<br />

Department of Plant Biology<br />

and Pathology<br />

Cook College-Rutgers University<br />

New Brunswick, New Jersey<br />

USA<br />

Mittag, Maria<br />

7Institute of General Botany<br />

Friedrich-Schiller-University Jena<br />

Am Planetarium 1<br />

07743 Jena<br />

Germany<br />

Mogge, Bernhard<br />

GSF–National Research Center for Environment<br />

and Health<br />

Institute of Soil Ecology<br />

Ingolstädter Landstrasse 1<br />

85764 Neuherberg/München<br />

Germany<br />

Moy, Melinda<br />

Department of Plant Biology<br />

and Pathology<br />

Cook College-Rutgers University<br />

New Brunswick, New Jersey<br />

USA<br />

Mulders, Ine H.M.<br />

Leiden University<br />

Institute of Biology<br />

Wassenaarseweg 64<br />

2333 AL Leiden<br />

The Netherlands<br />

Nehls, Uwe<br />

Physiologische Ökologie der Pflanzen<br />

Universität Tübingen<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany<br />

(e-mail: uwe.nehls@uni-tuebingen.de)<br />

Niini, Sara<br />

Department of Biosciences<br />

Plant Physiology<br />

P.O. Box 56<br />

00014 Helsinki University<br />

Finland<br />

Oberwinkler, Franz<br />

Universität Tübingen<br />

Lehrstuhl Spezielle Botanik<br />

und Mykologie<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany<br />

(e mail: franz.oberwinkler<br />

@uni-tuebingen.de)<br />

Oelmüller, Ralf<br />

Institute of General Botany<br />

Department of Environmental Sciences<br />

University of Jena<br />

Dornburger Strasse 159<br />

07743 Jena<br />

Germany<br />

Penrose, Donna M.<br />

Department of Biology<br />

University of Waterloo, Waterloo<br />

Ontario, Canada N2L 3G1<br />

Peskan, Tanja<br />

Institute of General Botany<br />

Department of Environmental Sciences<br />

University of Jena<br />

Dornburger Strasse 159<br />

07743 Jena<br />

Germany<br />

Pham, Giang Huong<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

Podila, Gopi K.<br />

Department of Biological Sciences<br />

University of Alabama<br />

Huntsville, AL-35899<br />

USA<br />

(e-mail: podilag@email.uah.edu)<br />

Prasad, Ram<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

Contributors XXIX


XXX<br />

Contributors<br />

Pukall, Rüdiger<br />

DSMZ–German Collection of Microbes<br />

and Cell Cultures GmbH<br />

Mascheroder Weg 1b<br />

38124 Braunschweig<br />

Germany<br />

Raudaskoski, Marjatta<br />

Department of Biosciences<br />

Plant Physiology<br />

P.O. Box 56<br />

00014 Helsinki University<br />

Finland<br />

(e-mail: marjatta.raudaskoski<br />

@helsinki.fi)<br />

Rexer, Karl-Heinz<br />

FB Biologie<br />

Spezielle Botanik und Mykologie<br />

Philipps-Universität Marburg<br />

35032 Marburg<br />

Germany<br />

Rothballer, Michael<br />

GSF–National Research Center for Environment<br />

and Health<br />

Institute of Soil Ecology<br />

Ingolstädter Landstrasse 1<br />

85764 Neuherberg/München<br />

Germany<br />

Sachdev, Minu<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

Saxena, Anil Kumar<br />

Division of Microbiology<br />

Indian Agricultural Research Institute<br />

New Delhi 110012<br />

India<br />

Schloter, Michael<br />

GSF–National Research Center for Environment<br />

and Health<br />

Institute of Soil Ecology<br />

Ingolstädter Landstrasse 1<br />

85764 Neuherberg/München<br />

Germany<br />

Schreiber, Lukas<br />

Institut für Zelluläre und Molekulare<br />

Botanik (IZMB)<br />

Abteilung Ökophysiologie<br />

Universität Bonn<br />

Kirschallee 1<br />

53115 Bonn<br />

Germany<br />

(e mail: lukas.schreiber@uni-bonn.de)<br />

Singh, Ajay<br />

Department of Biology<br />

University of Waterloo, Waterloo<br />

Ontario N2T 2J3<br />

Canada<br />

Singh, Anjana<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

Solaiman, M. Zakaria<br />

Soil Science and Plant Nutrition<br />

School of Earth and<br />

Geographical Sciences<br />

Faculty of Natural<br />

and Agricultural Sciences<br />

The University of Western Australia<br />

Crawley, WA 6009<br />

Australia<br />

Stabentheiner, Edith<br />

Institute for Plant Physiology<br />

Karl-Franzens University Graz<br />

University Street 51<br />

8010 Graz<br />

Austria<br />

Sullivan, Raymond<br />

Department of Plant Biology<br />

and Pathology<br />

Cook College-Rutgers University<br />

New Brunswick, New Jersey<br />

USA<br />

Tarkka, Mika<br />

Universität Tübingen<br />

Botanisches Institut<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany


Tripathi,K.K.<br />

Department of Biotechnology<br />

Ministry of Science and Technology<br />

C.G.O. Complex, Lodi Road<br />

New Delhi110 003<br />

India<br />

Varma, Ajit<br />

School of Life Sciences<br />

Jawaharlal Nehru University<br />

New Delhi 110067<br />

India<br />

(email: ajitvarma73@hotmail.com)<br />

Werner, Dietrich<br />

Fachbereich Biologie<br />

Fachgebiet Zellbiologie und<br />

Angewandte Botanik<br />

Philipps-University Marburg<br />

Germany<br />

(e-mail: werner@mailer.uni-marburg.de)<br />

Contributors XXXI<br />

White Jr., James F.<br />

Department of Plant Biology<br />

and Pathology<br />

Cook College-Rutgers University<br />

New Brunswick, New Jersey<br />

USA<br />

(e-mail: jwhite@AESOP.Rutgers.edu)<br />

Zhang, Chi<br />

Institute of Botany<br />

Department of Physiological Ecology<br />

of Plants<br />

University of Tübingen<br />

Auf der Morgenstelle 1<br />

72076 Tübingen<br />

Germany


1 The State of the Art<br />

Ajit Varma, Lynette K. Abbott, Dietrich Werner<br />

and Rüdiger Hampp<br />

As we enter the second century of research on associative and symbiotic<br />

microorganisms, it is heartening to see that attention is increasingly focused<br />

on the functions of these organisms in the natural and semi-natural systems<br />

in which it evolved. This volume, while encapsulating the spirit of the new<br />

adventure, also provides two further opportunities. It enables us to assess the<br />

strength of the platform from which we launch into this challenging area<br />

and to identify which experimental approaches might provide the most realistic<br />

evaluation of the roles played by <strong>surface</strong> microorganisms in natural<br />

communities. The long and difficult climb towards understanding the<br />

impacts of the microflora upon the species composition and dynamics,<br />

above and below ground, of <strong>plant</strong> communities is just beginning. This volume<br />

demonstrates both the strength and the weakness of the position from<br />

which we launch into the future. The strength may be that we have much<br />

precise information about microbial function under simplified conditions.<br />

The weakness, on the other hand, is that we have, as yet, little reliable information<br />

about the extent to which these functions are expressed under relevant,<br />

essentially multi-factorial circumstances of the kind that prevail in<br />

nature. The <strong>plant</strong> carries its major microbial community on its entire<br />

exposed <strong>surface</strong>s, from apical tip to root cap. These <strong>plant</strong> <strong>surface</strong>s represent<br />

an oozing, flaking layer of integument which discharges a wide range of substances<br />

that support a vast number of spatially discrete and specialized<br />

microbial communities, including parasites and symbionts, which can have<br />

a major impact on <strong>plant</strong> growth and development. In today’s scenario the<br />

<strong>plant</strong> <strong>surface</strong> is considered as a dynamic adaptable envelope, flexible in both<br />

its own right and the first barrier between the moist, concentrated, balanced<br />

<strong>plant</strong> cell and a hostile ever-changing external environment. It is well known<br />

that the microbial diversity on the <strong>plant</strong> <strong>surface</strong> and in the soil habitats is<br />

much greater compared to the insight using cultivation techniques. Manipulation<br />

of the <strong>plant</strong> <strong>surface</strong> microflora to improve its health is a desirable and<br />

much needed goal in <strong>plant</strong> <strong>microbiology</strong>. However, efforts to exploit this<br />

type of biological control have frequently been impeded because of major<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


2<br />

Ajit Varma et al.<br />

technical difficulties that must be overcome in order to fully understand the<br />

microbial ecology of this ecosystem, especially the lack of ability to extract<br />

in situ data that are both informative and quantifiable at spatial scales relevant<br />

to the ecological niches of the microorganisms involved. The entire volume<br />

is divided into five broad sections.<br />

The combining aspect of the chapters in sections A and B are microbial<br />

communities in their interactions with higher <strong>plant</strong>s. The communities are<br />

mainly dominated by a few species, however, a large number of other species<br />

may be equally important, although they are present only in the range of 1 %<br />

of the total population or less. Experimental studies concentrate, of course, on<br />

the major components of the communities. These representatives are also<br />

used for biotechnology purposes such as seed inoculation by Pseudomonas<br />

and Bacillus control strains (Chap. 2). The interactions of methanogens and<br />

methanotrophs independent of the <strong>plant</strong> photosynthesis and the <strong>plant</strong> root<br />

ecology is a major contribution to the global CH 4 cycle. These communities<br />

are especially present in anoxic sites in wetlands such as flooded rice fields.<br />

The different carbon sources affect the CH 4 to CO 2 ratio, an important aspect<br />

for the impact of different root components on the microbial communities in<br />

the rhizosphere, as described in Chapter 3. Abiotic factors also influence the<br />

colonization of Pseudomonas fluorescens on seeds and include, besides<br />

growth substrates, also temperature, soil humidity and pH (Chap. 3). The<br />

dynamics of microorganism populations in the rhizosphere is a topic where a<br />

large number of research groups worldwide are involved. This is related to the<br />

huge amount of organic carbon exudated from <strong>plant</strong> roots into the rhizosphere,<br />

in the order of 10 % or more of the total carbon assimilation by photosynthesis<br />

in higher <strong>plant</strong>s. All major nutrient cycles such as the carbon cycle,<br />

the nitrogen cycle, the sulfur cycle, the phosphorus cycle and the cycle for<br />

micronutrients are much more active in this rhizosphere soil compared to the<br />

bulk soil. The enormous diversity in this microhabitat is increased by the fact<br />

that many different <strong>plant</strong> families and species exudate different sets of components<br />

into the soil. In addition, the composition of lignins and hemicellulose<br />

in the cell walls can be quite different, leading to a different composition<br />

of the rhizosphere communities (Chap. 4). More information on the major<br />

groups of microorganisms in soils in general are covered in Chapter 5,<br />

describing especially the impact of microorganisms on <strong>plant</strong> development by<br />

mycorrhiza species, actinorhiza species, <strong>plant</strong> growth-promoting rhizobacteria<br />

(PGPR), phosphate-solubilizing microorganisms and the important group<br />

of lignocellulolytic microorganisms.<br />

Biotic signals from the microsymbionts inducing symbiosis and nodule<br />

development in legumes are even more specific in determining the interaction<br />

of the <strong>plant</strong>s with their specific associated bacteria such as Bradyrhizobium<br />

japonicum, Mesorhizobium loti, Sinorhizobium meliloti, Rhizobium<br />

tropici or Rhizobium etli. Flavonoids and nod factors (lipochitooligosaccharides)<br />

are the major components of the chemical language, in which the


1 The State of the Art 3<br />

microsymbionts and the host <strong>plant</strong>s communicate to each other. The signalling<br />

concept studied in this type of symbiosis is equally complicated as the<br />

mammalian notch homologues and the integrin-adhesion-receptor signalling<br />

in other multicellular organisms (Chap. 6). A large stimulus for ongoing<br />

and future research in the area of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> will be<br />

available from the use of already completed genome projects and on-going<br />

genome projects for prokaryotic and eukaryotic organisms. At present, about<br />

145 genome projects are finished and more than 580 projects are on-going<br />

(http://wit.integratedgenomics.com/GOLD/gold.html). A list of completed<br />

genomes present in the public data bases, available in June 2003, is presented<br />

in Table 1. It is interesting to note that <strong>plant</strong> symbiotic and parasitic bacteria<br />

such as Bradyrhizobium japonicum, Mesorhizobium loti, Sinorhizobium<br />

meliloti and Pseudomonas synringae have the largest procaryotic genomes.<br />

On the other side, there are some animal pathogenic organisms like Rickettsia<br />

Table 1. Complete genomes present in the public DataBases, June 2003<br />

(http://wit.integratedgenomics.com/GOLD/gold.html)<br />

Organism Size (kb) ORF number<br />

Archaeal<br />

Methanosarcina mazei 4.096 3,371 orfs MAP<br />

Methanobacterium thermoautotrophicum<br />

Bacterial<br />

1.751 1,918 orfs MAP<br />

Bradyrhizobium japonicum 9.105 8.317 orfs MAP<br />

Mesorhizobium loti 7.596 6.752 orfs MAP<br />

Sinorhizobium meliloti 6.690 6.205 orfs MAP<br />

Nostoc sp. PCC 7120 6.413 5.366 orfs MAP<br />

Pseudomonas synringae 6.397 5.615 orfs MAP<br />

Pseudomonas aeruginosa 6.264 5.570 orfs MAP<br />

Escherichia coli 0157:H7, Sakai 5.594 5.448 orfs MAP<br />

Xanthomonas campestris pv. Campestris 5.076 4.182 orfs MAP<br />

Agrobacterium tumefaciens 4.915 5.402 orfs MAP<br />

Bacillus subtilis 4.214 4.099 orfs MAP<br />

Escherichia coli 0157:H7, EDI.933 4.100 5.283 orfs MAP<br />

Nitrosomonas europeae 2.812 2.573 orfs MAP<br />

Borrelia burgdorferi B 31 1.230 1.256 orfs MAP<br />

Rickettsia prowazekii 1.111 834 orfs MAP<br />

Chlamydia trachomatis<br />

Eukaryal<br />

1.042 896 orfs MAP<br />

Orysa sativa L. ssp. indica 420.000 50.000 orfs<br />

Oryza sativa ssp. japonica 420.000 50.000 orfs<br />

Arabidopsis thaliana 115.428 25.498 orfs<br />

Neurospora crassa 43.000 10.082 orfs<br />

Schizosaccharomyces pombe 14.000 4.824 orfs<br />

Saccharomyces cerevisiae 12.069 6.294 orfs


4<br />

Ajit Varma et al.<br />

prowazekii with only 1.1 Mb, Chlamydia trachomatis with 1.04 Mb and Borellia<br />

burgdorferi with 1.23 Mb.<br />

Bacillus thuringiensis and Bt transgenic <strong>plant</strong>s are examples for biotechnology<br />

concentrated on a small number of well-studied soil microorganisms.<br />

The bio-insecticide protein is present only at a certain stage of sporulation in<br />

these organisms. Under natural conditions the spores have only a very limited<br />

survival time with less than 20 % present after 24 h (Chap. 7). The toxin from<br />

Bacillus thuringiensis released from transgenic <strong>plant</strong>s in the soil is much more<br />

stable with 25 % still present after 120 days. The toxin is protected from degradation<br />

by linkage and adsorption to clay minerals. Many other important signal<br />

molecules produced by <strong>plant</strong>s and microorganisms in the soil may also<br />

have very different half-life times by specific adsorption to soil minerals. The<br />

impact of increasing concentrations of these toxins in soils due to this biocontrol<br />

technique has not been sufficiently studied. Increases and decreases of<br />

specific subpopulations of soil microorganisms have been reported (Chap. 7).<br />

The other side of interactions, promotion instead of inhibition, is a topic of<br />

Chapter 8, which studies the mechanisms of <strong>plant</strong> growth-promoting rhizobacteria<br />

by phytohormones such as auxin and ethylene. An intermediate of<br />

ethylene synthesis is 1-aminocyclopropane-1-carboxylic acid (ACC). Microorganisms<br />

with an ACC deaminase gene increase stress tolerance of several<br />

<strong>plant</strong> species (Chap. 8). Compared to the rhizosphere, the communities in the<br />

phyllosphere have been studied less. The main reason is that the <strong>plant</strong> exudation<br />

from the rhizodermis is much larger than from the epidermis, due to the<br />

cuticles limiting carbon supply to the leaf <strong>surface</strong>s. In contrast to bacteria,<br />

fungi have the ability to penetrate the cuticles and get access to carbon supplies<br />

(Chap. 9). Future work may concentrate especially on conditions where<br />

oligotrophic situations persist and genotypes adapted to these conditions<br />

may be present and not been recognized so far. The presence of animals in the<br />

interface of <strong>plant</strong>s and microorganisms is another important aspect of communities,<br />

with the example of the Clavicipitaceae. It is very interesting to note<br />

that species of this family predominantly infect insects or the ancestors of<br />

grass-infecting species (Chap. 10). By sophisticated mechanisms, the fungi<br />

modify the <strong>plant</strong> tissues for nutrient acquisition. The shift from pathogenic<br />

interaction to mutualistic interaction in some species is a general aspect<br />

related to symbiosis and phytopathology. A completely new field of research<br />

has been developed, using the interaction of genetically modified <strong>plant</strong>s<br />

(GMP) with microbial communities or specific microorganisms (Chap. 11). In<br />

the list of GMP species, important crop <strong>plant</strong>s such as potatoes, maize, cotton,<br />

tobacco and alfalfa are used. The aspect of horizontal gene transfer (HGT)<br />

from GMP <strong>plant</strong>s to associated bacterial species and fungal species is a topic<br />

for several biotechnology research projects.<br />

Section C deals with interactions between <strong>plant</strong>s, fungi, and bacteria. The<br />

<strong>plant</strong> root constitutes an environment which forms the basis for multiple relationships<br />

with microorganisms. Fine roots of most <strong>plant</strong>s are associated with


1 The State of the Art 5<br />

symbiotic fungi, which facilitate uptake of nutrients and water.An example of<br />

such a symbiotic interaction (termed mycorrhiza), which occurs mainly with<br />

roots of trees in temperate and alpine regions is ectomycorrhiza. The formation<br />

of the resulting symbiotic structure is commonly associated with<br />

changes in root morphology. Properties of the root <strong>surface</strong> are obviously an<br />

important parameter which determines the establishment of the physical<br />

contact with soil fungi. Chapter 12 gives an overview about the current knowledge<br />

on this topic with regard to the interaction of soil bacteria and ectomycorrhiza-forming<br />

fungi. This includes recent data on the effects of a co-cultivation<br />

of a range of soil bacteria (Actinomycetes) with an important and widely<br />

distributed ectomycorrhiza-forming fungus, Amanita muscaria, as part of a<br />

model system. A specific topic is the interference of a bacterial strain, which<br />

highly promotes fungal growth with the protein complement of the latter.<br />

Chapter 13 deals with respective root properties such as type of root (long/<br />

short root) and <strong>surface</strong> chemistry. Here, hydrophobic cuticle layers obviously<br />

play an important role in hyphal attachment. In addition, compatible fungi<br />

are able to penetrate and digest this layer. How far this process is involved in<br />

altering the morphology of fungal hyphae when inside the root cortex (Hartig<br />

net formation) is discussed. As the data presented in this chapter originate<br />

mainly from ultrastructural investigations, possible pitfalls of such studies<br />

are also addressed.<br />

An integral part of root–fungus associations are soil bacteria. These can<br />

support the development of the root/fungus interaction by improving fungal<br />

root colonization, the availability of nutrients, or by producing exudates (e.g.,<br />

antibiotics) which can prevent attacks of pathogenic microorganisms. While<br />

ectomycorrhizas only constitute a small fraction of all root/fungus interactions<br />

known, another form of this symbiosis, namely endomycorrhiza, dominates<br />

by far, and facilitates nutrient uptake of many crop <strong>plant</strong>s. Fungi forming<br />

this type of mycorrhiza can usually not be cultured in the absence of a<br />

<strong>plant</strong> root. Chapter 14 focuses on structural studies of the interaction of these<br />

fungi with their host <strong>plant</strong>s. Electron microscopy reveals interaction-specific<br />

structures such as fungal deposits and interactive vesicles, which can be used<br />

for diagnostic purposes. Piriformospora indica is possibly an exception<br />

because this fungus can be cultivated separately and forms structures comparable<br />

to those of endomycorrhizas. Chapter 15 deals with the diverse interactions<br />

of this fungus with roots from a variety of <strong>plant</strong>s (from bryophytes to a<br />

wide range of angiosperms) and various groups of soil microorganisms,<br />

including bacteria of the rhizosphere (compare also Chap. 12) and other soil<br />

fungi such as Aspergillus or Gaeumannomyces (root pathogen).<br />

Interactions between smut fungi and their <strong>plant</strong> hosts are another topic of<br />

Section C. The term “smut fungus” characterizes fungi sharing similar organization<br />

and life strategies. As these fungi can considerably reduce crop<br />

yields, they are of economic importance. Most of them are members of the<br />

Ustilaginomycetes, which comprise a large number of species. Fungi can also


6<br />

Ajit Varma et al.<br />

parasite on other fungi. Basidiomycetes, e.g., include saprobes, mycorrhizaforming<br />

fungi, <strong>plant</strong> parasites, but also fungi which are parasites of other<br />

fungi. Hosts are both Basidiomycetes and Ascomycetes. Ultrastructural investigations<br />

of this kind of organismic interaction (Chap. 16) revealed two main<br />

types, the formation of colacosomes and the fusion between pathogen and<br />

host fungus cells. Colacosomes are unique organelles, which appear at the<br />

interface between parasite and host while fusion is based on specialized interactive<br />

cells (haustoria), which establish a direct cytoplasmic connection.<br />

Many microorganisms coexist with <strong>plant</strong>s in ways that do not lead to <strong>plant</strong><br />

disease, symbiosis, or other specific interactions. Some fungi or bacteria can<br />

be latent pathogens. Some have little or no influence on the <strong>plant</strong>, but may<br />

form toxic compounds that are damaging to grazing animals. Microorganisms<br />

that form more or less benign associations with <strong>plant</strong>s are generally<br />

termed ‘endophytes’ and are genetically diverse. A large number of fungal<br />

endophytes can be difficult to identify because they include a high proportion<br />

with sterile mycelia (Chap. 17). Overall, the roles of many of these organisms<br />

are poorly understood.<br />

The mechanisms for entry of endophytic organisms into <strong>plant</strong>s can be<br />

investigated using methodologies such as those applied to elucidate the<br />

cytoskeletal rearrangements of <strong>plant</strong> cells and fungal hyphae at the <strong>plant</strong>–<br />

microbe interface during colonization of roots by mycorrhizal fungi (Chap.<br />

18). Invading organisms have been shown to influence the expression of <strong>plant</strong><br />

genes for some filamentous structures within the cell cytoskeleton. Indirect<br />

immunofluorescence microscopy has been used to investigate the cytoskeleton<br />

of some mycorrhizal associations demonstrating the separation and<br />

invagination of the plasma membrane from the <strong>plant</strong> cell wall in response to<br />

growth of fungi inside the cell wall.<br />

Colonization of <strong>plant</strong>s by related and unrelated groups of microorganisms<br />

may occur simultaneously. For example, saprophytes, pathogens and mycorrhizal<br />

fungi may be associated with the same root systems and colonize roots<br />

to different degrees.Several species of arbuscular mycorrhizal fungi can simultaneously<br />

colonize the same sections of root,although they are generally separated<br />

in different cells or parts of the root cortex. Prior colonization by one<br />

organism can influence sequential colonization by other organisms. This<br />

occurs to varying degrees for different groups of <strong>plant</strong> endophytes, symbionts<br />

and pathogens. The relative extent to which roots become colonized by several<br />

species of arbuscular mycorrhizal fungi present in the same soil depends on<br />

the relative abundance of propagules of the fungi in the soil,the developmental<br />

stage of the hyphae associated with fungal propagules, the susceptibility of the<br />

roots to invasion and the physiological responses of the root to different<br />

species of fungi (Chap. 19). Investigations of the molecular communication<br />

between these fungi and their host <strong>plant</strong>s during root colonization and nutrient<br />

acquisition are now beginning to be understood in terms of gene expression<br />

in <strong>plant</strong>s and fungi. This provides a basis for predicting physiological


1 The State of the Art 7<br />

responses of <strong>plant</strong>s to colonization by communities of arbuscular mycorrhizal<br />

fungi comprising species with different capacities to take up phosphorus from<br />

soil, transport it along hyphae and transfer it to the <strong>plant</strong>.<br />

When microbial communities are established in association with roots,<br />

they may be affected by changes in rooting patterns and exudates (Chap. 20).<br />

Introduction of <strong>plant</strong> growth promoting rhizobacteria (PGPRs) into the<br />

soil/<strong>plant</strong>/microbial environment can influence organisms already present<br />

(e.g., pathogenic and mycorrhizal fungi) in addition to the roots themselves.<br />

Techniques for microbial community fingerprinting are being adapted for<br />

assessment of PGPRs, in addition to in situ methods such as confocal laser<br />

scanning microscopy, to understand root – microbial associations from the<br />

perspective of communities of organisms that perform different, and sometimes<br />

contrasting, functions.<br />

Nutrients introduced into the rhizosphere from <strong>plant</strong>s and decaying<br />

organic matter can influence physiological responses of microorganisms and<br />

their interactions with <strong>plant</strong>s. Gene regulation in some ectomycorrhizal fungi<br />

has been shown to be altered in nutrient-limiting environments and this<br />

could have consequences for nutrient uptake and transfer to <strong>plant</strong>s. For example,<br />

regulation of gene expression associated with some sugars has been<br />

shown to depend on the concentration of specific carbohydrates in the<br />

medium with threshold responses identified (Chap. 21). Expression of ammonium<br />

transporter genes can be stimulated for some fungi grown under nitrogen-limiting<br />

conditions and this could have important consequences for <strong>plant</strong><br />

establishment in nitrogen-limiting natural ecosystems. Different patterns of<br />

gene regulation have been identified for the ectomycorrhizal fungus Amanita<br />

muscaria in relation to carbon and nitrogen nutrition. Some genes are regulated<br />

by both nitrogen and carbon nutrition, while others by either nitrogen<br />

or carbon (Chap. 21). Recent advances in the adaptation of molecular techniques<br />

to studies of <strong>plant</strong> and fungal biochemistry have contributed to understanding<br />

nitrogen metabolism in <strong>plant</strong>s and microorganisms (Chap. 22). For<br />

some time, studies of nitrogen assimilation by ectomycorrhizal fungi have<br />

investigated nitrate and nitrite uptake kinetics, ammonium transport and<br />

amino acid transport. Techniques such as immunogold and 14 C labelling can<br />

now be combined with gene cloning to clarify physiological processes<br />

involved in nitrogen assimilation in ectomycorrhizal fungi to highlight their<br />

differences from saprophytic and pathogenic fungi.<br />

Section E deals with the sophisticated and novel techniques to formulate<br />

critical experiments and their design in order to retrieve excellent and reliable<br />

results. Background information for the selection of beneficial properties of<br />

Pseudomonas and Bacillus strains from the rhizospheric antagonistic to phytopathogenetic<br />

community requires elaboration, evaluation and bioassay<br />

(Chap. 23). After the selection of strains, these can be marked with a reporter<br />

gene and used to study cellular and molecular interactions between one or<br />

more beneficial microbes. These strains can also serve as a tool to study the


8<br />

Ajit Varma et al.<br />

interaction with soil-borne phytopathogens in the rhizosphere of their host<br />

<strong>plant</strong>s. Autofluorescent proteins can be used for the noninvasive study of rhizosphere<br />

interactions using epifluorescence and confocal laser scanning<br />

microscopy (CSLM). Autofluorescent proteins have become an outstanding<br />

and convenient tool for studying rhizosphere and other in situ environmental<br />

interactions and have allowed microbiologists to visualize the spatial distribution<br />

of various microorganisms. The advent of fluorescent proteins offers a<br />

broad range of applications to track bacteria and study gene expression in the<br />

rhizosphere. The whole procedure of isolation, screening of antifungal activity,<br />

determining disease suppression in bioassays, preparation and transformation<br />

of protoplasts, allows fast isolation of potential biocontrol strains. The<br />

gnotobiotic test system has proven to be a valuable test system to study interactions<br />

between biocontrol bacteria, phytopathogen, and host <strong>plant</strong>. Combined<br />

with the use of autofluorescent proteins, it provides us with an extraordinary<br />

opportunity to study the intricate cellular and molecular interactions<br />

that the key players use to mediate their actions in the rhizosphere. In depth<br />

characterization of bacterial communities residing in environmental habitats<br />

has been greatly stimulated by the application of molecular phylogenetic<br />

tools such as 16S ribosomal RNA-directed oligonucleotide probes derived<br />

from extensive 16S rDNA sequence analysis. These phylogenetic probes are<br />

successfully applied in diverse microbial habitats using the fluorescent in situ<br />

hybridization (FISH) technique. In addition, the application of the immunofluorescence<br />

techniques to detect specific subpopulations or enzymes and of<br />

fluorescence marker-tagged bacteria or reporter constructs enables a highly<br />

resolving population and functional analysis. Phylogenetic in situ studies of<br />

the population structure can thus be supplemented with functional or phenotypic<br />

in situ investigation approaches. Two experimental approaches to investigate<br />

root-associated bacterial communities are presented in Chapter 24. On<br />

one hand, population and functional studies can be conducted directly in the<br />

rhizoplane (in situ) by combining specific fluorescence probing with confocal<br />

laser scanning microscopy yielding detailed information about the localization<br />

and small scale distribution of bacterial cells and their activities on the<br />

root <strong>surface</strong>. On the other hand, the separated rhizosphere compartments and<br />

the bacteria extracted from these different compartments allow a variety of<br />

subsequent ex situ studies. The separation into the three compartments, bulk<br />

soil, ectorhizosphere and rhizoplane/endorhizosphere, has to be performed<br />

with great care and actually needs an optimization for each <strong>plant</strong> and soil type<br />

under study. The degree by which adhering soil particles (ectorhizosphere)<br />

are included in the rhizosphere studies considerably influences the outcome<br />

of the study, since these soil particles are carrying a microbial community<br />

resembling, to a varying extent, the soil situation as compared to the root <strong>surface</strong><br />

or rhizoplane situation. Certainly, in situ and ex situ studies (with separated<br />

rhizosphere compartments) both complement each other to give a more<br />

comprehensive picture. Although the microscopic in situ approach has the


1 The State of the Art 9<br />

great advantage of providing detailed spatial information about root <strong>surface</strong><br />

colonization, quantitative and qualitative data about the structural and functional<br />

diversity of root colonization can be obtained by a variety of complimentary<br />

ex situ approaches.<br />

The <strong>plant</strong> cuticle forms the solid <strong>surface</strong> environment for epiphyllic microorganisms.Detailed<br />

analysis of a variety of microbe – cuticle interactions combining<br />

physicochemical, ecophysiological and microbial aspects are presented<br />

in Chapter 25.Isolated cuticles are excellent model <strong>surface</strong>s to study the mechanisms<br />

of such interactions. Using the in vitro system, even minor changes in<br />

cuticular wax composition or permeability can be examined in relation to<br />

microbial growth.Working with entire leaves such changes would probably be<br />

masked by the physiological influence of the leaf.Therefore,this new approach<br />

might be very helpful to reveal possible mechanisms of interactions that occur,<br />

in reality, only in the scale of microhabitats. The impact of cuticular features<br />

will help us to understand the observed heterogeneous colonization of the leaf<br />

habitat and the formation of micro-colonies.Vice-versa the capacity of microbial<br />

cells to change cuticular properties might be of crucial importance for a<br />

successful colonization of the leaf <strong>surface</strong>s and could contribute substantially<br />

to microbial fitness of individual epiphyllic species.Changes in cuticular properties<br />

in relation to microbial growth can be assessed in vitro under controlled<br />

conditions. Pseudomonas putida GR12-2, a well-known <strong>plant</strong> growth promoting<br />

strain, contains the enzyme 1-aminocyclopropane-1-carboxylic acid<br />

(ACC) deaminase. This enzyme hydrolyses ACC, the immediate precursor of<br />

ethylene in <strong>plant</strong> tissues.Ethylene is required for seed germination and the rate<br />

of ethylene production increases during germination and seedling growth.<br />

One model has been suggested where ACC deaminase containing growth-promoting<br />

bacteria can lower ethylene levels and thus stimulate <strong>plant</strong> growth. A<br />

rapid and novel procedure for the isolation of ACC deaminase-containing bacteria<br />

has been described in Chapter 26. In order to be able to test the model, a<br />

method for measuring ACC in <strong>plant</strong> tissues is described. Since all of the available<br />

methods for ACC quantification had problems and limitations associated<br />

with their use,Waters AccQ.Tag Method,designed to measure amino acids,was<br />

successfully applied for ACC analysis. This procedure is simple and relatively<br />

sensitive.<br />

The protocol for understanding Rhizobium-legume root nodule symbiosis<br />

has been taken up by various microscopy techniques including bright-field,<br />

phase contrast, Nomarski interference contrast, polarized light, real time and<br />

time-lapse video, dark-field, conventional and laser scanning confocal epifluorescence,<br />

scanning electron, transmission electron, and field-emission scanning/transmission<br />

electron microscopies combined with visual counting<br />

techniques and manual interactive applications of image analysis. A new generation<br />

of innovative, customized image analysis software-CMEIAS (Center<br />

for Microbial Ecology Image Analysis System), designed specific digital<br />

images of microbial populations and communities and extracted all the infor-


10<br />

Ajit Varma et al.<br />

mative, quantitative data of in vitro microbial ecology from them at spatial<br />

scales relevant to the microbes themselves. New computer-assisted imaging<br />

technology has been successfully applied to the fascinating field of <strong>plant</strong> <strong>surface</strong><br />

<strong>microbiology</strong> (Chap. 27). CMEIAS software can “count what really<br />

counts” to enhance the quantitative analysis of microbial communities and<br />

populations in situ without cultivation.<br />

Knowledge of genetic diversity in the bacterial population has increased<br />

considerably over the last 15 years, due to the application of molecular techniques<br />

to microbial ecological studies. Among the molecular methods, the<br />

PCR-based techniques provide a powerful and high throughput approach for<br />

the study of genetic diversity in bacterial populations. Some of the most commons<br />

are the PCR-RFLP of specific sequences (16S rDNA, intergenic transcribed<br />

spacer, ITS), the repetitive extragenic palindromic-PCR and the BOX-<br />

PCR based on the presence of repetitive elements within the bacterial<br />

genome, the DNA amplification fingerprintings, RAPDs (random amplified<br />

polymorphic DNA, and AFLPs (amplified fragment length polymorphism).<br />

ITS, RAPD and AFLP have been shown to be particularly relevant for the<br />

study of genetic diversity within populations of bacteria belonging to the<br />

same or closely related species (Chap. 28). AFLP shows some advantages over<br />

the other methods due to high stringency PCR conditions which give reproducibility<br />

and easy application to <strong>plant</strong>, animal and bacterial genomic DNA.<br />

AFLP has a high informational content per single reaction, in fact, up to 100<br />

different bands can be displayed in a single lane and the scoring can be done<br />

with an automatic sequencer.<br />

While there is a considerable amount of knowledge based on the ecology<br />

and physiology of mycorrhizal fungi and their uses, the knowledge about cellular<br />

and molecular aspects leading to the growth and development of the<br />

mycorrhizal fungus, as well as the establishment of a functioning symbiosis is<br />

still limited. An appropriate approach to the study of these special fungi is to<br />

understand the molecular process leading to the host recognition, development<br />

and functioning of mycorrhiza through the analysis of expressed<br />

sequences.With the advent of many highly sophisticated techniques that have<br />

been successfully applied to the functional analysis of genes from many<br />

organisms, it is now possible to apply similar strategies to study the various<br />

aspects of the mycorrhizal symbiosis (Chap. 29). The protocol describes<br />

expressed sequence tags (EST) and macroarray techniques. These approaches<br />

provide efficient tools for mycorrhizal symbiosis research. They have the resolution<br />

and ability to obtain a more comprehensive view of various stages of<br />

mycorrhiza development or treatment effects due to nutritional changes or<br />

differences due to host responses. Data can be exchanged and compared<br />

between different laboratories and eventually will provide a platform to<br />

understand the key players (genes) that are markers for ectomycorrhizal and<br />

AM fungal symbiosis. A large number of media compositions are available in<br />

the literature for the cultivation of various groups of fungi, but almost no lit-


1 The State of the Art 11<br />

erature is available for axenic cultivation of symbiotic fungi. Chapter 30 deals<br />

with the possible methods and the tested media composition to cultivate Piriformospora<br />

indica. These media can be utilized to understand the morphological<br />

and functional properties, or to test possible biotechnological applications.<br />

Finally, for many groups of microorganisms, growth in axenic conditions is<br />

not yet possible. New methodologies for producing axenic cultures of the<br />

symbiotic fungus Piriformospora indica provide avenues for advancing the<br />

study of growth of other symbiotic organisms separately from their hosts.<br />

This is an important avenue of further studies, because it will allow us to<br />

understand a wider range of interactions between <strong>plant</strong>s and can more closely<br />

reflect the enormous diversity of <strong>plant</strong>/microbe associations that exist in<br />

every environment.


2 Root Colonisation Following Seed Inoculation<br />

Thomas F.C. Chin-A-Woeng and Ben J.J. Lugtenberg<br />

1 Introduction<br />

This chapter provides protocols for the use of a gnotobiotic sand system to<br />

study root colonisation after seed inoculation. The complete experimental<br />

setup for a gnotobiotic system to grow <strong>plant</strong>s for 7–14 days in the presence of<br />

inoculated bacteria or fungi is described. Subsequently, rhizosphere interactions<br />

and the in situ behaviour of inoculated organisms is visualised using<br />

autofluorescent proteins or other reporter systems. The behaviour of a good<br />

root-colonising Pseudomonas strain in this gnotobiotic system is described in<br />

terms of distribution, localisation, and root colonisation strategies as observed<br />

by microscopy.<br />

2 Bacterial Root Colonisation<br />

Microbial attachment to and proliferation on roots is generally referred to as<br />

root colonisation. Root colonisation is an important factor in <strong>plant</strong> pathogenesis<br />

of soil-borne microorganisms as well as in beneficial interactions used<br />

for microbiological control, biofertilisation, phytostimulation, and phytoremediation.<br />

Various methods for studying rhizosphere colonisation under axenic as<br />

well as under field soil conditions have been described and the experimental<br />

approaches taken often depend on the problems studied. In this chapter, we<br />

describe a method for studying bacterial colonisation of the <strong>plant</strong> root system<br />

after introduction by seed inoculation. This simple system can be extended to<br />

study the influence of individual biotic and abiotic factors such as those present<br />

in potting soil.<br />

Root colonisation is influenced by many variables. These factors can be<br />

biotic, such as genetic traits of the host <strong>plant</strong> and the colonising organism. For<br />

example, the possession of certain colonisation genes such as sss and/or<br />

colS/colR is necessary for efficient competitive root colonisation. In addition,<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


14<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

abiotic factors, such as growth substrate, soil humidity, soil and rhizosphere<br />

pH, and temperature heavily influence root colonisation. The study of the<br />

molecular mechanism of root colonisation of a host <strong>plant</strong> by one or more bacterial<br />

strains is complicated due to many biotic and abiotic field-soil variables<br />

which can be difficult to control. The use of a gnotobiotic system limits the<br />

biological variation and results in more reliable and reproducible experimental<br />

data. However, since the purpose of colonisation studies is to learn about<br />

the processes which occur under realistic conditions, we always test interesting<br />

gnotobiotic results in field or potting soil. With only one exception, the<br />

gnotobiotic results also appear to be the case in soil.<br />

Various visualisation systems, including light and electron microscopy and<br />

confocal laser scanning microscopy (CLSM) combined with reporter systems<br />

such as those using genes for autofluorescent proteins, b-glucuronidase, and<br />

b-galactosidase allow us to determine numbers of bacteria on the root and<br />

follow the fate of inoculant bacteria in the spermosphere after seed inoculation<br />

and along the root system after growth.<br />

In this chapter, we will also focus on the genetic and metabolic burdens in<br />

the rhizosphere as a consequence of genetic modification of the organisms<br />

required to enable the marking, tracking, recovery, and selection of bacteria<br />

in and from the rhizosphere. The gnotobiotic system provides a reproducible<br />

method to study root colonisation in terms of strategies and competition.<br />

Afterwards, the data should be verified under more natural conditions as<br />

emphasised before. Various growth substrates including sand, potting soil,<br />

field soil, and stonewool have been successfully used in the root colonisation<br />

system presented in this chapter. The system has been extended by introducing<br />

soil-borne pathogens, which allows the study of interactions between<br />

pathogen, microbes, and host <strong>plant</strong>s at the cellular level which may be important<br />

for applications such as biocontrol.<br />

3 Analysis of Tomato Root Tip Colonisation After Seed<br />

Inoculation Using a Gnotobiotic Assay<br />

3.1 Description of the Gnotobiotic System<br />

To assay colonisation, a gnotobiotic sand system comprised of two glass tubes<br />

is used. A silicone ring of 15 mm, cut from a silicone tube (25x35 mm, Rubber<br />

BV, Hilversum, The Netherlands), is placed around the top tube (outer diameter<br />

25 mm, inner diameter 21 mm, length 200 mm) at 5 cm from the end<br />

(Fig. 1). The same end is closed with gauze using a rubber band. This end is<br />

placed in a bottom tube (outer diameter 40 mm, inner diameter 35 mm, height<br />

95 mm) that contains 3 ml of water to prevent the tube content from desiccation.<br />

Subsequently, high quality quartz sand (quartz sand 0.1–0.3 mm;<br />

Wessem BV, Wessem, The Netherlands) is moisturised with <strong>plant</strong> nutrient


Fig. 1. Colonisation tube system (for explanation,<br />

see text)<br />

solution (PNS: 1.25 mM Ca(NO 3) 2, 1.25 mM KNO 3, 0.50 mM MgSO 4, 0.25 mM<br />

KH 2 PO 4 and trace elements (0.75 mg/l KI, 3.00 mg/l H 3 BO 3 , 10.0 mg/l<br />

MnSO 4◊H 2O, 2.0 mg/l ZnSO 4◊5H 2O, 0.25 mg/l Na 2MoO 4◊2H 2O, 0.025 mg/l<br />

CuSO 4 ◊5H 2 O, 0.025 mg/l CoCl 2 ◊6H 2 O, pH adjusted to 5.8; 10 % v/w).After thorough<br />

mixing, the top tubes are loosely filled with about 60 g of moisturised<br />

sand and closed with a cotton plug. The entire system is sterilised at 120 °C for<br />

20 min.<br />

3.2 Seed Disinfection<br />

2 Root Colonisation Following Seed Inoculation 15<br />

Many ways have been described to disinfect the <strong>surface</strong> of seeds of various<br />

crop <strong>plant</strong>s without causing notable decreased seed germination efficiency.<br />

Common household bleach (sodium hypochlorite) or ethanol is often used<br />

for seed <strong>surface</strong> treatments. Most bacteria and fungi on the seed coat are<br />

killed after treatment with these disinfectants. Higher concentrations of up to


16<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

50 % (v/v) sodium hypochlorite can be prepared from commercial stocks. The<br />

effectiveness of a certain procedure is dependent upon the species and source<br />

of the seeds. To ensure sterility, checks should be performed by placing the<br />

disinfected seeds on rich agar medium. Care should be taken to remove traces<br />

of the disinfectant since this may influence germination efficiency as well as<br />

the survival of the bacteria after coating or inoculation of the seed. Sterilised<br />

tomato (Lycopersicon esculentum) seeds are obtained by rinsing tomato seeds<br />

with household bleach (adjusted to approximately 5 % sodium hypochlorite)<br />

and stirring in a sterile flask for 3 min. Not all seeds sink to the bottom of the<br />

flask despite stirring. After 3 min, sterilised demineralised water is added and<br />

most, if not all, seeds will then sink to the bottom of the flask. Seeds that<br />

remain floating are discarded. The hypochlorite is removed by washing the<br />

seeds five times extensively with 20 ml sterile water, followed by 2-h washing<br />

in sterile water during which the water is replaced at least three times. Contamination<br />

checks, carried out by placing the disinfected seeds on King’s<br />

medium B agar (KB), show whether the seeds are free of contaminating<br />

microorganisms. For colonisation assays, this method is a reliable disinfection<br />

method. For disinfection of grass and wheat seeds, NaOCl/0.1 % SDS<br />

solutions can be used.<br />

If seedlings are used instead of seeds, the <strong>surface</strong> disinfected seeds are<br />

placed on PNS solidified with 1.8 % Bacto Agar and placed in the dark to allow<br />

germination. Prior to transfer to a suitable temperature for germination (e.g.<br />

28 °C for tomato), the seeds are incubated overnight at 4 °C, which often<br />

improves the germination efficiency and enhances synchronous germination<br />

of the seeds. For seeds such as tomato, wheat, or radish, it subsequently takes<br />

1–2 days before 3–5-mm root tips appear. Seeds are inspected for proper germination<br />

and seedlings with the same length of root tips are selected.<br />

3.3 Growth and Preparation of Bacteria<br />

Liquid cultures of bacterial strains are grown overnight on a rotary shaker.<br />

For colonisation experiments with a mixture of strains (e.g. wild type versus<br />

mutant) a suspension of washed bacteria is prepared in a 1:1 ratio. A volume<br />

of 1.0 ml of an overnight culture is sedimented by centrifugation and the<br />

supernatant is discarded. The cells are washed with 1 ml phosphate buffered<br />

saline (PBS: 20 mM sodium phosphate, 150 mM NaCl, pH 7.4) and resuspended<br />

in PBS. The concentration of bacteria in this suspension is determined<br />

by measuring the optical density (OD 600 nm ). The strains are diluted to<br />

a concentration of 1◊10 8 CFU/ml. If a mixture of strains is to be used for inoculation,<br />

the cells are mixed prior to inoculation of the seeds or seedlings, e.g.<br />

in a 1:1 ratio. The suspension is vortexed vigorously to yield a homogenous<br />

suspension of two strains.


3.4 Seed Inoculation<br />

Seeds are placed in the bacterial suspension with sterile forceps and shaken<br />

gently for a few seconds. After approximately 10 min, the inoculated seeds are<br />

aseptically <strong>plant</strong>ed in the sand column of the gnotobiotic system, 5 mm below<br />

the sand <strong>surface</strong>.At a concentration of the inoculation mixture of 10 8 CFU/ml,<br />

the number of bacteria attaching to tomato seeds or seedlings is close to saturation<br />

(approximately 10 4 CFU/seed) and lowering the inoculation concentration<br />

to 10 4 CFU/ml does not appear to have an effect on the numbers and distribution<br />

of bacteria on the root system after 7 days of growth. Care should be<br />

taken not to damage the roots of the seedling since this will induce formation<br />

of lateral roots. The seedlings are grown in a climate-controlled chamber<br />

(19 °C, 16/8 h day/night cycles, 70 % relative humidity) for 7 days, or until the<br />

root tips penetrate the gauze.<br />

The gnotobiotic system can be used to study the root colonisation behaviour<br />

of bacteria or be used to test strains for their competitive colonisation<br />

abilities. To screen for mutants that are impaired in competitive root colonisation,<br />

two mutants can be employed. Depending on the selectable properties<br />

of the strains (one strain must be marked with an antibiotic resistance or a<br />

reporter) the suspension can be plated on an appropriate selective medium to<br />

check the ratio of the strains. The use of Tn5lacZ marked strains allows the<br />

discrimination between wild type and mutant on 5-bromo-4-chloro-3indolyl-b-galactopyranoside<br />

(X-gal) plates after reisolation of the bacteria<br />

from part of the root system. Since chances are small that two randomly<br />

picked mutants are both colonisation mutants, one Tn5 (white) mutant can be<br />

tested against a Tn5lacZ mutant (blue), which allows faster screening for<br />

colonisation mutants, after which each mutant has to be tested against the<br />

wild-type strains.<br />

3.5 Analysis of the Tomato Root Tip<br />

2 Root Colonisation Following Seed Inoculation 17<br />

To reisolate bacteria from the rhizosphere, the complete sand column is carefully<br />

removed from the tube. Most of the still adhering rhizosphere sand is<br />

removed and a length of 1–2 cm root tip is cut off with caution to prevent<br />

cross-contamination from upper root parts. If the complete root system is to<br />

be analysed, the root can be divided into segments. The root segments are<br />

shaken in 1 ml sterile PBS in the presence of the adhering rhizosphere sand or<br />

sterile glass beads to release tightly associated bacteria from the root <strong>surface</strong><br />

on an Eppendorf shaker for 20 min. The bacterial suspension thus obtained is<br />

diluted with PBS and plated using a spiral plater on solid medium supplemented<br />

with X-gal when lacZ is used as a marker. The use of an automatic<br />

plating system and counter usually allows fast and accurate bacterial counts<br />

covering five orders of magnitude using a single dilution step.


18<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

With P. fluorescens strains WCS365 and WCS365::Tn5lacZ,a 10 4 dilution of<br />

the resuspended bacteria is plated with a spiral plater on KB medium containing<br />

X-gal (40 mg/ml). After growth, the numbers of white and blue<br />

colonies are determined. Since the bacteria are lognormally distributed in the<br />

rhizosphere, the data are log 10(CFU+1)/cm transformed prior to statistical<br />

analysis with ANOVA followed by the non-parametric Wilcoxon-Mann-Whitney<br />

U-test to test significance between sample data. Details of the statistical<br />

approaches when handling these experimental data have been reviewed.<br />

Alternatively, root sections can be prepared for visualisation by light, electron,<br />

or confocal laser scanning microscopy to obtain details of the distribution<br />

pattern of the bacteria on the root <strong>surface</strong>.<br />

3.6 Confocal Laser Scanning Microscopy<br />

Autofluorescent proteins have been successfully expressed in bacterial cells<br />

and are widely used to monitor the localisation of bacterial cells or gene<br />

expression in cells. Autofluorescent proteins can be detected in living cells<br />

without staining or invasive detection methods and require no cofactors. Furthermore,<br />

the generation and discovery of various forms of autofluorescent<br />

proteins, such as BFP, CFP, YFP, DsRed, with differing luminescent and spectral<br />

properties have spurred additional interest in the use of these proteins as<br />

reporters. Autofluorescent protein-labelled strains have been used to study<br />

microbial communities in various environmental applications such as the<br />

study of dynamics and distribution of bacteria in soil, water systems, rhizospheres,<br />

activated sludges, biodegradation/bioremediation, biofilms, and root<br />

nodulation. The protein can also be used to study gene expression and gene<br />

transfer in bacterial populations.<br />

The analysis of autofluorescent proteins using CLSM is a very powerful<br />

technique to visualise microorganisms in complex environments such as in<br />

biofilms and the rhizosphere.<br />

Computer-assisted CLSM provides high resolution imaging under noninvasive<br />

conditions. With software for three-dimensional image analysis, a<br />

spatial arrangement of the distribution of labelled bacteria can be determined.<br />

4 Genetic Tools for Studying Root Colonisation<br />

4.1 Marking and Selecting Bacteria<br />

While antibiotic resistance can be very well applied as a marker to select bacteria<br />

in vitro, field conditions often require other or additional selection<br />

methods. There are numerous ways to track bacteria in the rhizosphere, asso-


2 Root Colonisation Following Seed Inoculation 19<br />

ciated habitats, and phyllosphere. Commonly used marker genes include the<br />

gusA, lacZ, phoA, xylE, luxAB, luc, and celB genes (Table 1).<br />

The use of reporter genes such as b-galactosidase or b-glucuronidase as<br />

reporter genes has greatly facilitated the localisation of bacteria on the root<br />

<strong>surface</strong>. For b-galactosidase staining, roots or root sections can be directly<br />

fixed in 1.25 % (v/v) glutaraldehyde in Z buffer (10 mM KCl, 1 mM MgSO 4,<br />

50 mM KH 2PO 4, 50 mM K 2HPO 4, pH 7.0) for 30 min. Subsequently, the roots<br />

are washed twice in Z buffer for 30 min and stained overnight at 28 °C in a<br />

solution of X-Gal (0.8 mg/ml). The roots can be mounted for light microscopic<br />

analysis after thorough rinsing in Z buffer. The use of cross-linking fixation<br />

immobilises the bacteria on the root <strong>surface</strong> and the enzyme in the tissue.<br />

Although <strong>plant</strong>s are known to possess endogenous b-galactosidase<br />

activity, this method gives no background of b-galactosidase activity from a<br />

number of <strong>plant</strong> root systems including tomato and Arabidopsis, since<br />

endogenous <strong>plant</strong> b-galactosidases are inactivated at high temperatures. By<br />

making cross-sections of roots after staining, the method can also be used to<br />

study bacterial-root associations in which bacteria penetrate deeper into the<br />

root tissue. In a similar way bacteria carrying a b-glucuronidase gene can be<br />

detected on the root system after staining with 5-bromo-4-chloro-3-indolylb-D-glucuronide.<br />

The major advantage of the use of b-glucuronidase is that<br />

<strong>plant</strong>s do not possess endogenous b-glucuronidase activity.<br />

The optimal reporter system should provide an easy and non-invasive way<br />

to follow the fate of individual cells in the rhizosphere. In addition, it should<br />

provide the possibility to quantify the activity of specific promoters in the rhizosphere.<br />

Many of the reporters have several drawbacks and restrictions,<br />

which limit their application. Some make use of specific substrates, have high<br />

background signals, or require sophisticated and expensive equipment for<br />

detection (Table 1). Compared to these reporters, autofluorescent proteins<br />

possess several advantages and have been shown to be good tools for the<br />

detection of cells (see Chap. 23, Visualisation of rhizosphere interactions of<br />

Pseudomonas and Bacillus biocontrol strains), and are promising tools for the<br />

measurement of gene activities in the rhizosphere. Nowadays, an argon laser<br />

(488-nm wavelength) is often used to excite red-shifted gfp-variants. An epifluorescence<br />

microscope equipped with a standard fluorescein isothiocyanate<br />

filter is effective for the detection of gfp red-shifted mutants which have excitation<br />

and emission maxima at 488 and 510 nm, respectively. A DAPI (4¢6diamidino-2-phenylindole)<br />

filter set with excitation at 330–380 nm and barrier<br />

filters at 435 nm can be used to detect wild-type Gfp. Autofluorescently<br />

labelled colonies on agar plates can be detected under a hand-held UV-lamp<br />

or a low-resolution binocular microscope equipped with a UV lamp. Other<br />

methods such as flow cytometry can be used to quantify gfp-labelled bacteria.<br />

Individual cells can be detected, quantified, and sorted with high speed and<br />

accuracy. On media without added iron, fluorescent pseudomonads tend to<br />

emit background fluorescence, which can obscure the GFP fluorescence. For


20<br />

Table 1. Reporter genes commonly used for the detection of bacteria in environmental applications<br />

Gene Gene product or function Advantages and disadvantages for use in the rhizophere References<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

gusA b-Glucuronidase No background in rhizobia and <strong>plant</strong>s. Requires substrate. Sharma and Signer (1990);<br />

Streit et al. (1992)<br />

lacZ b-Galactosidase High background in most <strong>plant</strong>s and bacteria. Drahos et al. (1986);<br />

Katupitiya et al. (1992);<br />

Krishnan and Pueppke (1992)<br />

phoA Alkaline phosphatase High background in most <strong>plant</strong>s and bacteria. Reuber et al. (1991)<br />

xylE Catechol 2,3-dioxygenase Soluble end product. Winstanley et al. (1991)<br />

luxA, luc Luciferase Amplification and or photographic exposure for detection. O’Kane et al. (1988);<br />

de Weger et al. (1991);<br />

Low resolution. Silcock et al. (1992);<br />

de Weger et al. (1997)<br />

celB b-Glucosidase Detection after denaturation of endogenous enzymes. Voorhorst et al. (1995)<br />

tfdA 2,4-Dichlorophenoxyacetate Low resolution. King et al. (1991)<br />

monooxygenase<br />

gfp, bfp, yfp, Autofluorescent protein High resolution. Real-time application. Requires oxygen Chalfie et al. (1994)<br />

cfp, rfp for proper folding.<br />

Antibiotic Antibiotic resistance Requires plate counting. Hagedorn (1994)<br />

resistance<br />

Heavy metal Heavy metal resistance Requires plate counting. de Lorenzo (1994)<br />

resistance


selection using GFP-expressing bacteria, this can be easily overcome by the<br />

addition of 0.45 mM FeSO 4 ◊H 2 O. Since most GFP gene sequences are known,<br />

gfp-tagged cells can also be detected by molecular methods such as gene<br />

probing, DNA hybridisation, or PCR.<br />

4.2 Rhizosphere-Stable Plasmids<br />

To understand the biological significance of genes and mutations, they need<br />

to be studied or expressed in the context in which they are assumed to function.<br />

Also, the complementation of rhizosphere-expressed mutations and<br />

expression of reporter genes need to be performed in situ. One consideration<br />

when studying processes in complex living systems, such as under soil or rhizosphere<br />

conditions, is that antibiotic selection often cannot be applied. In<br />

addition, bacteria in the rhizosphere are assumed to be covered by a mucigel<br />

layer or form biofilms which are known to have increased resistance to antibiotics.<br />

Therefore, field and rhizosphere studies often require the use of rhizosphere-stable<br />

plasmids, e.g. for complementation of mutations or for tracking<br />

bacteria. While naturally occurring plasmids are often stably maintained<br />

within a bacterial population in the absence of selection pressure, many<br />

cloning vectors disappear without the appropriate selection. Plasmids with<br />

genes for complementation or reporter studies should therefore be stably<br />

maintained in strains without antibiotic pressure or be integrated into the<br />

chromosome.<br />

The Pseudomonas replicon pVS1 is stably maintained in many genera<br />

including Pseudomonas, Agrobacterium, Rhizobium, Burkholderia, Aeromonas,<br />

and Comamonas. Cloning vectors harbouring a 3.8-kb region of pVS1<br />

with functions for replication (rep) and stability (sta) also appear to be stably<br />

maintained. pVS1 derivatives pVSP41, pWTT2081, pME6010, pME6030,<br />

pME6040, and derivatives have been shown to be completely stable in various<br />

rhizosphere bacteria in the rhizospheres of various crop <strong>plant</strong>s. Although the<br />

incompatibility group of pVS1 has not been determined, the replicon appears<br />

to be compatible with IncP-1, IncP-4, IncP-8, IncP-10, and IncP-11 plasmids in<br />

P. aeruginosa.<br />

4.3 Genetic and Metabolic Burdens<br />

2 Root Colonisation Following Seed Inoculation 21<br />

Another consideration when introducing foreign or additional DNA on plasmids<br />

into bacterial strains is a plasmid or metabolic burden. The presence of<br />

a plasmid may confer a metabolic burden on the cells because of the presence<br />

of additional DNA and/or the expression of the reporter gene. Although the<br />

effects are often not visible under laboratory conditions, the presence of a<br />

plasmid may very well cause a genetic or metabolic burden in the rhizosphere


22<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

and e.g. negatively affect the colonisation ability of a strain, as was shown for<br />

the presence of the rhizosphere-stable plasmid pWTT2081 in P. fluorescens<br />

WCS365 in the tomato rhizosphere. In competitive colonisation studies it is,<br />

therefore, of crucial importance to restore the balance by introducing the<br />

same empty vector in other strains when they are compared. Similarly, some<br />

biocontrol strains marked with autofluorescent proteins show decreased control<br />

of disease compared to the wild type such as in the control of seed-borne<br />

net blotch by Pseudomonas chlororaphis MA 342. E. coli cells harbouring<br />

DsRed also appear to be smaller than untransformed bacteria.<br />

5 Behaviour of Root-Colonising Pseudomonas Bacteria in a<br />

Gnotobiotic System<br />

5.1 Colonisation Strategies of Bacteria<br />

Using light, electron, or confocal laser scanning microscopy, bacteria can be<br />

directly visualised on the root <strong>surface</strong> and as such allow determination of distribution<br />

and colonisation patterns. Although light microscopy offers an easy<br />

way of visualising bacteria on the root, the resolution is often just below that<br />

necessary for detailed studies. More recently, CLSM has provided much more<br />

detailed information on the distribution and interactions in the rhizosphere.<br />

The number of bacteria present on the root system can also be simply followed<br />

by dilution plating of cell suspensions of bacteria that have been reisolated<br />

from root sections. On many <strong>plant</strong> root systems bacteria appear to be<br />

distributed lognormally rather than in a uniform way. In a typical bioassay<br />

with tomato seedlings grown for 7 days in a gnotobiotic sand system bacteria<br />

also appear to be distributed lognormally. High bacterial numbers are found<br />

at the root base (10 7 –10 8 CFU/cm) which rapidly decrease to 10 3 –10 4 CFU/cm<br />

at the root tip. Under the same growth conditions, bacterial numbers on one<br />

of the many roots of wheat are one order of magnitude higher, whereas in<br />

competition with indigenous rhizobacteria the numbers are usually one order<br />

of magnitude lower.<br />

The pattern of microbial occupation of root sites by bacteria varies considerably<br />

with <strong>plant</strong> species and conditions under which <strong>plant</strong>s are grown, but<br />

the percentage of root <strong>surface</strong> covered is usually estimated less than 10 %.<br />

Often, the distribution within a small area of the <strong>plant</strong> root <strong>surface</strong> appears to<br />

consist of heavily populated areas, whereas other parts are practically devoid<br />

of bacteria. Pseudomonas cells on the tomato root are mainly present as elongated<br />

stretches on indented areas, such as junctions between epidermal cells<br />

and the deeper parts of the root epidermis, and root hairs.<br />

Transmission (TEM) and scanning electron microscopy (SEM) of the<br />

root–soil interface can reveal more details regarding the spatial relationships<br />

of microorganisms, soil, and roots than light microscopy.After removal of the


<strong>plant</strong> roots from the sand, these can be directly fixed and prepared according<br />

to standard protocols for TEM or SEM analyses. A prominent feature<br />

observed with these techniques is the mucilage or biofilm which surrounds<br />

the root and in which microorganisms develop. This biofilm is believed to<br />

provide a contact between soil and roots for diffusion of nutrients and may<br />

give some protection from other microorganisms. Although the film is also<br />

produced by the <strong>plant</strong> under axenic conditions, it appears to be thicker in<br />

non-sterile roots, where bacterial capsular material such as exopolysaccharides<br />

(EPS) may contribute significantly to this layer. The biofilm can also be<br />

visualised using confocal laser scanning microscopy combined with fluorescently<br />

marked bacteria. The encapsulation of bacteria in a mucigel may have<br />

considerable consequences for the action of certain diffusible compounds<br />

such as autoinducer molecules involved in quorum sensing. This phenomenon<br />

also complicates proper visualisation of marked bacteria that have penetrated<br />

deeper into the <strong>surface</strong> layers of the root. CLSM usually can cope with<br />

these difficulties since the system can focus on multiple planes of the specimen.<br />

An in-depth study of the stages of root colonisation by CLSM has shown<br />

that P. fluorescens WCS365 microcolonies on the root <strong>surface</strong> are usually<br />

formed from one single cell, since mature microcolonies that have been visualised<br />

on the root <strong>surface</strong> usually consist of one type of bacterium. The lognormal<br />

distribution of bacteria on the root tip indicates that most bacteria<br />

remain close to the inoculation site after seed inoculation. It is believed that<br />

occasionally, single cells detach from older parts of the root and travel along<br />

the growing root tip to establish new colonies. In later stages, mixed microcolonies<br />

can be observed with CLSM, indicating that other bacteria can join at<br />

some stage of microcolony formation.<br />

5.2 Competitive Colonisation Studies<br />

2 Root Colonisation Following Seed Inoculation 23<br />

For a long-lasting effect, biocontrol bacteria must compete with the native<br />

microflora and establish themselves for several months at a high level in the<br />

rhizosphere. Successful colonisation of the <strong>plant</strong> root is often considered to be<br />

important for the success of various applications for beneficial purposes and<br />

for suppression of <strong>plant</strong> diseases. When studying colonisation traits in our<br />

laboratory, we therefore determine competitive root colonisation of two or<br />

more strains on the root.<br />

It was assumed that various bacterial traits contribute to the ability of a<br />

bacterial strain to colonise the rhizosphere and that loss of such a trait<br />

reduces the ability to establish itself effectively in the rhizosphere and, hence,<br />

also reduces its beneficial effects. Using initially competitive root tip colonisation<br />

in the gnotobiotic system as the assay, various competitive colonisation<br />

genes and traits were identified. One of the identified traits involved in coloni-


24<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

sation is chemotaxis towards root exudate. cheA – chemotaxis mutants of various<br />

P. fluorescens strains appear to be strongly reduced in competitive root<br />

colonisation (de Weert et al. 2002). Chemotaxis was also suggested to be the<br />

first step in establishment of bacterial seed and root colonisation.<br />

Flagella-less Pseudomonas strains, when tested in competition with the<br />

wild type after application on seeds, are severely impaired in colonisation of<br />

the root tip of potato and tomato. A non-motile mutant of the Fusarium oxysporum<br />

f. sp. radicis-lycopersici (F.o.r.l.) antagonist P. chlororaphis PCL1391,<br />

was 1000-fold impaired in competitive tomato root tip colonisation.<br />

Agglutination and attachment of Pseudomonas cells to <strong>plant</strong> roots are likely<br />

to play a role in colonisation. Compounds that can mediate attachment or<br />

agglutination are adhesins, fimbriae, pili, cell <strong>surface</strong> proteins, and polysaccharides.<br />

The degree of attachment to tomato roots is correlated with the<br />

number of type 4 fimbriae on bacterial cells of P. fluorescens WCS365. The<br />

outer membrane protein OprF of P. fluorescens OE28.3 is involved in attachment<br />

to <strong>plant</strong> roots. A root-<strong>surface</strong> glycoprotein agglutinin was shown to<br />

mediate agglutination of P. putida isolate Corvallis, but had no effect on<br />

colonisation.<br />

Various Pseudomonas mutant derivatives lacking the O-antigen side chain<br />

of lipopolysaccharide (LPS) are impaired in colonisation. The colonisation<br />

defect in strains with defective LPS can be explained by assuming that for the<br />

optimal functioning of nutrient uptake systems, an intact outer membrane is<br />

required.<br />

Genes for the biosynthesis of amino acids and vitamin B1 and for utilisation<br />

of root exudate components such as organic acids are also important for<br />

colonisation of P. fluorescens WCS365 on tomato roots (Simons et al. 1997;<br />

Wijfjes et al. in preparation) and P. chlororaphis PCL1391. Putrescine is an<br />

important root exudate component of which the uptake level must be carefully<br />

regulated. P. fluorescens mutants with an increased putrescine level have<br />

a decreased growth rate resulting in a colonisation defect.<br />

Other traits that are likely to influence colonisation include generation<br />

time, osmotolerance, resistance to predators, host <strong>plant</strong> cultivar, and soil type.<br />

Genes of which the role in colonisation were not obvious were identified<br />

after screening of a random Tn5 mutant library of P. fluorescens WCS365 in<br />

competition with the parental strain. They include the nuoD gene which is<br />

part of a 14-gene operon encoding NADH dehydrogenase NDH-1 (Camacho<br />

et al. 2002). The biocontrol strain P. fluorescens WCS365 possesses two NADH<br />

dehydrogenases, and apparently, the absence of NDH-1 cannot be adequately<br />

compensated for by the other NADH dehydrogenase under rhizosphere conditions,<br />

resulting in lower fitness on the root.<br />

A two-component regulatory system consisting of the colS and colR genes,<br />

which have homology to sensor kinases and response regulators, respectively,<br />

was also shown to be involved in efficient root colonisation of strain P. fluorescens<br />

WCS365. It was concluded that an environmental stimulus is impor-


tant for colonisation, but neither the nature of the stimulus, nor the target<br />

genes are known.<br />

The sss gene, encoding a protein of the lambda integrase gene family of<br />

site-specific recombinases, to which XerC and XerD also belong, is necessary<br />

for adequate root colonisation of P. fluorescens WCS365 and P. chlororaphis<br />

PCL1391. It was postulated that a certain bacterial subpopulation, which<br />

expresses an as yet unknown cell <strong>surface</strong> component regulated by a site-specific<br />

recombinase, is important for competitive colonisation of P. fluorescens<br />

WCS365.<br />

For some strains the production of secondary metabolites contributes to<br />

the ecological competence of strains as was indeed shown for the phenazineproducing<br />

strains P. fluorescens 2–79 and P. aureofaciens 30–84 using<br />

phenazine biosynthetic mutants. Phenazine-minus strains had a reduced survival<br />

and a diminished ability to compete with the resident microflora. However,<br />

production of the antifungal factor 2,4-diacetylphloroglucinol in P. fluorescens<br />

strain F113 did not influence its persistence in the soil.<br />

5.3 Monocots Versus Dicots<br />

Differences in colonisation of bacterial strains may be attributed to different<br />

root exudate compositions of the host <strong>plant</strong>. Sugars, organic acids, and amino<br />

acids are considered to be the major readily metabolisable exudate compounds.<br />

The role of root exudate composition in colonisation behaviour was<br />

studied for a number of <strong>plant</strong>s including tomato and grass. The amount of<br />

organic acid in tomato root exudate appears to be five times higher than that<br />

of exudate sugars. Using mutants of P. fluorescens WCS365, it was shown that<br />

organic acids are the nutritional basis for tomato root colonisation by this<br />

strain (Wijfjes et al. 2002, in prep.), whereas sugars appear to be less essential<br />

for colonisation. For monocots such as wheat and grass, a ten times higher<br />

number of Pseudomonas bacteria was found on the root compared to dicots<br />

such as tomato, radish, or potato. Since dicots and monocots have different<br />

organic acid and sugar compositions, increased root colonisation efficiency<br />

by certain strains might be related to a better growth on root exudates of<br />

monocots.<br />

6 Influence of Abiotic and Biotic Factors<br />

6.1 Abiotic Factors<br />

2 Root Colonisation Following Seed Inoculation 25<br />

Commercial inoculants are mostly attached to the seed or are applied in the<br />

furrow where the bacteria can reach the seedling. However, for laboratory<br />

studies, bacterisation of seedlings instead of seeds will increase reproducibil-


26<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

ity since the experiments start with a homogenous set of seedlings and this<br />

eliminates problems associated with irregular seed germination. The use of a<br />

sterile system not only ensures more reproducible bacterial numbers on the<br />

root system, but also results in higher numbers on the root due to the absence<br />

of competition by indigenous soil bacteria.<br />

Various environmental conditions influence root colonisation efficiency in<br />

the gnotobiotic sand system. The effect of a number of biotic and abiotic factors<br />

on colonisation was determined in a tomato-P. fluorescens WCS365 system.<br />

These factors include growth substrate, temperature, soil humidity, pH,<br />

and the presence of (competing) indigenous bacteria. Usually, ten times lower<br />

bacterial numbers are found on the tomato root system when experiments are<br />

performed in non-sterile potting soil instead of sterile quartz sand, which<br />

might be explained by the presence of indigenous competing organisms. The<br />

choice of material to sustain growth of seedlings is mainly determined by the<br />

system of interest. The use of chemically clean sand ensures a reliable experimental<br />

approach, but cannot be applied for studies requiring field conditions.<br />

Sand can be replaced by potting or field soil and the soil can be practically<br />

freed from indigenous organisms by gamma irradiation. Rockwool drained in<br />

<strong>plant</strong> nutrient solution also supports <strong>plant</strong> growth and bacterial colonisation.<br />

For more compact soil systems, such as clay-containing soils, the soil can be<br />

amended with sand to facilitate the recovery of roots from the system. The<br />

gnotobiotic system has been tested for tomato, radish, potato, cucumber,<br />

grass, and wheat, and may well be suitable for growth of other <strong>plant</strong> species.<br />

Although our seedlings in the gnotobiotic sand system are normally grown<br />

for 7 days, they can be grown for up to 14 days without watering.<br />

To determine the influence of a number of abiotic factors on colonisation<br />

in the gnotobiotic system, P. fluorescens WCS365 was marked with a b-glucuronidase<br />

reporter and singly inoculated on tomato seedlings. The overall<br />

bacterial distribution of the marked bacteria was determined using dilution<br />

plating and visualised using root prints (unpublished data). Increasing fluid<br />

content from 10 up to 20 % (v/w) in sand results in an overall increase of bacterial<br />

numbers on the tomato root tip. The increased colonisation may be due<br />

to increased motility or passive transport of bacteria down the root. Utilisation<br />

of 5 % (v/w) nutrient solution severely limits <strong>plant</strong> growth and consequently,<br />

bacterial numbers are lower. Temperatures at which <strong>plant</strong>s are grown<br />

need to be selected depending on the <strong>plant</strong> species. Although we grow tomato<br />

seedlings at an intermediate temperature of 19 °C, growth is significantly<br />

enhanced at higher temperatures (e.g. 28 °C). This is also reflected in the number<br />

of bacteria sustained by the <strong>plant</strong> root system, possibly due to the effect of<br />

increased root exudation on bacterial growth.


6.2 Biotic Factors<br />

2 Root Colonisation Following Seed Inoculation 27<br />

In potting soil, numbers of inoculated bacteria on the root system are usually<br />

ten-fold lower. Decreased root colonisation is not only caused by competition<br />

with soil-borne bacteria since numbers of inoculated bacteria on roots in<br />

non-sterilised and gamma-irradiated soil are comparable.<br />

Sometimes <strong>plant</strong> roots grown in potting soil are difficult to remove from<br />

the glass colonisation tube. In such cases, a mixture of potting soil/sand (1:3<br />

w/w) can be used as a compromise between the wish to use potting soil and<br />

that to experimentally study colonisation.<br />

When a fungal pathogen is included in the system, it is possible to determine<br />

biocontrol abilities of strains under controlled conditions. In our lab,<br />

bioassays with tomato and the fungal pathogens Fusarium oxysporum f. sp.<br />

radicis-lycopersici (F.o.r.l.), Rhizoctonia solani, and Pythium ultimum systems<br />

have been successfully employed to determine antifungal abilities of<br />

pseudomonads and bacilli (Lagopodi et al. unpublished data) and to perform<br />

microscopic analyses of rhizosphere interactions (see Chap. 23, Visualisation<br />

of Rhizosphere Interactions of Pseudomonas and Bacillus Biocontrol Strains).<br />

The pathogen can be introduced together with the biocontrol agent onto the<br />

seed or mixed with the sand as a spore or mycelium suspension, depending on<br />

the question under study. For the tomato-F.o.r.l. system, spores are collected<br />

from a 3-day-old culture of F.o.r.l. grown in liquid Czapek-Dox medium.<br />

Mycelium obtained from a PDA agar culture was used for inoculation of the<br />

culture. Spores are collected after passage through a miracloth filter, washed<br />

with water, and resuspended in PNS. Numbers of spores can be determined<br />

using a haemocytometer. Finally, the spores are mixed through the sand to a<br />

final concentration of 50 CFU/g sand.<br />

P. ultimum is grown for 3–4 weeks in clarified V8-medium (20 % V8 vegetable<br />

juice [Campbell Foods, Inc.], 25 mM CaCO 3,30mg/ml cholesterol).<br />

Prior to use,V8 is clarified by sedimentation at 6000 rpm for 30 min. Alternatively,<br />

the fungus can be cultured in hemp (Cannabis sp.) seed extract for<br />

1–2 weeks. Oospores that are abundantly produced during incubation are collected<br />

and freed from the mycelium. The fungal mycelium is washed three<br />

times in sterile water and blended in 0.1 M sucrose for 1–2 min. The culture is<br />

incubated for 2 h at 130 rpm at 28 °C. The suspension is sedimented by centrifugation<br />

at 4000 rpm for 10 min., resuspended in 1 M sucrose, and incubated<br />

at –20 °C for 12 h to kill the mycelium fragments. After washing with<br />

water, the suspension is layered over 1 M sucrose and centrifuged at 2351 rpm<br />

for 1 min. Consecutive washing steps remove most of the mycelium fragments.<br />

Oospores are added to the sand to a final concentration of 3–24<br />

oospores/g sand.<br />

Plants are judged according to a fixed disease index based upon disease<br />

symptoms (Table 2). The presence of the fungus on diseased <strong>plant</strong>s can be<br />

confirmed by dipping suspected diseased parts in 0.05 % household bleach


28<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

Table 2. Pythium ultimum and Fusarium oxysporum f. sp. radicis-lycopersici disease<br />

indices<br />

Disease symptoms Disease index<br />

No visible symptoms 0<br />

Small brown spots on the main root and/or the crown 1<br />

Brown spots on the central root and extensive discoloration of crown 2<br />

Damping-off or wilting 3<br />

Dead <strong>plant</strong> 4<br />

for 30 s and rinsing in sterile water to <strong>surface</strong>-disinfect the sample, followed<br />

by incubation on PDA agar medium.<br />

7 Conclusions<br />

The sand gnotobiotic system has proven to be a good tool to study rhizosphere<br />

interactions. Environmental and biotic conditions can be more carefully<br />

controlled in this system than in natural soils. Under controlled conditions,<br />

it also allows the enrichment of strains for particular traits. In our lab<br />

mutant screening has resulted in numerous mutants involved in root colonisation,<br />

which subsequently have been genetically characterised. Combined<br />

with the use of autofluorescent proteins and CLSM, the gnotobiotic system is<br />

a powerful tool to study the interactions between biocontrol bacteria, the<br />

pathogen, and the host <strong>plant</strong>. Unstable fluorescent proteins provide the tools<br />

for study of gene expression in the rhizosphere. Rhizosphere-associated phenomena<br />

such as bacterial cell-to-cell signalling events and signalling between<br />

pathogens and rhizosphere bacteria can be investigated in a clean and reproducible<br />

way.<br />

References and Selected Reading<br />

Bahme JB, Schroth MN (1987) Spatial-temporal colonization patterns of a rhizobacterium<br />

on underground organs of potato. Phytopathology 77:1093–1100<br />

Bloemberg GV, O’Toole GA, Lugtenberg BJJ, Kolter R (1997) Green fluorescent protein as<br />

a marker for Pseudomonas spp. Appl Environ Microbiol 63:4543–4551<br />

Bloemberg GV,Wijfjes AH, Lamers GE, Stuurman N, Lugtenberg BJ (2000) Simultaneous<br />

imaging of Pseudomonas fluorescens WCS365 populations expressing three different<br />

autofluorescent proteins in the rhizosphere: new perspectives for studying microbial<br />

communities. Mol Plant-Microbe Interact 13:1170–1176<br />

Bowen GD, Rovira AD (1976) Microbial colonization of <strong>plant</strong> roots. Annu Rev Phytopathol<br />

14:121–144


2 Root Colonisation Following Seed Inoculation 29<br />

Buell CR, Anderson AJ (1993) Expression of the aggA locus of Pseudomonas putida in<br />

vitro and in <strong>plant</strong>a as detected by the reporter gene, xylE. Mol Plant-Microbe Interact<br />

6:331–340<br />

Bull CT, Weller DM, Thomashow LS (1991) Relationship between root colonization and<br />

suppression of Gaeumannomyces graminis var. tritici by Pseudomonas fluorescens<br />

strain 2–79. Phytopathology 81:954–959<br />

Camacho MM (2001) Molecular characterization of type 4 pili, NDHI and PyrR in rhizosphere<br />

colonization of Pseudomonas fluorescens WCS365. PhD Thesis, Universiteit<br />

Leiden, Leiden<br />

Camacho Carvajal MM, Wijfjes AHM, Mulders IHM, Lugtenberg BJJ, Bloemberg GV<br />

(2002) Characterization of NADH dehydrogenases of Pseudomonas fluorescens<br />

WCS365 and their role in competitive root colonisation. Mol Plant-Microbe Interact<br />

15:662–671<br />

Campbell R, Rovira AD (1973) The study of the rhizosphere by scanning electron<br />

microscopy. Soil Biol Biochem 5:747–752<br />

Caroll H, Moënne-Loccoz Y, Dowling D, O’Gara F (1995) Mutational disruption of the<br />

biosynthesis genes coding for the antifungal metabolite 2,4-diacetylphloroglucinol<br />

does not influence the ecological fitness of Pseudomonas fluorescens F113 in the rhizosphere<br />

of sugar beets. Appl Environ Microbiol 61:3002–3007<br />

Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC (1994) Green fluorescent protein<br />

as a marker for gene expression. Science 263:802–805<br />

Chin-A-Woeng TFC, de Priester W, van der Bij AJ, Lugtenberg BJJ (1997) Description of<br />

the colonization of a gnotobiotic tomato rhizosphere by Pseudomonas fluorescens<br />

biocontrol strain WCS365, using scanning electron microscopy. Mol Plant-Microbe<br />

Interact 10:79–86<br />

Chin-A-Woeng TFC, Bloemberg GV, van der Bij AJ, van der Drift KMGM, Schripsema J,<br />

Kroon B, Scheffer RJ, Keel C, Bakker PAHM, Tichy HV, de Bruijn FJ, Thomas-Oates JE,<br />

Lugtenberg BJJ (1998) Biocontrol by phenazine-1-carboxamide-producing Pseudomonas<br />

chlororaphis PCL1391 of tomato root rot caused by Fusarium oxysporum f. sp.<br />

radicis-lycopersici. Mol Plant-Microbe Interact 11:1069–1077<br />

Chin-A-Woeng TFC, Bloemberg GV, Mulders IHM, Dekkers LC, Lugtenberg BJJ (2000)<br />

Root colonization by phenazine-1-carboxamide-producing bacterium Pseudomonas<br />

chlororaphis PCL1391 is essential for biocontrol of tomato foot and root rot. Mol<br />

Plant-Microbe Interact 13:1340–1345<br />

Christensen BB, Sternberg C, Molin S (1996) Bacterial plasmid conjugation on semisolid<br />

<strong>surface</strong>s monitored with the green fluorescent protein (GFP) from Aequorea victoria<br />

as a marker. Gene 173:59–65<br />

Clarholm M (1984) Heterothrophic, free-living protozoa: neglected microorganisms<br />

with an important task in regulating bacterial populations. In: Klug MJ, Reddy CA<br />

(eds) Current perspectives in microbial ecology. American Society of Microbiology,<br />

Washington, DC, pp 321–326<br />

Cormack BP, Valdivia RH, Falkow S (1996) FACS-optimized mutants of the green fluorescent<br />

protein (GFP). Gene 173:33–38<br />

Davies KG, Whitbread R (1989) A comparison of methods for measuring the colonisation<br />

of a root system by fluorescent pseudomonads. Plant Soil 116:239–246<br />

de Lorenzo V (1994) Designing microbial systems for gene expression in the field. Trends<br />

Biotechnol 12:365–371<br />

de Weert S, Vermeiren H, Mulders IHM, Kuiper I, Hendrickx N, Bloemberg GV, Vanderleyden<br />

J, DeMot R, Lugtenberg BJJ (2002) Flagella-driven chemotaxis towards exudate<br />

components is an important trait for tomato root colonization by Pseudomonas fluorescens.<br />

Mol Plant-Microbe Interact 15:1173–1180


30<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

de Weger LA, Bakker PAHM, Schippers B, van Loosdrecht MCM, Lugtenberg BJJ (1989)<br />

Pseudomonas spp. with mutational changes in the O-antigenic side chain of their<br />

lipopolysaccharide are affected in their ability to colonize potato roots. In: Lugtenberg<br />

BJJ (ed) Signal molecules in <strong>plant</strong>s and <strong>plant</strong>-microbe interactions. NATO ASI<br />

Series H, Springer, Berlin Heidelberg New York, pp 197–202<br />

Dekkers LC (1997) Isolation and characterization of novel rhizosphere colonization<br />

mutants of Pseudomonas fluorescens WCS365. PhD Thesis, Leiden University, Leiden,<br />

The Netherlands<br />

Dekkers LC, van der Bij AJ, Mulders IHM, Phoelich CC, Wentwood RAR, Glandorf DCM,<br />

Wijffelman CA, Lugtenberg BJJ (1998a) Role of the O-antigen of lipopolysaccharide,<br />

and possible roles of growth rate and of NADH:Ubiquinone oxidoreductase (nuo) in<br />

competitive tomato root-tip colonization by Pseudomonas fluorescens WCS365. Mol<br />

Plant-Microbe Interact 11:763–771<br />

Dekkers LC, Phoelich CC, van der Fits L, Lugtenberg BJJ (1998b) A site-specific recombinase<br />

is required for competitive root colonization by Pseudomonas fluorescens<br />

WCS365. Proc Natl Acad Sci USA 95:7051–7056<br />

Dekkers LC, Bloemendaal CP, de Weger LA, Wijffelman CA, Spaink HP, Lugtenberg BJJ<br />

(1998 c) A two-component system plays an important role in the root-colonizing ability<br />

of Pseudomonas fluorescens strain WCS365. Mol Plant-Microbe Interact 11:45–56<br />

Dekkers LC, Mulders IH, Phoelich CC, Chin-A-Woeng TFC, Wijfjes AH, Lugtenberg BJ<br />

(2000) The sss colonization gene of the tomato-Fusarium oxysporum f. sp. radicislycopersici<br />

biocontrol strain Pseudomonas fluorescens WCS365 can improve root colonization<br />

of other wild-type Pseudomonas spp. bacteria. Mol Plant-Microbe Interact<br />

13:1177–1183<br />

DeMot R, Veulemans B, Vanderleyden J (1991) Root-adhesive protein of Pseudomonas<br />

fluorescens OE28–3. In: Keel C, Knoller B, Défago G (eds) Plant growth-promoting<br />

rhizobacteria. Progress and prospects. International organization for biological and<br />

integrated control of noxious animals and <strong>plant</strong>s. Proceedings of the 2nd International<br />

Workshop on PGPR. WPRS Bulletin XIV/8, 308–312<br />

de Weger LA, Dunbar P, Mahafee WF, Lugtenberg BJJ, Sayler G (1991) Use of bioluminescence<br />

markers to detect Pseudomonas spp. in the rhizosphere. Appl Environ Microbiol<br />

57:3641–3644<br />

de Weger LA, Kuiper I, van der Bij AJ, Lugtenberg BJJ (1997) Use of a lux-based procedure<br />

to rapidly visualize root colonisation by Pseudomonas fluorescens in the wheat<br />

rhizisphere. Anton Leeuw Int J G 72:365–372<br />

Drahos DJ, Hemming BC, McPherson S (1986) Tracking recombinant organisms in the<br />

environment: b-galactosidase as a selectable non-antibiotic marker for fluorescent<br />

pseudomonads. Bio/Technology 4:439–444<br />

Errampalli D, Okamura H, Lee H, Trevors JT, van Elsas JD (1998) Green fluorescent protein<br />

as a marker to monitor survival of phenanthrene-mineralizing Pseudomonas sp.<br />

UG14Gr in creosote-contaminated soil. FEMS Microbiol Ecol 26:181–191<br />

Errampalli D, Leung K, Cassidy MB, Kostrzynska M, Blears M, Lee H, Trevors JT (1999)<br />

Applications of the green fluorescent protein as a molecular marker in environmental<br />

microorganisms. J Microbiol Meth 35:187–199<br />

Foster RC (1986) The ultrastructure of the rhizoplane and rhizosphere. Annu Rev Phytopathol<br />

24:211–234<br />

Glandorf DCM, Sluis I, Anderson AJ, Bakker PAHM, Schippers B (1994) Agglutination,<br />

adherence, and root colonization by fluorescent pseudomonads.Appl Environ Microbiol<br />

60:1726–1733<br />

Greaves MP, Darbyshire JF (1972) The ultrastructure of the mucilaginous layer on <strong>plant</strong><br />

roots. Soil Biol Biochem 4:443–449<br />

Habte M, Alexander M (1977) Further evidence for the regulation of bacterial populations<br />

in soil by protozoa. Arch Microbiol 113:181–183


2 Root Colonisation Following Seed Inoculation 31<br />

Hagedorn C (1994) Spontaneous and intrinsic antibiotic resistance markers. In: Weaver<br />

RS, Angle S, Bottomley P (eds) Methods of soil analysis, Part 2, Microbiological and<br />

chemical properties. Soil Science of America, Inc, Madison, WI, pp 575–591<br />

Heeb S, Itoh Y, Nishijyo T, Schnider U, Keel C, Wade J, Walsh U, O’Gara F, Haas D (2000)<br />

Small, stable shuttle vectors based on the minimal pVS1 replicon for use in gram-negative,<br />

<strong>plant</strong>-associated bacteria. Mol Plant-Microbe Interact 13:232–237<br />

Hoffland E, Findenegg GR, Nelemans JA (1989) Solubilization of rock phosphate by rape.<br />

Plant Soil 113:161–165<br />

Howie WJ, Cook RJ, Weller DM (1987) Effect of soil matric potential and cell motility on<br />

wheat root colonization by fluorescent pseudomonads suppressive to take-all. Phytopathology<br />

77:286–292<br />

Itoh Y, Haas D (1985) Cloning vectors derived from the Pseudomonas plasmid pVSP1.<br />

Gene 36:27–36<br />

Itoh Y, Watson JM, Haas D, Leisinger T (1984) Genetic and molecular characterization of<br />

the Pseudomonas plasmid pVS1. Plasmid 11:206–220<br />

Jakobs S, Subramaniam V, Schonle A, Jovin TM, Hell SW (2000) EFGP and DsRed<br />

expressing cultures of Escherichia coli imaged by confocal, two-photon and fluorescence<br />

lifetime microscopy. FEBS Lett 479:131–135<br />

Jenny H, Grossenbacher K (1963) Root-soil boundary zones as seen in the electron<br />

microscope. Soil Sci Soc Am Proc 27:273–277<br />

Katupitiya S, New PB, Elmerich C, Kennedy IR (1992) Improved N 2 -fixation in 2,4-Dtreated<br />

wheat roots associated with A. lipoferum: studies of colonisation using<br />

reporter genes. Soil Biol Biochem 27:447–452<br />

King EO, Ward MK, Raney DE (1954) Two simple media for the demonstration of<br />

pyocyanin and fluorescein. J Lab Clin Med 44:301–307<br />

King RJ, Short KA, Seidler RJ (1991) Assay for detection and enumeration of genetically<br />

engineered microorganisms which is based on the activity of a deregulated 2,3dichlorophenoxyacetate<br />

monooxygenase. Appl Environ Microbiol 57:1790–1792<br />

Kloepper JW, Beauchamp CJ (1992) A review of issues related to measuring colonization<br />

of <strong>plant</strong> roots by bacteria. Can J Microbiol 38:1219–1232<br />

Knudsen IMB, Hockenhull J, Jensen DF, Gerhardson B, Hokeberg M, Tahvonen R, Teperi<br />

E, Sundheim L, Henriksen B (1997) Selection of biological control agents for controlling<br />

soil and seed-borne diseases in the field. Eur J Plant Pathol 103:775–784<br />

Krishnan HB, Pueppke SG (1992) A nolC-lacZ gene fusion in Rhizobium fredii facilitates<br />

direct assessment of competition for nodulation of soybean. Can J Micriobiol<br />

38:515–519<br />

Kuiper I, Bloemberg GV, Lugtenberg BJ (2001a) Selection of a <strong>plant</strong>-bacterium pair as a<br />

novel tool for rhizostimulation of polycyclic aromatic hydrocarbon-degrading bacteria.<br />

Mol Plant-Microbe Interact 14:1197–1205<br />

Kuiper I, Bloemberg GV, Noreen S, Thomas-Oates JE, Lugtenberg BJJ (2001b) Increased<br />

uptake of putrescine in the rhizosphere inhibits competitive root colonization by<br />

Pseudomonas fluorescens strain WCS365. Mol Plant-Microbe Interact 14:1096–1104<br />

Lagopodi AL, Ram AFJ, Lamers GEM, Punt PJ, van den Hondel CAMJJ, Lugtenberg BJJ,<br />

Bloemberg GV (2002) Novel aspects of tomato root colonization and infection by<br />

Fusarium oxysporum f. sp. radicis-lycopersici revealed by confocal laser scanning<br />

microscopic analysis using the green fluorescent protein as a marker. Mol Plant-<br />

Microbe Interact 15:172–179<br />

Lewis K (2001) Riddle of biofilm resistance. Antimicrob Agents Chemother 45:999–1007<br />

Loper JE, Suslow TV, Schroth MN (1984) Lognormal distribution of bacterial populations<br />

in the rhizosphere. Phytopathology 74:1454–1460<br />

Loper JE, Haack C, Schroth MN (1985) Population dynamics of soil pseudomonads in<br />

rhizosphere of potato (Solanum tuberosum L.). Appl Environ Microbiol 49:416–422


32<br />

Thomas F.C. Chin-A-Woeng and Ben J. J. Lugtenberg<br />

Lugtenberg BJJ, de Weger LA, Schippers B (1994) Bacterization to protect seed and rhizosphere<br />

against disease. BCPC Monograph 57:293–302<br />

Lugtenberg BJJ, Dekkers LC, Bansraj M, Bloemberg GV, Camacho M, Chin-A-Woeng<br />

TFC, van den Hondel C, Kravchenko L, Kuiper I, Lagopodi AL, Mulders I, Phoelich C,<br />

Ram A, Tikhonovich I, Tuinman S, Wijffelman C, Wijfjes A (1999a) Pseudomonas<br />

genes and traits involved in tomato root colonization. In: de Wit PJGM, Bisseling T,<br />

Stiekema WJ (eds) 1999 IC-MPMI Congress Proceedings: Biology of <strong>plant</strong>-microbe<br />

interactions, volume 2. International Society for Molecular Plant-Microbe Interactions,<br />

St. Paul, MN, pp 324–330<br />

Lugtenberg BJJ, Kravchenko LV, Simons M (1999b) Tomato seed and root exudate sugars:<br />

composition, utilization by Pseudomonas biocontrol strains and role in rhizosphere<br />

colonization. Environ Microbiol 1:439–446<br />

Lugtenberg BJJ, Dekkers LC, Bloemberg GV (2001) Molecular determinants of rhizosphere<br />

colonization by Pseudomonas. Annu Rev Phytopathol 39:461–490<br />

Mah TF, O’Toole GA (2001) Mechanisms of biofilm resistance to antimicrobial agents.<br />

Trends Microbiol 9:34–39<br />

Mazzola M, Cook RJ, Thomashow LS, Weller DM, Pierson LS (1992) Contribution of<br />

phenazine antibiotic biosynthesis to the ecological competence of fluorescent<br />

pseudomonads in soil habitats. Appl Environ Microbiol 58:2616–2624<br />

McLoughlin AJ (1994) Plasmid stability and ecological competence in recombinant cultures.<br />

Biotechnol Adv 12:279–324<br />

Miller WG, Lindow SE (1997) An improved GFP cloning cassette designed for prokaryotic<br />

transcriptional fusions. Gene 191:149–153<br />

Möller S, Steinberg C,Andersen JB, Christensen BB, Ramos JL, Givskov M, Molin S (1998)<br />

In situ gene expression in mixed-culture biofilms evidence of metabolic interactions<br />

between community members. Appl Environ Microbiol 64:721–732<br />

Nordström K (1989) Mechanisms that contribute to the stable segregation of plasmids.<br />

Annu Rev Genet 23:37–69<br />

O’Kane DJ, Lingle WL, Wampler JE, Legocki RP, Szalay AA (1988) Visualization of bioluminescence<br />

as a marker of gene expression in rhizobium-infected soybean root nodules.<br />

Plant Mol Biol 10:387–399<br />

Okker RJ, Spaink H, Hille J, van Brussel TA, Lugtenberg B, Schilperoort RA (1984) Plantinducible<br />

virulence promoter of the Agrobacterium tumefaciens Ti plasmid. Nature<br />

312:564–566<br />

Palmer RJJ, Sternberg C (1999) Modern microscopy in biofilm research: confocal<br />

microscopy and other approaches. Curr Opin Biotechnol 10:263–268<br />

Partridge JE, Smith FD, Harpending PR, Rasmussen JL, Sanford JC (1991) Preparation of<br />

mycelium-free suspensions of oospores of Phytophtera megasperma var. sojae.Appl<br />

Environ Microbiol:480–485<br />

Prosser JI (1994) Molecular marker systems for detection of genetically engineered<br />

micro-organisms in the environment. Microbiology 140:5–17<br />

Rengel Z, Ross G, Hirsch P (1998) Plant genotype and micronutrient status influence colonization<br />

of wheat roots by soil bacteria. J Plant Nutr 21:99–113<br />

Reuber TL, Long SL, Walker GC (1991) Regulation of Rhizobium meliloti exo genes in<br />

free-living cells and in <strong>plant</strong>a examined using TnphoA fusions. J Bacteriol 173:426–<br />

434<br />

Rhodes DJ, Powell KA (1994) Biological seed treatments – the development process.<br />

BCPC Monograph 57:303–310<br />

Rovira AD, Sands DC (1974) Quantitative assessment of the rhizoplane microflora by<br />

direct microscopy. Soil Biol Biochem 6:211–216<br />

Rovira AD, Campbell R (1975) A scanning electron microscope study of interactions<br />

between micro-organisms and Gaeumannomyces graminis (syn. Ophiobolus graminis)<br />

on wheat roots. Microbial Ecol 3:177–185


2 Root Colonisation Following Seed Inoculation 33<br />

Ryder MH, Pankhurst CE, Rovira AD, Corell RL, Ophel Keller KM (1994) Detection of<br />

introduced bacteria in the rhizosphere using marker genes and DNA probes. In:<br />

O’Gara F, Dowling DN, Boesten B (eds) Molecular ecology of rhizosphere microorganisms.VCH,<br />

Weinheim, pp 29–47<br />

Scher FM, Kloepper JW, Singleton CA (1985) Chemotaxis of fluorescent Pseudomonas<br />

spp. to soybean seed exudates in vitro and in soil. Can J Microbiol 31:570–574<br />

Sharma SB, Signer ER (1990) Temporal and spatial regulation of the symbiotic genes of<br />

Rhizobium meliloti in <strong>plant</strong>a revealed by transposon Tn5-gusA. Genes Dev. 4:344–356<br />

Silcock DJ, Waterhouse RN, Glover LA, Prosser JI, Killham K (1992) Detection of a single<br />

genetically engineered modified bacterial cell in soil by using charge coupled deviceenhanced<br />

microscopy. Appl Environ Microbiol 58:2444–2448<br />

Simons M, van der Bij AJ, Brand J, de Weger LA, Wijffelman CA, Lugtenberg BJJ (1996)<br />

Gnotobiotic system for studying rhizosphere colonization by <strong>plant</strong> growth-promoting<br />

Pseudomonas bacteria. Mol Plant-Microbe Interact 9:600–607<br />

Simons M, Permentier HP, de Weger LA, Wijffelman CA, Lugtenberg BJJ (1997) Amino<br />

acid synthesis is necessary for tomato root colonization by Pseudomonas fluorescens<br />

strain WCS365. Mol Plant-Microbe Interact 10:102–106<br />

Singleton LL, Mihail JD, Rush CM (1992) Methods for research on soil-borne phytopathogenic<br />

fungi. American Phytopathological Society Press, St. Paul, Minnesota<br />

Sokal RR, Rohlf FJ (1981) Biometry: the principles and practice of statistics in biological<br />

research. Freeman, New York, pp 432–436<br />

Stanisich VA, Bennett PM, Richmond MH (1977) Characterization of a translocation unit<br />

encoding resistance to mercuric ions that occurs on a nonconjugative plasmid in<br />

Pseudomonas aeruginosa. J Bacteriol 129:1227–1233<br />

Streit W, Kosch K, Werner D (1992) Nodulation competitiveness of Rhizobium leguminosarum<br />

bv. phaseoli and Rhizobium tropici strains measured by glucuronidase (gus)<br />

gene fusion. Biol Fertil Soils 14:140–144<br />

Teeri TH, Lehvaslaiho H, Franck M, Uotila J, Heino P, Palva ET,Van Montagu M, Herrera-<br />

Estrella L (1989) Gene fusions to lacZ reveal new expression patterns of chimeric<br />

genes in transgenic <strong>plant</strong>s. EMBO J 8:343–350<br />

Timms-Wilson TM, Bailey MJ (2001) Reliable use of green fluorescent protein in fluorescent<br />

pseudomonads. J Microbiol Meth 46:77–80<br />

Tombolini R, van der Gaag DJ, Gerhardson B, Jansson JK (1999) Colonization pattern of<br />

the biocontrol strain Pseudomonas chlororaphis MA 342 on barley seeds visualized by<br />

using green fluorescent protein. Appl Environ Microbiol 65:3674–3680<br />

van der Bij AJ, de Weger LA, Tucker WT, Lugtenberg BJJ (1996) Plasmid stability in<br />

Pseudomonas fluorescens in the rhizosphere. Appl Environ Microbiol 62:1076–1080<br />

Voorhorst WGB, Eggen RIL, Luesink EJ, de Vos WM (1995) Characterization of the celB<br />

gene coding for b-glucosidase from the hyperthermophilic archaeon Pyrococcus<br />

furiosus and its expression and mutation analysis in Escherichia coli. J Bacteriol<br />

177:7105–7111<br />

Ward DM (1989) Molecular probes for analysis of microbial communities. In: Characklis<br />

WG, Wilderer PA (eds) Structure and function of biofilms. Wiley, New York, pp<br />

145–155<br />

Warren TM, Williams V, Flechcher M (1992) Influence of solid <strong>surface</strong>, adhesive ability,<br />

and inoculum size on bacterial colonization in microcosm studies. Appl Environ<br />

Microbiol 58:2954–2959<br />

Weller DM (1988) Biological control of soilborne <strong>plant</strong> pathogens in the rhizosphere<br />

with bacteria. Annu Rev Phytopathol 26:379–407<br />

Winstanley G, Morgan JAW, Pickup RW, Saunders JR (1991) Use of a xylE marker gene to<br />

monitor survival of recombinant Pseudomonas putida populations in lake water by<br />

culture on nonselective media. Appl Environ Microbiol 57:1905–1913


3 Methanogenic Microbial Communities Associated<br />

with Aquatic Plants<br />

Ralf Conrad<br />

1 Introduction<br />

Methanogenic microbial communities are typically active at anoxic sites that<br />

are depleted in electron acceptors other than CO 2 and H + .At these sites CH 4 is<br />

one of the major products of degradation of organic matter. The degradation<br />

products of cellulose, for example, which has an oxidation state of zero, would<br />

be CH 4 and CO 2 in a ratio of 1:1. Organic matter with a higher or lower oxidation<br />

state would yield respectively less or more CH 4 (Yao and Conrad 2000).<br />

Consequently, anoxic methanogenic habitats can be significant sources in the<br />

global CH 4 cycle. The global CH 4 cycle is important with respect to atmospheric<br />

chemistry and climate, since CH 4 is an important greenhouse gas and<br />

has tripled in abundance over the last two centuries (Cicerone and Oremland<br />

1988; Ehhalt 1999). The most important individual source for atmospheric<br />

CH 4 is wetlands (including flooded rice fields), which account for about<br />

175 Tg CH 4 per year or 33 % of the total atmospheric CH 4 budget (Conrad<br />

1997; Aulakh et al. 2001). The general <strong>microbiology</strong> and that of methanogenic<br />

microbial communities in flooded soils has recently been reviewed in detail<br />

(Kimura 2000; Liesack et al. 2000; Conrad and Frenzel 2002). In the following<br />

I will concentrate on methanogenic microbial communities associated with<br />

aquatic <strong>plant</strong>s.<br />

2 Role of Plants in Emission of CH 4 to the Atmosphere<br />

Aquatic <strong>plant</strong>s are an integral part of wetland ecosystems that emit CH 4 into<br />

the atmosphere. Aquatic <strong>plant</strong>s interact in three different ways with the<br />

microbial CH 4 cycling, i.e., by serving as gas conduits, by supplying O 2 to the<br />

rhizosphere and by supplying organic substrates to the soil (Fig. 1).<br />

Aquatic <strong>plant</strong>s live in anoxic soil habitats and thus have to make sure that<br />

their roots are supplied with O 2. The supply of O 2 is accomplished by vascular<br />

gas transport and aerenchyma systems. These systems and their mode of<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


36<br />

CH 4<br />

Ralf Conrad<br />

O 2<br />

CH 4<br />

methanogenic<br />

substrates<br />

O 2<br />

straw &<br />

stubbles<br />

C<br />

sloughed-off cells<br />

exudates<br />

B<br />

aerenchymatous<br />

leaf sheeth<br />

Fig. 1. Role of aquatic <strong>plant</strong>s for cycling of CH 4 by serving as gas conduits (C): cross section<br />

through an aerenchymatous leaf sheath, by supplying O 2 to the rhizosphere (B), and<br />

by supplying organic substrates to the soil; A, B and C are sites where O 2 is available<br />

(taken from Frenzel 2000)<br />

operation can be different in the different <strong>plant</strong> species (Armstrong 1979;<br />

Grosse et al. 1996; Jackson and Armstrong 1999). However, they all allow for<br />

transport of O 2 to the roots and vice-versa allow for the transport of CH 4 from<br />

the anoxic soil into the atmosphere. In rice fields, up to about 90 % of total<br />

CH 4 emission can be accomplished by ventilation through the rice <strong>plant</strong>s<br />

(Holzapfel-Pschorn et al. 1986; Aulakh et al. 2001). The exact contribution of<br />

rice <strong>plant</strong>s to the transport of CH 4 from the soil into the atmosphere depends<br />

on the size of the rice <strong>plant</strong>s and their capacity for gas transport (Aulakh et al.<br />

A


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 37<br />

2001). Other aquatic <strong>plant</strong>s have similar features (Chanton and Dacey 1991;<br />

Grosse et al. 1996). Due to ventilation through aquatic <strong>plant</strong>s, only a few bubbles<br />

accumulate in the soil and the ratio of CH 4 to N 2 in soil gas is relatively<br />

low (Chanton and Dacey 1991). Transport through the aquatic <strong>plant</strong>s results<br />

in the fractionation of the stable isotope composition of CH 4 (delaying transport<br />

of heavy carbon and hydrogen), the extent being dependent on the mode<br />

of gas transport, e.g., by molecular diffusion or thermo-osmosis (Chanton<br />

and Dacey 1991; Chanton and Whiting 1996).<br />

By supplying O 2 to the rhizosphere, aquatic <strong>plant</strong>s create a habitat there<br />

that is partially oxic. The presence of O 2 and increase in the redox potential<br />

have been demonstrated in the rhizosphere of aquatic <strong>plant</strong>s (Frenzel 2000).<br />

However, O 2 availability may be spatially and temporarily restricted. Leakage<br />

of O 2 from the roots only occurs at specific sites, e.g., at the tips and where lateral<br />

roots emerge (Armstrong 1967).As the root grows, the soil sites which are<br />

affected by O 2 leakage change also (Flessa and Fischer 1992; Flessa 1994). A<br />

further factor, which affects the availability of O 2 , is microbial respiration of<br />

organic substrates, which also varies in time and space. Availability of useful<br />

substrates can dramatically limit the availability of O 2 in the rhizosphere (Van<br />

Bodegom et al. 2001a, b). The creation of oxic microhabitats may have dramatic<br />

effects on methanogenic microbial communities that also occur in the<br />

rhizosphere. First, O 2 is probably toxic to most of the anaerobic microorganisms<br />

and to methanogenic ones in particular (see below). Second, availability<br />

of O 2 allows the microbial and/or chemical oxidation of reduced inorganic<br />

compounds such as ammonia, sulfide and ferrous iron. These oxidation activities<br />

in turn result in the availability of inorganic oxidants and an increase of<br />

the redox potential in the rhizosphere beyond the zone where molecular O 2 is<br />

available. The availability of nitrate, sulfate and ferric iron, in turn, allows the<br />

operation of microbial nitrate reduction, sulfate reduction and iron reduction<br />

interfering with the activity of methanogenesis (Conrad 1993; Conrad and<br />

Frenzel 2002). Most of all, however, availability of O 2 allows the partial oxidation<br />

of CH 4 produced by the methanogenic microbial community. In fact, a<br />

significant percentage of the CH 4 produced in the anoxic soil and/or the rhizosphere<br />

is oxidized by methanotrophic bacteria (Frenzel 2000). The methanotrophic<br />

bacteria live by oxidation of CH 4 with O 2 to CO 2 and thus depend on<br />

the availability of both CH 4 and O 2 . The methanotrophic activity in the rhizosphere<br />

of aquatic <strong>plant</strong>s scavenges a significant part of the produced CH 4<br />

which otherwise would be emitted into the atmosphere. The percentage of<br />

oxidized CH 4 varies with circumstances, but is typically in the order of 30 % of<br />

the CH 4 produced (Frenzel 2000). The CH 4 that escapes oxidation is generally<br />

enriched in isotopically heavy carbon (Chanton et al. 1997; Tyler et al. 1997;<br />

Krüger et al. 2002). The methanotrophs live directly on the root <strong>surface</strong>, partially<br />

even penetrating into the root (Gilbert et al. 1998), but may also be<br />

active a short distance away from the root <strong>surface</strong> if O 2 is available (Van Bodegom<br />

et al. 2001a).


38<br />

Ralf Conrad<br />

Finally, aquatic <strong>plant</strong>s stimulate the methanogenic microbial communities<br />

in the rhizosphere and the bulk soil by providing additional organic substrates<br />

that can be methanogenically degraded. Theoretically, we may expect<br />

two different classes of organic substrates that originate from the <strong>plant</strong>s, i.e.,<br />

soluble exudates that are released from the roots briefly after being generated<br />

through photosynthesis, and structural organic matter provided by <strong>plant</strong><br />

debris.<br />

3 Role of Photosynthates and Plant Debris<br />

for CH 4 Production<br />

Field observations suggested that root exudate-driven CH 4 production might<br />

play a major role in CH 4 emission from flooded rice fields (Holzapfel-<br />

Pschorn et al. 1986). Another preliminary indication of photosynthesis<br />

affecting CH 4 production came from field observations that CH 4 emission<br />

from various wetlands correlates with primary productivity (Whiting and<br />

Chanton 1993; Joabsson and Christensen 2001) and that CO 2 enrichment of<br />

the atmosphere results in increased CH 4 emission (Dacey et al. 1994; Hutchin<br />

et al. 1995; Megonigal and Schlesinger 1997; Ziska et al. 1998). However, CO 2<br />

enrichment and increased temperature caused in Florida rice fields a<br />

decreased CH 4 emission, probably because of enhanced delivery of O 2 into<br />

the rhizosphere (Schrope et al. 1999). This study is in contrast to that by<br />

Ziska et al. (1998) on rice fields in the Philippines, and shows that field<br />

observations have to be interpreted with care due to the highly complex<br />

interactions in the ecosystem.<br />

However, there are more direct indications that <strong>plant</strong> photosynthesis<br />

affects the methanogenic microbial community in the rhizosphere. For example,<br />

CH 4 production is correlated to the extent of root exudation in rice<br />

(Aulakh et al. 2001). Pulse labeling studies with rice and other aquatic <strong>plant</strong>s<br />

have shown that different percentages (acetate>CH 4, indicates the<br />

degradation pathway of the excreted organic substrate to CH 4.<br />

Other pulse-labeling studies have shown that photosynthate-derived CH 4<br />

contributes more than 50 % to the total CH 4 emission from flooded rice fields<br />

(Minoda et al. 1996; Watanabe et al. 1999). These studies also confirmed the<br />

speculations from earlier field work (Holzapfel-Pschorn et al. 1986; Schütz et<br />

al. 1989) that seasonal peaks in CH 4 emission were due to decomposition of<br />

rice straw, followed by stimulation through root exudation and finally<br />

through decay of roots (Fig. 3).


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 39<br />

Fig. 2. Transfer of carbon via rice<br />

<strong>plant</strong>s to the soil and into CH 4 .<br />

Above After pulse labeling of the<br />

rice <strong>plant</strong>s with 14 CO 2 , radioactivity<br />

transiently accumulates in soil<br />

organic compounds and is ultimately<br />

converted to 14 CH 4 ; below<br />

specific radioactivities indicate<br />

that radioactive compounds are<br />

converted in the sequence lactate>propionate>acetate>CH<br />

4<br />

(taken from Dannenberg and Conrad<br />

1999)<br />

Radioactivity [Bq ml -1 ]<br />

Specific radioactivity [Bq nmol -1 ]<br />

500<br />

400<br />

300<br />

200<br />

100<br />

Experiment #1<br />

lactate<br />

propionate<br />

acetate<br />

CH 4<br />

0<br />

0 2 4 6 8 10 12 14 16<br />

Time [d]<br />

0<br />

0 2 4 6<br />

Besides the more direct effect of photosynthesis through root exudation,<br />

CH 4 production in wetlands is furthermore stimulated by <strong>plant</strong> debris. This<br />

may be decaying roots or dead aboveground <strong>plant</strong> material. In rice fields, for<br />

example, rice straw from the previous season is often plowed under to<br />

improve soil quality. Also, composted <strong>plant</strong> material is used as soil fertilizer.<br />

There are numerous studies which show that addition of such organic matter<br />

dramatically increases CH 4 emission rates (Denier van der Gon 1999).<br />

Decomposition of isotopically labeled rice straw contributes significantly to<br />

production and emission of CH 4 during the early season (Chidthaisong and<br />

Watanabe 1997; Watanabe et al. 1998, 1999)<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

Time [d]<br />

A<br />

lactate<br />

propionate<br />

acetate<br />

CO 2<br />

CH 4<br />

B


40<br />

Ralf Conrad<br />

Fig. 3. Emission of CH 4 from strawfertilized,<br />

<strong>plant</strong>ed rice field soil and<br />

partitioning of the primary carbon<br />

from which CH 4 was formed, determined<br />

by using 13 C-labeled CO 2 , rice<br />

straw and soil organic matter; rice<br />

<strong>plant</strong> C1 is carbon released within<br />

2 weeks after assimilation of 13 CO 2 ;<br />

rice <strong>plant</strong> C2 is other <strong>plant</strong>-derived<br />

carbon, presumably from sloughed-off<br />

root cells or decaying roots (taken<br />

from Watanabe et al. 1999)<br />

4 Methanogenic Microbial Communities on Plant Debris<br />

Straw and decomposing roots are important <strong>plant</strong> debris in flooded rice<br />

fields. Rice straw mainly consists of cellulose, hemicellulose and lignin and<br />

is encrusted with silica (Tsutsuki and Ponnamperuma 1987; Watanabe et al.<br />

1993). After a rapid mineralization of 80–90 % of the straw during the first<br />

year, a more resistant fraction of organic matter remains. The latter is<br />

degraded slowly with a half-life of about 2 years (Neue and Scharpenseel<br />

1987). Rice straw is colonized by microorganisms and the structure of the<br />

leave blades and sheaths gradually disintegrates. The degradation process<br />

becomes visually apparent after about 3 weeks (Kimura and Tun 1999; Tun<br />

and Kimura 2000) with a dry weight loss of about 50 % during the first<br />

30 days (Glissmann and Conrad 2002). Degradation of rice straw proceeds<br />

via hydrolysis, fermentation of the released sugars, syntrophic conversion of<br />

primary fermentation products to acetate, CO 2 and H 2, and conversion of<br />

acetate and H 2/CO 2, respectively, to CH 4 (Glissmann and Conrad 2002). The<br />

same degradation pathway is generally found in methanogenic environments<br />

such as lake sediments or anaerobic digestors (Zinder 1993). The<br />

methanogenic degradation pathway of rice straw is similar to that of the<br />

organic matter present in flooded soil to which no rice straw was added, but<br />

the rate of CH 4 production is lower in the unamended soil (Glissmann and


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 41<br />

Conrad 2000). Under steady state conditions, the conversion of rice straw to<br />

CH 4 is limited by the hydrolysis of the straw polysaccharides, which become<br />

increasingly recalcitrant to decomposition (Glissmann and Conrad 2002). It<br />

is likely that rice straw is in this way gradually converted to soil organic matter<br />

and humus.<br />

The bacteria that colonize and degrade rice straw mainly consist of<br />

clostridia (Weber et al. 2001a) which belong to the same taxonomic clusters<br />

as found in unamended soil (Chin et al. 1999; Hengstmann et al. 1999; Lüdemann<br />

et al. 2000). On the other hand, the community of methanogenic<br />

archaea on rice straw is less diverse and abundant than in the bulk soil<br />

(Weber et al. 2001b). The genus Methanosaeta, in particular, was lacking in<br />

degrading straw. This genus is common in bulk soil (Grosskopf et al. 1998)<br />

and especially becomes abundant at limiting acetate concentrations (Fey and<br />

Conrad 2000). Consistent with the low abundance of methanogens on rice<br />

straw is the observation that straw pieces retrieved from the soil mainly<br />

exhibit fermentative production of H 2 and fatty acids, while the subsequent<br />

conversion of the fatty acids to CH 4 takes place in the bulk soil to where the<br />

fatty acids are released (Glissmann et al. 2001). Hence, methanogenic degradation<br />

of rice straw is compartmentalized in a way that methanogenesis<br />

occurs in the soil at some distance to the microbial community that colonizes<br />

the straw (Fig. 4).<br />

Fig. 4. Conceptual model<br />

of methanogenic degradation<br />

of rice straw, and<br />

the localization of the<br />

major processes either<br />

on the straw or in the soil<br />

slurry (taken from Glissmann<br />

et al. 2001)<br />

Slurry<br />

Straw<br />

Biopolymers<br />

Hydrolysis<br />

of polymers<br />

Monoand<br />

oligomers<br />

Fermentation<br />

Fatty acids<br />

and alcohols<br />

Fermentation<br />

and syntrophic<br />

degradation<br />

Homoacetogenesis<br />

H 2 + CO2<br />

Acetate<br />

Methanogenesis<br />

CH 4


42<br />

Ralf Conrad<br />

The general colonization patterns of rice roots with microorganisms and<br />

their potential involvement in degradation of the dead roots have been<br />

reviewed by Kimura (2000). It is likely that dead roots are decomposed in a<br />

similar way to rice straw, but detailed studies are lacking.<br />

5 Methanogenic Microbial Communities on Roots<br />

Plant roots are apparently colonized by methanogenic microorganisms. This<br />

evidence came from incubation of excised roots of aquatic <strong>plant</strong>s under<br />

anoxic conditions resulting in substantial CH 4 production (Kimura et al. 1991;<br />

King 1994; Frenzel and Bosse 1996). Subsequently, it was shown by Grosskopf<br />

et al. (1998) that rice roots are indeed inhabited by a diverse community of<br />

methanogenic archaea, which can be retrieved by DNA extraction, and amplification<br />

of the archaeal SSU rRNA genes. Archaeal diether lipids were also<br />

detected on rice roots (Reichardt et al. 1997).<br />

Production of CH 4 on rice roots is dominated by H 2/CO 2-utilizing<br />

methanogens (Lehmann-Richter et al 1999; Conrad et al. 2000). The most<br />

prominent group of methanogens on rice roots is that of the uncultivated rice<br />

cluster I (Grosskopf et al. 1998). Since this cluster is also dominant in soils in<br />

which CH 4 is exclusively produced from H 2 /CO 2 (Fey et al. 2000) and in<br />

methanogenic enrichment cultures on H 2/CO 2 (Lueders et al. 2001), it is likely<br />

that it is responsible for the observed H 2/CO 2 dependent methanogenesis on<br />

rice roots. However, methanogens belonging to Methanobacteriaceae and<br />

Methanomicrobiaceae, i.e., groups that are able to utilize H 2/CO 2, have also<br />

been detected (Grosskopf et al. 1998). Populations of acetoclastic Methanosarcina,<br />

on the other hand, only developed at a later stage of anoxic incubation<br />

of excised rice roots, when sufficient acetate had accumulated and only in the<br />

absence of phosphate. Phosphate concentrations higher than 10 mM were<br />

found to prohibit the activity of acetoclastic methanogenesis (Conrad et al.<br />

2000). Collectively, these observations suggest that the methanogenic flora in<br />

situ produces CH 4 mainly from H 2/CO 2 rather than from acetate. This is a<br />

major difference to the behavior in the soil, where acetate is the dominant<br />

methanogenic substrate.<br />

Consequently, the stable isotopic signature of the produced CH 4 was<br />

found to be different for the methanogenic microbial communities in the<br />

soil and on the roots (Conrad et al. 2000, 2002). This fact may have implications<br />

for estimates dealing with the budget of atmospheric CH 4 and the<br />

global CH 4 cycle, for which the stable isotopic signature of CH 4 is an important<br />

constraint (Stevens 1993). Unfortunately, we presently do not know how<br />

much the methanogenic microbial community on rice roots, or on the roots<br />

of aquatic <strong>plant</strong>s in general, contribute to the CH 4 source strength of wetland<br />

ecosystems compared to the methanogenic microbial communities in the<br />

anoxic soil.


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 43<br />

Another implication of the observations concerns the structure of the<br />

methanogenic microbial community on the roots, which seem to be very simple,<br />

consisting only of H 2-producing fermenting and H 2-consuming methanogenic<br />

microorganisms. However, experiments with excised rice roots have<br />

demonstrated a more complex community of fermenting bacteria including<br />

vigorous fermentative production of acetate, propionate and butyrate (Conrad<br />

and Klose 1999, 2000). Interestingly, a significant percentage (up to 60 %)<br />

of these fatty acids was produced by reduction of CO 2. The stable isotope signature<br />

of the produced acetate was consistent with the production by CO 2<br />

reduction (Conrad et al. 2002). Acetate production from CO 2 indicates that<br />

homoactogenic bacteria were active, a likely conclusion, since homoacetogenic<br />

Sporomusa are members of the rice root microflora (Rosencrantz et al.<br />

1999). Homoactogens have also been found on the roots of sea grass (Küsel et<br />

al. 1999, 2001). Approximately 30 % of the root epidermal cells of sea grass<br />

were colonized with microorganisms that hybridized with an archaeal probe<br />

suggesting the presence of methanogens (Küsel et al. 1999). Presently, little is<br />

known about the fate of the produced fatty acids. Propionate and butyrate can<br />

potentially be further converted to acetate, CO 2 and H 2 by syntrophic bacteria,<br />

which are present in the anoxic rice soil, followed by H 2/CO 2-dependent<br />

methanogenesis (Krylova et al. 1997). Syntrophic oxidation of acetate, however,<br />

is unlikely since [2- 14 C]acetate was hardly turned over in root preparations<br />

(Lehmann-Richter et al. 1999). The most likely fate of the acetate produced<br />

by the root microflora is its escape into the bulk soil where it is<br />

methanogenically decomposed (Fig. 5). Alternatively, acetate may be a substrate<br />

for anaerobic bacteria using nitrate, ferric iron or sulfate as electron<br />

Fig. 5. Conceptual model of the<br />

localization of methanogenic<br />

archaea (MA), homoacetogenic<br />

bacteria (HAB), methane-oxidizing<br />

bacteria (MOB) and aerobic<br />

bacteria (AB) in vicinity of rice<br />

roots and to each other, and the<br />

flow of organic carbon. The insertion<br />

of lateral roots (and root<br />

tips) are the most likely sites<br />

where O 2 and organic substrates<br />

(e.g., sugars) are released into the<br />

soil


44<br />

Ralf Conrad<br />

acceptor. However, little is known about the activity of these functional<br />

groups on the roots of aquatic <strong>plant</strong>s (Bodelier et al. 1997; Nijburg and Laanbroek<br />

1997; King and Garey 1999; Küsel et al. 1999; Wind et al. 1999; Arth and<br />

Frenzel 2000).<br />

6 Interaction of Methanogens and Methanotrophs<br />

Although it has become evident that methanogenesis is stimulated by <strong>plant</strong><br />

photosynthesis (see above), it has been a rather unexpected result that<br />

methanogenic activity is obviously localized directly on the root <strong>surface</strong>. This<br />

result was surprising, since textbook knowledge suggests that methanogenic<br />

archaea need an absolutely O 2-free environment, which the root <strong>surface</strong> does<br />

not provide (see above). Quite the contrary, roots have been shown to be the<br />

site of methanotrophic activity (Frenzel 2000). This discrepancy between<br />

roots being colonized by both aerobic and anaerobic microorganisms has not<br />

been completely reconciled.<br />

One possible explanation is a spatially heterogeneous colonization of the<br />

roots. The aerobic methanotrophs would colonize only those parts where O 2<br />

is leaking from the roots and the methanogens only those that stay anoxic.<br />

However, methanogens would probably only colonize the roots if provision of<br />

the substrate (H 2) is better there than in the bulk soil. Production of H 2<br />

requires microbial fermentation activity and this in turn requires the provision<br />

of a degradable substrate. Hence, colonization of roots by methanogens<br />

is most likely at the sites with high leakage rates of organic substrates. The<br />

localization of sites with exudation of organic substrates along the root is not<br />

quite clear, but the root tips were found to be most actively excreting sucrose<br />

in an annual grass (Jaeger et al. 1999). However, root tips are also the most<br />

active sites of O 2 leakage (Armstrong 1967). Thus, we have to expect that the<br />

optimal sites for colonization by methanotrophs and methanogens are the<br />

same.<br />

Another possible explanation is that the methanogens are largely protected<br />

from O 2, because they are living in the vicinity of O 2-consuming methanotrophs.<br />

A similar close association of methanotrophs and methanogens has<br />

been hypothesized for pelagic microbial assemblages, thus explaining the formation<br />

of CH 4 in oxic ocean <strong>surface</strong> water (Sieburth 1991). Although<br />

Sieburth’s hypothesis has so far not been confirmed in pelagic microbial flocs<br />

(Ploug et al. 1997), experiments in microbial chemostat cultures have shown<br />

that anaerobic methanogens can co-exist with aerobic microorganisms under<br />

aerated conditions (Gerritse and Gottschal 1993).<br />

Moreover, at least some of the species of methanogens seem to be more<br />

resistant to exposure to O 2 than generally expected. For example,<br />

methanogens in rice field soil have been found to survive desiccation of the<br />

soil and prolonged exposure to air (Fetzer et al. 1993). Methanosarcina bark-


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 45<br />

eri was found to be able to initiate CH 4 production despite a positive redox<br />

potential of the medium (Fetzer and Conrad 1993). Methanogenic activity has<br />

been detected in just that part of the termite gut that is oxygenated (Brune<br />

and Friedrich 2000), and Methanobrevibacter isolates were able to grow<br />

slowly despite the presence of low O 2 concentrations (Leadbetter and Breznak<br />

1996). Recently, some methanogenic species were found to contain catalase<br />

and superoxide dismutase to protect against oxidative stress (Shima et al.<br />

1999, 2001; Brioukhanov et al. 2000). Unfortunately, we do not know the O 2<br />

resistance of the methanogenic species that inhabit rice roots, in particular<br />

the uncultivated rice cluster I methanogens (Lueders et al. 2001).<br />

As methanogens and methanotrophs live in the same environments in<br />

close vicinity, it might be possible that they communicate with each other not<br />

only by transfer of CH 4, but also more directly through gene exchange. R.K.<br />

Thauer (Germany) and his group have recently put forward this idea. It is<br />

indeed intriguing that methanotrophic bacteria contain genes and coenzymes,<br />

which had been postulated to be specific for methanogens. Thus,<br />

methanotrophs seem to contain tetrahydromethanopterin in addition to<br />

tetrahydrofolate, and utilize tetrahydromethanopterin-dependent enzymes<br />

for catabolic C1 transfer reactions similarly to the methanogens (Chisdoserdova<br />

et al. 1998; Vorholt et al. 1999). It is likely that methanotrophs have<br />

acquired the necessary genes from methanogens. The other way round,<br />

methanogens seem to have acquired genes that are of bacterial origin. Thus,<br />

Methanosarcina and Methanobrevibacter species contain a monofunctional<br />

catalase. Such an enzyme is unexpected for Archaea, which generally contain<br />

a bifunctional catalase (Shima et al. 2001). Roots of aquatic <strong>plant</strong>s would be a<br />

possible habitat where such gene transfers between methanogenic Archaea<br />

and methanotrophic Bacteria might occur.<br />

Acknowledgements. I thank Peter Frenzel and Rolf Thauer for discussion.<br />

References and Selected Reading<br />

Armstrong W (1967) The use of polarography in the assay of oxygen diffusing from<br />

roots in anaerobic media. Physiol Plantarum 20:540–553<br />

Armstrong W (1979) Aeration in higher <strong>plant</strong>s. Adv Bot Res 7:226–332<br />

Arth I, Frenzel P (2000) Nitrification and denitrification in the rhizosphere of rice: the<br />

detection of processes by a new multi-channel electrode. Biol Fertil Soils 31:427–435<br />

Aulakh MS, Wassmann R, Rennenberg H (2001) Methane emissions from rice fields –<br />

Quantification, mechanisms, role of management, and mitigation options.Adv Agron<br />

70:193–260<br />

Bodelier PLE, Wijlhuizen AG, Blom CWPM, Laanbroek HJ (1997) Effects of photoperiod<br />

on growth of and denitrification by Pseudomonas chlororaphis in the root zone of<br />

Glyceria maxima, studied in a gnotobiotic microcosm. Plant Soil 190:91–103


46<br />

Ralf Conrad<br />

Brioukhanov A, Netrusov A, Sordel M, Thauer RK, Shima S (2000) Protection of<br />

Methanosarcina barkeri against oxidative stress: identification and characterization<br />

of an iron superoxide dismutase. Arch Microbiol 174:213–216<br />

Brune A, Friedrich M (2000) Microecology of the termite gut: structure and function on<br />

a microscale [Review]. Curr Opin Microbiol 3:263–269<br />

Chanton JP, Dacey JW (1991) Effects of vegetation on methane flux, reservoirs, and carbon<br />

isotopic composition. In: Rogers JE, Whitman WB (eds) Trace gas emissions by<br />

<strong>plant</strong>s. Academic Press, San Diego, pp 65–92<br />

Chanton JP, Whiting GJ (1996) Methane stable isotopic distributions as indicators of gas<br />

transport mechanisms in emergent aquatic <strong>plant</strong>s. Aquat Bot 54:227–236<br />

Chanton JP, Whiting GJ, Blair NE, Lindau CW, Bollich PK (1997) Methane emission from<br />

rice: Stable isotopes, diurnal variations, and CO 2 exchange. Global Biogeochem Cycles<br />

11:15–27<br />

Chidthaisong A, Watanabe I (1997) Methane formation and emission from flooded rice<br />

soil incorporated with 13 C-labeled rice straw. Soil Biol Biochem 29:1173–1181<br />

Chin KJ, Hahn D, Hengstmann U, Liesack W, Janssen PH (1999) Characterization and<br />

identification of numerically abundant culturable bacteria from the anoxic bulk soil<br />

of rice paddy microcosms. Appl Environ Microbiol 65:5042–5049<br />

Chistoserdova L, Vorholt JA, Thauer RK, Lidstrom ME (1998) C-1 transfer enzymes and<br />

coenzymes linking methylotrophic bacteria and methanogenic archaea. Science<br />

281:99–102<br />

Cicerone RJ, Oremland RS (1988) Biogeochemical aspects of atmospheric methane.<br />

Global Biogeochem Cycles 2:299–327<br />

Conrad R (1993) Mechanisms controlling methane emission from wetland rice fields. In:<br />

Oremland RS (ed) The biogeochemistry of global change: radiative trace gases. Chapman<br />

and Hall, New York, pp 317–335<br />

Conrad R (1997) Production and consumption of methane in the terrestrial biosphere.<br />

In: Helas G, Slanina J, Steinbrecher R (eds) Biogenic volatile organic carbon compounds<br />

in the atmosphere. SBP Academic Publ, Amsterdam, pp 27–44<br />

Conrad R, Klose M (1999) Anaerobic conversion of carbon dioxide to methane, acetate<br />

and propionate on washed rice roots. FEMS Microbiol Ecol 30:147–155<br />

Conrad R, Klose M (2000) Selective inhibition of reactions involved in methanogenesis<br />

and fatty acid production on rice roots. FEMS Microbiol Ecol 34:27–34<br />

Conrad R, Frenzel P (2002) Flooded soils. In: Bitton G (ed) The encyclopedia of environmental<br />

<strong>microbiology</strong>. Wiley, New York, pp 1316.1333<br />

Conrad R, Klose M, Claus P (2000) Phosphate inhibits acetotrophic methanogenesis on<br />

rice roots. Appl Environ Microbiol 66:828–831<br />

Conrad R, Klose M, Claus P (2002) Pathway of CH 4 formation in anoxic rice field soil and<br />

rice roots determined by 13C-stable isotope fractionation. Chemosphere 47:797–806<br />

Dacey JWH, Drake BG, Klug MJ (1994) Stimulation of methane emission by carbon dioxide<br />

enrichment of marsh vegetation. Nature 370:47–49<br />

Dannenberg S, Conrad R (1999) Effect of rice <strong>plant</strong>s on methane production and rhizospheric<br />

metabolism in paddy soil. Biogeochemistry 45:53–71<br />

Denier van der Gon H (1999) Changes in CH 4 emission from rice fields from 1960 to<br />

1990s – 2. The declining use of organic inputs in rice farming. Global Biogeochem<br />

Cycles 13:1053–1062<br />

Ehhalt DH (1999) Gas phase chemistry in the troposphere. In: Zellner R (ed) Global<br />

aspects of atmospheric chemistry. Springer, Berlin Heidelberg New York, pp 21–109<br />

Fetzer S, Conrad R (1993) Effect of redox potential on methanogenesis by Methanosarcina<br />

barkeri. Arch Microbiol 160:108–113<br />

Fetzer S, Bak F, Conrad R (1993) Sensitivity of methanogenic bacteria from paddy soil to<br />

oxygen and desiccation. FEMS Microbiol Ecol 12:107–115


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 47<br />

Fey A, Conrad R (2000) Effect of temperature on carbon and electron flow and on the<br />

archaeal community in methanogenic rice field soil. Appl Environ Microbiol<br />

66:4790–4797<br />

Flessa H (1994) Plant-induced changes in the redox potential of the rhizospheres of the<br />

submerged vascular macrophytes Myriophyllum verticillatum L and Ranunculus circinatus<br />

L. Aquat Bot 47:119–129<br />

Flessa H, Fischer WR (1992) Plant-induced changes in the redox potentials of rice rhizospheres.<br />

Plant Soil 143:55–60<br />

Frenzel P (2000) Plant-associated methane oxidation in rice fields and wetlands<br />

[Review]. Adv Microb Ecol 16:85–114<br />

Frenzel P, Bosse U (1996) Methyl fluoride, an inhibitor of methane oxidation and<br />

methane production. FEMS Microbiol Ecol 21:25–36<br />

Gerritse J, Gottschal JC (1993) Two-membered mixed cultures of methanogenic and aerobic<br />

bacteria in O 2 -limited chemostats. J Gen Microbiol 139:1853–1860<br />

Gilbert B, Assmus B, Hartmann A, Frenzel P (1998) In situ localization of two methanotrophic<br />

strains in the rhizosphere of rice <strong>plant</strong>s. FEMS Microbiol Ecol 25:117–128<br />

Glissmann K, Conrad R (2000) Fermentation pattern of methanogenic degradation of<br />

rice straw in anoxic paddy soil. FEMS Microbiol Ecol 31:117–126<br />

Glissmann K, Conrad R (2002) Saccharolytic activity and its role as a limiting step in<br />

methane formation during the anaerobic degradation of rice straw in rice paddy soil.<br />

Biology and Fertility of Soils 35:62–67<br />

Glissmann K, Weber S, Conrad R (2001) Localization of processes involved in methanogenic<br />

degradation of rice straw in anoxic paddy soil. Environ Microbiol 3:502–511<br />

Grosse W, Armstrong J, Armstrong W (1996) A history of pressurised gas-flow studies in<br />

<strong>plant</strong>s. Aquat Bot 54:87–100<br />

Großkopf R, Stubner S, Liesack W (1998) Novel euryarchaeotal lineages detected on rice<br />

roots and in the anoxic bulk soil of flooded rice microcosms. Appl Environ Microbiol<br />

64:4983–4989<br />

Hengstmann U, Chin KJ, Janssen PH, Liesack W (1999) Comparative phylogenetic<br />

assignment of environmental sequences of genes encoding 16S rRNA and numerically<br />

abundant culturable bacteria from an anoxic rice paddy soil. Appl Environ<br />

Microbiol 65:5050–5058<br />

Holzapfel-Pschorn A, Conrad R, Seiler W (1986) Effects of vegetation on the emission of<br />

methane from submerged paddy soil. Plant and Soil 92:223–233<br />

Hutchin PR, Press MC, Lee JA,Ashenden TW (1995) Elevated concentrations of CO 2 may<br />

double methane emissions from mires. Global Change Biol 1:125–128<br />

Jackson MB, Armstrong W (1999) Formation of aerenchyma and the processes of <strong>plant</strong><br />

ventilation in relation to soil flooding and submergence [review]. Plant Biol 1:274–<br />

287<br />

Jaeger CH III, Lindow SE, Miller S, Clark E, Firestone MK (1999) Mapping of sugar and<br />

amino acid availability in soil around roots with bacterial sensors of sucrose and<br />

tryptophan. Appl Environ Microbiol 65:2685–2690<br />

Joabsson A, Christensen TR (2001) Methane emissions from wetlands and their relationship<br />

with vascular <strong>plant</strong>s: an Arctic example. Global Change Biol 7:919–932<br />

Kimura M (2000) Anaerobic <strong>microbiology</strong> in waterlogged rice fields. In: Bollag JM,<br />

Stotzky G (eds) Soil biochemistry, vol 10. Marcel Dekker, New York, pp 35–138<br />

Kimura M, Tun CC (1999) Microscopic observation of the decomposition process of leaf<br />

sheath of rice straw and colonizing microorganisms during the cultivation period of<br />

paddy rice. Soil Sci Plant Nutr 45:427–437<br />

Kimura M, Murakami H, Wada H (1991) CO 2 ,H 2 ,and CH 4 production in rice rhizosphere.<br />

Soil Sci Plant Nutr 37:55–60<br />

King GM (1994) Associations of methanotrophs with the roots and rhizomes of aquatic<br />

vegetation. Appl Environ Microbiol 60:3220–3227


48<br />

Ralf Conrad<br />

King GM, Garey MA (1999) Ferric iron reduction by bacteria associated with the roots of<br />

freshwater and marine macrophytes. Appl Environ Microbiol 65:4393–4398<br />

King JY, Reeburgh WS (2002) A pulse-labeling experiment to determine the contribution<br />

of recent <strong>plant</strong> photosynthates to net methane emission in arctic wet sedge tundra.<br />

Soil Biol Biochem 34:173–180<br />

Krüger M, Eller G, Conrad R, Frenzel P (2002) Seasonal variation in pathways of CH 4 production<br />

and in CH 4 oxidation in rice fields determined by stable isotopes and specific<br />

inhibitors. Global Change Biol 8:265–280<br />

Krylova NI, Janssen PH, Conrad R (1997) Turnover of propionate in methanogenic<br />

paddy soil. FEMS Microbiol Ecol 23:107–117<br />

Küsel K, Pinkart HC, Drake HL, Devereux R (1999) Acetogenic and sulfate-reducing bacteria<br />

inhabiting the rhizoplane and deep cortex cells of the sea grass Halodule<br />

wrightii. Appl Environ Microbiol 65:5117–5123<br />

Küsel K, Karnholz A, Trinkwalter T, Devereux R, Acker G, Drake HL (2001) Physiological<br />

ecology of Clostridium glycolicum RD-1, an aerotolerant acetogen isolated from sea<br />

grass roots. Appl Environ Microbiol 67:4734–4741<br />

Leadbetter JR, Breznak JA (1996) Physiological ecology of Methanobrevibacter cuticularis<br />

sp. nov and Methanobrevibacter curvatus sp. nov, isolated from the hindgut of<br />

the termite Reticulitermes flavipes. Appl Environ Microbiol 62:3620–3631<br />

Lehmann-Richter S, Großkopf R, Liesack W, Frenzel P, Conrad R (1999) Methanogenic<br />

archaea and CO 2 -dependent methanogenesis on washed rice roots. Environ Microbiol<br />

1:159–166<br />

Liesack W, Schnell S, Revsbech NP (2000) Microbiology of flooded rice paddies<br />

[Review]. FEMS Microbiol Rev 24:625–645<br />

Lüdemann H,Arth I, Liesack W (2000) Spatial changes in the bacterial community structure<br />

along a vertical oxygen gradient in flooded paddy soil cores. Appl Environ<br />

Microbiol 66:754–762<br />

Lueders T, Chin KJ, Conrad R, Friedrich M (2001) Molecular analyses of methyl-coenzyme<br />

M reductase alpha-subunit (mcrA) genes in rice field soil and enrichment cultures<br />

reveal the methanogenic phenotype of a novel archaeal lineage. Environ Microbiol<br />

3:194–204<br />

Megonigal JP, Schlesinger WH (1997) Enhanced CH 4 emissions from a wetland soil<br />

exposed to elevated CO 2 . Biogeochemistry 37:77–88<br />

Megonigal JP,Whalen SC, Tissue DT, Bovard BD,Albert DB,Allen AS (1999) A <strong>plant</strong>-soilatmosphere<br />

microcosm for tracing radiocarbon from photosynthesis through<br />

methanogenesis. Soil Sci Soc Am J 63:665–671<br />

Minoda T, Kimura M,Wada E (1996) Photosynthates as dominant source of CH 4 and CO 2<br />

in soil water and CH 4 emitted to the atmosphere from paddy fields. J Geophys Res<br />

101:21091–21097<br />

Neue HU, Scharpenseel HW (1987) Decomposition pattern of 14 C-labeled rice straw in<br />

aerobic and submerged rice soils of the Philippines. Sci Total Environ 62:431–434<br />

Nijburg JW, Laanbroek HJ (1997) The fate of 15 N-nitrate in healthy and declining Phragmites<br />

australis stands. Microb Ecol 34:254–262<br />

Ploug H, Kühl M, Buchholz-Cleven B, Joergensen BB (1997) Anoxic aggregates – an<br />

ephemeral phenomenon in the pelagic environment? Aquat Microb Ecol 13:285–294<br />

Reichardt W, Mascarina G, Padre B, Doll J (1997) Microbial communities of continuously<br />

cropped, irrigated rice fields. Appl Environ Microbiol 63:233–238<br />

Rosencrantz D, Rainey FA, Janssen PH (1999) Culturable populations of Sporomusa spp.<br />

and Desulfovibrio spp. in the anoxic bulk soil of flooded rice microcosms. Appl Environ<br />

Microbiol 65:3526–3533<br />

Schrope MK, Chanton JP, Allen LH, Baker JT (1999) Effect of CO 2 enrichment and elevated<br />

temperature on methane emissions from rice, Oryza sativa. Global Change Biology<br />

5:587–599


3 Methaogenic Microbial Communities Asociated with Aquatic Plants 49<br />

Schütz H, Holzapfel-Pschorn A, Conrad R, Rennenberg H, Seiler W (1989) A 3-year continuous<br />

record on the influence of daytime, season, and fertilizer treatment on<br />

methane emission rates from an Italian rice paddy. J Geophys Res 94:16405–16416<br />

Shima S, Netrusov A, Sordel M, Wicke M, Hartmann GC, Thauer RK (1999) Purification,<br />

characterization, and primary structure of a monofunctional catalase from Methanosarcina<br />

barkeri. Arch Microbiol 171:317–323<br />

Shima S, Sordel-Klippert M, Brioukhanov A, Netrusov A, Linder D, Thauer RK (2001)<br />

Characterization of a heme-dependent catalase from Methanobrevibacter arboriphilus.<br />

Appl Environ Microbiol 67:3041–3045<br />

Sieburth JM (1991) Methane and hydrogen sulfide in the pycnocline: a result of tight<br />

coupling of photosynthetic and “benthic” processes in stratified waters. In: Rogers JE,<br />

Whitman WB (eds) Microbial production and consumption of greenhouse gases:<br />

methane, nitrogen oxides, and halomethanes. American Society for Microbiology,<br />

Washington, DC, pp 147–174<br />

Stevens CM (1993) Isotopic abundances in the atmosphere and sources. In: Khalil MAK<br />

(ed) Atmospheric methane: sources, sinks, and role in global change, Springer, Berlin<br />

Heidelberg New York, pp 62–88<br />

Tsutsuki K, Ponnamperuma FN (1987) Behavior of anaerobic decomposition products<br />

in submerged soils . Effects of organic material amendment, soil properties, and temperature.<br />

Soil Sci Plant Nutr 33:13–33<br />

Tun CC, Kimura M (2000) Microscopic observation of the decomposition process of leaf<br />

blade of rice straw and colonizing microorganisms in a Japanese paddy field soil during<br />

the cultivation period of paddy rice. Soil Sci Plant Nutr 46:127–137<br />

Tyler SC, Bilek RS, Sass RL, Fisher FM (1997) Methane oxidation and pathways of production<br />

in a Texas paddy field deduced from measurements of flux, d 13 C, and dD of<br />

CH 4 . Global Biogeochem Cycles 11:323–348<br />

Van Bodegom P, Goudriaan J, Leffelaar P (2001a) A mechanistic model on methane oxidation<br />

in a rice rhizosphere. Biogeochem 55:145–177<br />

Van Bodegom P, Stams F, Mollema L, Boeke S, Leffelaar P (2001b) Methane oxidation and<br />

the competition for oxygen in the rice rhizosphere. Appl Environ Microbiol 67:3586–<br />

3597<br />

Vorholt JA, Chistoserdova L, Stolyar SM, Thauer RK, Lidstrom ME (1999) Distribution of<br />

tetrahydromethanopterin-dependent enzymes in methylotrophic bacteria and phylogeny<br />

of methenyl tetrahydromethanopterin cyclohydrolases. J Bacteriol<br />

181:5750–5757<br />

Watanabe A, Katoh K, Kimura M (1993) Effect of rice straw application on CH 4 emission<br />

from paddy fields. 2. contribution of organic constituents in rice straw. Soil Sci Plant<br />

Nutr 39:707–712<br />

Watanabe A, Yoshida M, Kimura M (1998) Contribution of rice straw carbon to CH 4<br />

emission from rice paddies using 13 C-enriched rice straw. J Geophys Res 103:8237–<br />

8242<br />

Watanabe A, Takeda T, Kimura M (1999) Evaluation of origins of CH 4 carbon emitted<br />

from rice paddies. J Geophys Res 104:23623–23629<br />

Weber S, Stubner S, Conrad R (2001a) Bacterial populations colonizing and degrading<br />

rice straw in anoxic paddy soil. Appl Environ Microbiol 67:1318–1327<br />

Weber S, Lueders T, Friedrich MW, Conrad R (2001b) Methanogenic populations<br />

involved in the degradation of rice straw in anoxic paddy soil. FEMS Microbiol Ecol<br />

38:11–20<br />

Whiting GJ, Chanton JP (1993) Primary production control of methane emission from<br />

wetlands. Nature 364:794–795<br />

Wind T, Stubner S, Conrad R (1999) Sulphate-reducing bacteria in rice field soil and on<br />

rice roots. Syst Appl Microbiol 22:269–279


50<br />

Ralf Conrad<br />

Yao H, Conrad R (2000) Electron balance during steady-state production of CH 4 and CO 2<br />

in anoxic rice soil. Eur J Soil Sci 51:369–378<br />

Zinder SH (1993) Physiological ecology of methanogens. In: Ferry JG (ed) Methanogenesis:<br />

ecology, physiology, biochemistry and genetics. Chapman and Hall, New York, pp<br />

128–206<br />

Ziska LH, Moya TB,Wassmann R, Namuco OS, Lantin RS,Aduna JB,Abao E, Bronson KF,<br />

Neue HU, Olszyk D (1998) Long-term growth at elevated carbon dioxide stimulates<br />

methane emission in tropical paddy rice. Global Change Biology 4:657–665


4 Role of Functional Groups of Microorganisms on<br />

the Rhizosphere Microcosm Dynamics<br />

Galdino Andrade<br />

1 Introduction<br />

This chapter discusses the role of functional microorganism groups that live<br />

in the rhizosphere and contribute to nutrient cycling. Soil ecology has much<br />

to contribute to our knowledge of important processes at the ecosystem<br />

level, such as how <strong>plant</strong> growth is affected by the rhizosphere biota, organic<br />

matter dynamics and nutrient cycling, and soil structure dynamics (Brussaard<br />

1998).<br />

Many groups work directly on <strong>plant</strong> nutrition, such as rhizobia and mycorrhiza<br />

fungi which are symbiotic. These groups have been studied extensively<br />

in the last few decades, but little has been investigated about the relationship<br />

between other functional groups, notwithstanding that many other interactions<br />

exist in the rhizosphere that are ecologically important to maintain life<br />

on Earth and consequently in the soil, since this is a part of the whole.<br />

Many steps of nutrient cycling are made exclusively by microorganism<br />

populations, and some of them may participate in one or more biogeochemical<br />

cycles. The understanding of these interactions between different populations<br />

according to specific phenotypes could give a better perspective about<br />

the processes that are occurring. A percentage of the microbial community<br />

can be grown in culture medium under laboratory conditions, if cultured<br />

microorganisms are considered as a sample of microbial community in soil<br />

microcosms. Grouping the microbial communities by phenotypes is more<br />

realistic than determining the species that are involved in these process.<br />

Although only a small amount of high quality data can be obtained, it is possible<br />

to monitor the effects of hazardous chemical products, environmental<br />

disturbance, and disturbances in nutrient cycling and soil fertility controlled<br />

by these organisms, and also ecosystem health.<br />

Functionality aspects are more important than biodiversity in natural or<br />

sustainable agriculture systems. Some questions could be raised concerning<br />

biodiversity. The first question that should be asked is: what is more important<br />

to the Earth? The number of species that compose the functional group<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


52<br />

Galdino Andrade<br />

or the transformation power of one group? On the other hand, other questions<br />

could be asked such as: what is the importance of one species inside the<br />

biological dynamic system? What is the capacity of one species to influence<br />

nutrient cycling? What does a species represent within the biological<br />

dynamic? What importance can one species have in nutrient cycling? These<br />

questions could lead us to conclude that we need to review our vision of the<br />

soil microcosm, extend our understanding of the biological processes and<br />

interactions that occur in the soil – <strong>plant</strong> system, assuming that these<br />

processes are a whole, and each functional group is only a small fraction of<br />

the whole. Only in this way can we improve the determination of the environmental<br />

impact of any disturbance effect on the soil microbial community, not<br />

only on one specific group of microorganisms. The several functional groups<br />

which take part in different stages of the carbon, phosphorus, nitrogen and<br />

sulphur biogeochemical cycles should be assessed, looking for a correlation<br />

between them and a response in <strong>plant</strong> growth.<br />

Microflora biodiversity is important for other objectives, such as searching<br />

for specific microbial phenotypes to use in food or the pharmaceutical industry.<br />

Its importance in the environment should also be investigated, since molecular<br />

biology does not permit assessment of the microbial interaction mechanisms<br />

in the soil microcosm.<br />

2 General Aspects of Functional Groups of Soil<br />

Microorganisms<br />

In a soil microbial ecosystem individual cells grow and form populations<br />

(Fig. 1). Metabolically related populations constitute groupings called functional<br />

groups, and sets of functional groups conducting complementary physiological<br />

processes interact to form microbial communities. Microbial communities<br />

then interact with communities of macroorganisms to define the<br />

whole biosphere.<br />

We can define the functional groups of microbial populations that take<br />

part in the same transformation of nutrients in the soil, where the same population<br />

of microorganisms may participate in different steps in different<br />

Individual Population<br />

Fig. 1. The individual cells grow and form populations


Population 1<br />

Population 2<br />

Population 3<br />

Population 4<br />

Population 5<br />

cycles (Fig. 2). An example is the cellulolytic functional group; if the soil suspension<br />

is inoculated in Petri dishes with selective culture media for cellulolytic<br />

microorganisms, where cellulose is the only carbon source, and the<br />

culture is incubated at 28 °C for 3 days, some colonies will form halos around<br />

the colonies after staining with Congo red. If we count the different organisms<br />

by decreasing order of numbers, we can observe colonies forming units<br />

of fungi, actinomycetes and then bacteria. Many species will be observed<br />

within the fungi group, as will also occur with actinomycetes and bacteria<br />

populations.<br />

The number of colony forming units (CFU) and the ratio between colony<br />

size and degradation halo diameter should be considered in an evaluation<br />

study, while assessing the cellulolytic activity. These parameters determine<br />

the community size and/or the activity of the individuals that compose it. The<br />

biodiversity of the fungi, actinomycetes and bacteria that form this functional<br />

group are secondary parameters when assessing the functionality of the biogeochemical<br />

cycle under study.<br />

3 Carbon Cycle Functional Groups<br />

4 Microorganisms on the Rhizosphere Microcosm 53<br />

Celulase<br />

producers<br />

Protease<br />

producers<br />

Functional<br />

group of<br />

celulolytic<br />

Functional<br />

group of<br />

Proteolytic<br />

Fig. 2. Many populations of microorganisms may participate in one or more<br />

biogeochemical cycling<br />

C cycle<br />

N cycle<br />

The largest carbon reservoir is present in the sediments and rocks of the<br />

Earth, but the turnover time is so long that flow from this compartment is relatively<br />

insignificant on a human scale. From the viewpoint of living organisms,<br />

a large amount of organic carbon is found in land <strong>plant</strong>s. This represents<br />

the carbon of forests and grasslands and constitutes the major site of photosynthetic<br />

CO 2 fixation. However, more carbon is present in dead organic<br />

material, called humus, than in living organisms (Madigan et al. 2000)<br />

Plant residues are the largest fraction of all organic carbon entering the<br />

soil. Plants contain 15–60 % cellulose, 10–30 % hemicellulose, 2–30 % lignin,


54<br />

Galdino Andrade<br />

and 2–15 % protein. Soluble substances, such as sugars, amino sugars, organic<br />

acids, and amino acids, can constitute 10 % of the dry weight (Paul and Clark<br />

1989). Soil microbes use residue components as substrates for energy and also<br />

as carbon sources in the synthesis of new cells. The presence or absence of<br />

substrates can increase or decrease the populations.<br />

Microorganism populations capable of cellulose, starch and both animal<br />

and <strong>plant</strong> protein hydrolisation can be assessed in the carbon cycle. These<br />

polymers are broken into smaller units of sugars and amino acids, respectively<br />

(Fig. 3).<br />

The functional group of cellulotic microorganisms is formed by fungi,<br />

actinomycetes and bacteria. These microorganisms can produce exoenzymes<br />

called cellulases. The term cellulase describes a diversity of enzyme complexes<br />

that act in two distinct stages. First, there is a loss of the crystalline<br />

Photosynthesis<br />

POLYMERS<br />

Celulose, Starch, Protein<br />

Hidrolytic<br />

Activity<br />

UNITS<br />

Sugar, Amino acids<br />

Aerobic<br />

Microorganisms<br />

Plant<br />

Biosynthesis<br />

Sun Light<br />

CO2<br />

CO2 + H2O<br />

Energy<br />

Biomass<br />

Fig. 3. The activity of some functional groups of<br />

microorganisms in the carbon cycle


4 Microorganisms on the Rhizosphere Microcosm 55<br />

structure, and then the depolymerisation itself occurs. The resultant disaccharide,<br />

cellobiose, is hydrolysed by the enzyme cellobiase to glucose (Paul<br />

and Clark 1989).<br />

The amylolytic group hydrolyses starch, which is a common reserve of<br />

polysaccharide that serves as an energy storage product in <strong>plant</strong>s. Starch is<br />

called amylose when it is a linear polymer of glucose linked in the a1-4 position.<br />

The a1-4 linkage facilitates a more rapid breakdown rate than the b1-4<br />

linkage found in cellulose. Glucose can also be found linked in a1-6 positions<br />

to produce a polymer known as amylopectin.<br />

Extracellular enzymes known as amylases are produced by numerous<br />

fungi, actinomycetes and some bacteria. a-amylases hydrolyse both amylose<br />

and amylopectin to units consisting of several glucose molecules. b-amylase<br />

reduces amylose to maltose (two glucose units), subsequent hydrolysis of maltose<br />

by an a1-4 glucosidase (maltase) yields glucose, and amylopectin is broken<br />

down to a mix of maltose and dextrins.<br />

The proteolytic functional group can act in both the carbon and nitrogen<br />

cycles, described later. Many microorganisms, such as fungi, actinomycetes<br />

and bacteria may produce extra-cell enzymes called proteinases and peptidases.<br />

The proteinases degrade proteins releasing peptides which in turn are<br />

attacked by the peptidases releasing amino acids which are transported inside<br />

the cells (Fig. 3).<br />

The amino acids may be used as a source of either carbon or nitrogen. In<br />

the carbon cycle, the amino acids are catabolised into various compounds, as<br />

intermediate metabolites of the glucolytic path or tricarboxylic acid cycle. In<br />

this conversion, the amino acid undergoes a de-amination process where the<br />

amine group is removed and converted into ammonia (NH 3 + ) which may be<br />

excreted by the cells. The carboxylic group can enter in the tricarboxylic acid<br />

cycle or undergo a process of de-carboxylisation (removal of COOH) and dehydrogenisation,<br />

releasing carbon dioxide and nitrogen compounds, such as<br />

amines and di-amines.<br />

4 Functional Groups of Microrganisms of the Nitrogen Cycle<br />

Plants, animals, and most microorganisms require combined forms of nitrogen<br />

for incorporation into cellular biomass, but the ability to fix atmospheric<br />

nitrogen is restricted to a limited number of bacteria and symbiotic associations.Whereas<br />

many habitats depend on <strong>plant</strong>s for a supply of organic carbon<br />

that can be used as a source of energy, all organisms depend on the bacterial<br />

fixation of atmospheric nitrogen (Atlas and Bartha 1993).<br />

Several functional groups in the nitrogen cycle can be used as bioindicators<br />

of disturbances in the soil. Among these, the groups to be considered are the<br />

symbiotic or free-living nitrogen fixers for legumes and non-legumes <strong>plant</strong>s,<br />

respectively, and others which participate in the mineralisation process of the


56<br />

Galdino Andrade<br />

organic nitrogen in the soil such as free-living ammonifiers and protozoans,<br />

which also have an important function of mobilisation and mineralisation of<br />

nitrogen compounds (Fig. 4). The choice of these groups within the nitrogen<br />

cycle was based on their ability to produce ammonia as an end product. Both<br />

the nitrogen fixers and the ammonifiers such as the protozoa release ammonia<br />

into the rhizosphere. However, the pathway production is different: (1) the<br />

first group uses atmospheric nitrogen that by biological fixation produces<br />

ammonia, (2) the second group takes part in the mineralisation process of<br />

nitrogen organic compounds and, (3) the third group, such as microorganism<br />

predators, obtain proteins from their prey and excrete ammonia, among other<br />

substances.<br />

Atmospheric nitrogen fixation is a fundamental process for the maintenance<br />

of the biosphere, as all organisms require proteins. Nitrogenase is an<br />

enzyme complex which is responsible for nitrogen fixation and requires great<br />

quantities of energy for its activity. Non-symbiotic biological fixation of<br />

nitrogen is carried out by some free-living bacteria genera which are associ-<br />

Protozoans<br />

Feeding<br />

Bacteria<br />

Excretion<br />

Microbiota<br />

Proteins<br />

Aminoacids<br />

Excretion<br />

Celular death<br />

Proteases<br />

Peptidases<br />

Microbial<br />

Mineralization<br />

NH4 + N2<br />

Nitrogen<br />

fixation<br />

Fig. 4. The activity of some functional groups of microorganisms in the nitrogen cycle


4 Microorganisms on the Rhizosphere Microcosm 57<br />

ated with the <strong>plant</strong> rhizosphere. The symbiotic association of microorganisms<br />

and legumes is the most effective in terms of the quantity of nitrogen<br />

fixed. The quantity of nitrogen fixed per year by these microorganism groups<br />

is much greater than free-living fixing.<br />

The mineralisation of nitrogen compounds in the soil (ammoniation and<br />

nitrification) is an essentially microbiological process. The two phases are<br />

equally important because the <strong>plant</strong>s are capable of absorbing the nitrogen in<br />

the two forms (NH 3 + and NO3 – ). When there is no addition of nitrogen fertilisers,<br />

as in the case of natural areas, nitrification depends on the ammoniation<br />

rate for the supply of NH 3 + substrate.<br />

Ammoniation which occurs in the de-amination process of nitrogen<br />

organic compounds is carried out by a large variety of heterotrophic microorganisms<br />

that can use amino acids as a source of nitrogen and carbon.<br />

The protozoans are composed of the three groups flagellates, amoebae and<br />

ciliates and are important in maintaining <strong>plant</strong>-available nitrogen and the<br />

mineralisation process. The role of protozoa in the soils is still unclear, but<br />

evidence for their central position is now accumulating. Protozoans can consume<br />

150–900 g of bacteria m –2 year –1 , which is equal to a production of 15–85<br />

times standing crop (Stout and Heal 1967). This means that preying on bacteria<br />

is an important mechanism in nutrient uptake, resulting in greater mineralisation<br />

and higher nitrogen release by <strong>plant</strong>s (Juma 1993; Fig. 4).<br />

The correlation among these functional groups is obvious and very important<br />

in maintaining the nitrogen cycle and soil fertility. Any factor which<br />

alters the populations of these groups will have an immediate response in<br />

<strong>plant</strong> growth.<br />

5 Functional Groups of Microrganisms of the Sulphur Cycle<br />

Plants, algae, and many heterotrophic microorganisms assimilate sulphur in<br />

the form of sulphate. For incorporation into amino acids biosynthesis as cysteine,<br />

methionine and coenzymes in the form of sulphydril (S-H) groups, sulphate<br />

needs to be reduced to the sulphide level by assimilatory sulphate<br />

reduction.<br />

The stages assessed in the sulphur cycle involve the organic sulphur mineralisers<br />

and the sulphate reducers. These two functional groups participate at<br />

different stages of the sulphur cycle and have hydrogen sulphide (H 2S) formation<br />

as an end product. Hydrogen sulphide, which is volatile, may decrease the<br />

sulphur concentration if it does not complex with other compounds in the soil<br />

(Fig. 5).<br />

Mineralisation of organic sulphur in soil is greatly mediated by microbial<br />

activity. Carbon-linked sulphur is mineralised either though oxidative (aerobic)<br />

decomposition or a desulphirisation (anaerobic) process. The mineralisation<br />

process may be direct (cell-mediated), involving enzymes such as sul-


58<br />

SO4<br />

Galdino Andrade<br />

Assimilatory<br />

sulfate<br />

Reduction<br />

Dissimilatory<br />

Sulfate<br />

reduction<br />

Living organisms<br />

Proteins<br />

Sulphur<br />

aminoacids<br />

H2S<br />

Excretion<br />

Celular death<br />

Proteases<br />

Peptidases<br />

Dissimilatory<br />

Sulfate<br />

Reduction<br />

Fig. 5. The activity of some functional<br />

groups of microorganisms in the sulphur<br />

cycle<br />

phatases where elements such as nitrogen and sulphur-linked carbon mineralised<br />

by microorganisms oxidize the organic carbon compounds to obtain<br />

energy. The heterotrophic soil microorganisms decompose organic sulphur to<br />

form sulphide. In the case of indirect mineralisation, those elements that exist<br />

as sulphate esters are hydrolysed by endo or exoenzymes. This process occurs<br />

by positive feedback or negative control (Sylvia et al. 1998).<br />

The activity of these microorganisms may be aerobic or anaerobic. Anaerobic<br />

microorganisms exist in fairly low numbers in the rhizosphere of <strong>plant</strong>s<br />

which live in non-flooded soils. Bearing in mind that sulphate is fundamental<br />

for <strong>plant</strong> metabolism and that the turnover of organic to inorganic sulphate<br />

implies availability of the nutrient for <strong>plant</strong> growth, the study of these populations<br />

may complement the analysis of functional microorganism groups as<br />

indicators of environmental impact or of biotic fertility indexes in sustainable<br />

agricultural systems or natural areas.


6 Functional Groups of Microrganisms<br />

of the Phosphorus Cycle<br />

4 Microorganisms on the Rhizosphere Microcosm 59<br />

The main functional groups of the phosphorus cycle are the mycorrhizal<br />

fungi and the inorganic phosphate solubiliser microorganisms. The interaction<br />

between these two microbial groups is fundamental for the nutrition of<br />

the majority of native <strong>plant</strong>s and is also of agronomic interest.<br />

The phosphate solubiliser functional group can include fungi, actinomycetes<br />

and bacteria that are capable of solubilizing inorganic phosphate by<br />

production and excretion of organic and inorganic acids, of a phosphatase<br />

group of enzymes and of carbon dioxide (CO 2) in the rhizosphere soil solution.<br />

Carbon dioxide can cause the solubilisation of calcium, magnesium and<br />

Heterotrophic<br />

Microorganisms<br />

Insoluble<br />

Inorganic<br />

Phosphate<br />

Nitrifying<br />

Bacteria<br />

CO2<br />

Organic<br />

Acids Soluble<br />

Inorganic<br />

Phosphate<br />

Mycorrhiza<br />

Fungi<br />

Plant<br />

Root<br />

Nitric Acid<br />

Sulfur Oxiding<br />

Sulfur Reducing<br />

H2SO4<br />

H2S<br />

Fig. 6. The activity of some functional groups of microorganisms in the phosphorus<br />

cycle


60<br />

Galdino Andrade<br />

phosphate compounds. The nitrifying, sulphur oxidants and sulphur- reducing<br />

bacteria can also solubilise insoluble phosphate salts and produce H 2S<br />

under anaerobic conditions. Many microorganisms and <strong>plant</strong>s can produce<br />

organic acids by acting as solubilizing agents and quelants and releasing<br />

orthophosphate in the soil solution (Sylvia et al. 1998). Soluble phosphate in<br />

the soil solution can be absorbed and transported to the <strong>plant</strong> by arbuscular<br />

mycorrhizal (AM) fungus mycelia. The interaction between the phosphate<br />

solubilisers and the mycorrhizae can stimulate mycorrhizal colonisation<br />

and/or <strong>plant</strong> growth by increasing the phosphorus levels (Fig. 6).<br />

The arbuscular mycorrhizal fungi are symbiotic fungi of <strong>plant</strong> roots. This<br />

symbiosis is present in almost all <strong>plant</strong>s in the most different ecosystems<br />

(Hayman 1982). The symbiotic relationship between <strong>plant</strong> roots and mycorrhizal<br />

fungi improves <strong>plant</strong> mineral nutrient acquisition from the soil, especially<br />

immobile elements such as P, Zn and Cu, but also more mobile ions such<br />

as S, Ca, K, Fe, Mg, Mn, Cl, Br and N (Tinker 1984). In soils where such elements<br />

may be deficient or less available, mycorrhizal fungi increase efficiency<br />

of mineral uptake, resulting in increased <strong>plant</strong> growth (Linderman 1988).<br />

The mycorrhizal complex (AM fungi and root) changes the nutritional and<br />

physicochemical conditions of the rhizosphere, and has a large negative or<br />

positive impact on the functional microorganism groups. This effect depends<br />

on the cycle to which the functional group belongs. However, in spite of the<br />

importance of the mycorrhizae, these groups should not be assessed in isolation.<br />

7 Dynamics of the Rhizosphere Functional Groups<br />

of Microrganisms<br />

The interaction of specific biological systems, in a ecosystem or microcosm,<br />

depends on the interplay of three general factors – environment, biological<br />

community structure (biodiversity), and biological activity (function). The<br />

role of diversity, particularly of microorganisms, and the relationship<br />

between microbial diversity and function is largely unknown (Griffiths et al.<br />

1997). As can be seen, each functional group can interact with different biogeochemical<br />

soil cycles and the environmental impact caused by an agent can<br />

be determined by the changes observed in the populations, as a determined<br />

environmental condition can affect the microbial activity without affecting<br />

the community biodiversity (Griffiths et al. 1997). The dynamic behaviour of<br />

perturbed communities is a branch of general ecology closely related to the<br />

study of natural and artificial disturbances in microbial habitats. Another<br />

important factor is the relationship between resistance and resilience, whose<br />

combined effects determine the ecosystem stability. Resistance is the inherent<br />

capacity of the system to hold disturbance, whereas resilience is the capacity<br />

to recover after disturbance.


4 Microorganisms on the Rhizosphere Microcosm 61<br />

8 Relationship Among r and k Strategist Functional Groups<br />

The determination of the r and k strategists (Andrews 1984) is also related to<br />

soil disturbance, resilience and health. The r strategist microorganism has a<br />

high reproductive rate with few competitive adaptations. On the other hand,<br />

the k strategist microorganism reproduces more slowly than the r strategist,<br />

and is usually a more stable and permanent member of the community.<br />

Fungi and actinomycetes are normally k strategists and are involved in the<br />

carbon cycle degrading cellulose and structural proteins among other macromolecules.<br />

The stability of these compounds and the slow k strategist growth<br />

rate render them not very sensitive to swift environmental changes. On the<br />

other hand, the r strategists such as bacteria are more sensitive to quick environmental<br />

changes. Heterotrophic bacteria populations are affected by the<br />

lack of carbohydrates which occurs due to changes in the rhizosphere carbon<br />

flow in the photosynthesis function variations between day and night. Only<br />

heterotrophic bacteria populations that have metabolic diversity and can<br />

manage to use other compounds, such as amino acids for obtaining carbon<br />

and energy, will keep their numbers in the rhizosphere. The other populations<br />

decrease their CFU number. Plants begin photosynthesis at daybreak with a<br />

consequent increase in carbon concentration in the exudates, and the heterotrophic<br />

bacteria community returns to its previous composition.<br />

9 Arbuscular Mycorrhizal Fungi Dynamics in the<br />

Rhizosphere<br />

The MA can also be considered k strategists and influence several biogeochemical<br />

soil cycles: (1) the carbon cycle due to alterations in the flow of carbon<br />

compounds from the exudates, (2) the phosphorus cycle due to stimulus<br />

to phosphate-solubilising bacteria activity and absorption of soluble phosphorus<br />

by <strong>plant</strong>s (Toro 1998), (3) the nitrogen cycle due to stimulus to symbiotic<br />

(Toro 1998) and non-symbiotic fixation (Vosátka and Gryndler 1999) and<br />

to the rhizosphere ammoniation process (Amora-Lazcano et al. 1998). The<br />

sulphur cycle is also influenced by alterations in the autotrophic sulphur oxidising<br />

and sulphate reducing bacteria populations (Amora-Alzcano and Azón<br />

1997).<br />

The term mycorrhizosphere (Oswald and Ferchau 1968) refers to the zone<br />

of influence of the mycorrhiza (fungus-root) in the soil. The mycorrhizosphere<br />

has two components. One is the rhizosphere, a thin layer of soil that surrounds<br />

the root and is under the joint, direct influence of the root, root hairs,<br />

and AM hyphae adjacent to the root. The other, the hyphosphere, is not<br />

directly influenced by the root. The hyphosphere is a zone of AM hypha-soil<br />

interactions (Marschner 1995), and may be more or less densely permeated by<br />

the AM soil mycelium.


62<br />

Galdino Andrade<br />

In our laboratory, hypha colonisation of some MA fungus species by bacteria<br />

in spores germinated in 1 % agar-water medium on a Petri dish was<br />

observed. These bacteria had as their single nutrient source the products<br />

excreted by the MA mycelia in the medium (Fig. 7). The bacteria formed a<br />

dense cell layer around the hypha in an experiment with Glomus etunicatum.<br />

From this layer, as the exudate excretion increased the medium nutrient to<br />

optimum levels, the bacteria developed and colonised the remaining mycelia<br />

(Fig. 8).<br />

In an axenic conditions experiment with maize <strong>plant</strong>s and colonised Glomus<br />

clarum mycelia, the bacteria which colonised the G. clarum mycelia without<br />

<strong>plant</strong>s continued to prefer products excreted by the mycelia, and no<br />

colonies were observed in the <strong>plant</strong> roots (Fig. 9). These results seem to indicate<br />

that the fungus mycelia produce some growth factor essential for the bacteria<br />

growth, which is not found in the maize root exudates. However, the<br />

mechanisms involved in this interaction are not yet known.<br />

H<br />

A B<br />

Fig. 7. Bacterial growth around arbuscular mycorrhiza hyphae in water-agar 1 %. BC<br />

Bacteria colonies , H hyphae. A Scutellospora heterogama (x40), B corresponds to black<br />

box indicated in A (x100) C Glomus clarum (x100)<br />

BC<br />

C<br />

BC<br />

H


4 Microorganisms on the Rhizosphere Microcosm 63<br />

Fig. 8. Bacterial colonising AM hyphae of Glomus etunicatum in 1 % water-agar. BC Bacteria<br />

colonies, H hyphae. A General aspects of mycelia colonised by bacteria (x20), B corresponds<br />

to black box indicated in the A, where bacteria is growing around hyphae<br />

(x100), C bacteria growing around hyphae (x400)<br />

In the soil, Andrade et al. (1997) observed sorghum <strong>plant</strong>s inoculated with<br />

several exotic or native Glomus species either exotic or native to the test soil.<br />

The soils adhering to the root were considered rhizosphere or not adhering to<br />

the root were considered hyphosphere. Bacterial numbers were greater in<br />

rhizo- than in hyphosphere soil. Isolates of Bacillus and Arthrobacter were<br />

most frequent in hyphosphere and Pseudomonas in rhizosphere soils. More<br />

bacterial species were found in hyphosphere than in rhizosphere soil, and<br />

bacterial communities varied within and among AM treatments. The development<br />

of the AM mycelium in soil had little influence on the composition of<br />

the microflora in the hyphosphere, while AM root colonisation was positively<br />

related with bacterial numbers in the hyphosphere and with the presence of<br />

Pseudomonas in the rhizosphere.<br />

In another experiment, Andrade et al. (1998) inoculated Alcaligenes eutrophus<br />

and Arthrobacter globiformis in sorghum <strong>plant</strong>s. The first is an isolate<br />

of the Glomus mosseae hyphosphere and the second an isolate of the G.<br />

mosseae and G. intraradices mycorrhizosphere. Ten days after inoculation,


64<br />

Galdino Andrade<br />

Fig. 9. Bacteria colonising mycelia of Glomus clarum in the hyphosphere of maize <strong>plant</strong>s<br />

grown under axenic conditions in 1 % water-agar. Bacteria did not colonise maize roots,<br />

colonies were observed only around mycelia (x40). BC Bacteria colonies, H hyphae, R<br />

root<br />

the A. globiformis population present in bulk soil, in the rhizosphere and<br />

hyphosphere were similar, but that present in the mycorrhizosphere was<br />

larger. A. eutrphus was dependent on the presence of G. mosseae in the soil,<br />

indicating that even in soil some bacteria may depend on MA-excreted<br />

metabolic products.<br />

These results show that the MA-<strong>plant</strong> system is very complex and the influence<br />

of these microorganisms is fundamental for the regulation of the biogeochemical<br />

cycles in the rhizosphere system. On the other hand, the<br />

microorganisms of other cycles also influenced the mycorrhizal activity and<br />

root infection with direct consequences on the <strong>plant</strong> growth and soil fertility.<br />

In degraded areas of tropical regions, the soil is compacted displaying minimum<br />

aeration and draining capacity, aluminium and manganese toxicity<br />

and low fertility indices especially for nitrogen, phosphorus and organic matter.<br />

In these areas, the re-vegetation process is directly related to the interaction<br />

between the <strong>plant</strong> roots and the functional microorganism groups. The<br />

pioneer <strong>plant</strong>s are the first to colonise these low fertility areas, and they are<br />

very dependent on AM for phosphorus. The pioneer <strong>plant</strong>s in this process are<br />

r strategists which improve the physicochemical characteristics of the soil<br />

and fertility levels with time, allowing other groups of more demanding


4 Microorganisms on the Rhizosphere Microcosm 65<br />

<strong>plant</strong>s (k strategists) to establish in the area and to form a forest in equilibrium.<br />

The pioneer <strong>plant</strong>s can survive under adverse conditions due to the presence<br />

in the rhizosphere of microorganisms which supply nutrients for their<br />

metabolism, and in turn, their exudates maintain these rhizosphere microorganisms.<br />

The k strategist mycorrhizae are sufficiently stable to maintain the<br />

required nutrient levels for this <strong>plant</strong> group. In this sense, groups of r strategist<br />

microorganisms succeed each other, maintaining the dynamic of the system<br />

and the reconstitution of other biogeochemical cycles until the system<br />

equilibrium is reached with the establishment of late secondary and climax<br />

<strong>plant</strong> groups.<br />

10 Dynamics Among the Functional Microrganism Groups<br />

of the Carbon, Nitrogen, Phosphorus and Sulphur Cycles<br />

There are several stages in each biogeochemical cycle, and many microorganisms<br />

can take part in one or more cycles depending on the diversity of their<br />

metabolic path (Fig. 10). Microbiota metabolic versatility makes a single bacteria<br />

species able to use various carbohydrates, such as glucose, fructose and<br />

saccharose, as a carbon and energy source, and in their absence they can use<br />

amino acids or other compounds.<br />

The biosphere is composed of all living organisms which depend on matter<br />

transformation for their maintenance. The functional microorganism groups<br />

are inserted in this system which transforms matter and maintains the levels<br />

of nutrients available on Earth. Due to their functional importance, they can<br />

be used as biological indicators to determine any natural or artificial impact<br />

which may occur in the soil. It is obvious that the complexity of the biological<br />

interactions occurring on the soil–<strong>plant</strong> interface must be simplified to allow<br />

quick and accurate assessment of these microorganism populations. Thus,<br />

only those stages of the biogeochemical cycles which directly influence <strong>plant</strong><br />

growth should be chosen. However, different stages can be selected according<br />

to the experimental objective.<br />

Autotrophic organisms have the important function of matter de-mineralisation<br />

and transform it into organic molecules. In this group are <strong>plant</strong>s that<br />

de-mineralise carbon, i.e. transform carbon dioxide (CO 2) into glucose, which<br />

is then polymerised mainly into starch, cellulose, hemicellulose and lignin.<br />

Plants are also responsible for transforming NO 3 – ,NH3 + ,and SO4 2– into amino<br />

acids, PO 4 2– into nucleic acids while ATP, NADP, and SO4 2– can be transformed<br />

into glutathione.<br />

In a simplified way, <strong>plant</strong>s can be considered as nutrients from the soil<br />

solution plus solar energy accumulated in chemical form. Plants generally<br />

release organic molecules into the soil in two ways: (1) by depositing dead<br />

<strong>plant</strong> material to form the litter; and, (2) by exuding excretion and lysates into


66<br />

Galdino Andrade<br />

Plants<br />

Carbon<br />

Desmineralization<br />

Starch<br />

Celulose<br />

Lignin<br />

Hidrolytic<br />

Activity<br />

Sugars<br />

Carbon<br />

Source<br />

Heterotrophic<br />

Microorganisms<br />

CO2<br />

Organic Acids<br />

Soluble<br />

Inorganic<br />

Phosphate<br />

Carbon Source<br />

N<br />

Desmineralization<br />

Carbon Source<br />

Sulfur<br />

Source<br />

Carbon<br />

Nitrogen<br />

Source<br />

SO4 -2<br />

BIOSPHERE<br />

H2SO4<br />

Insoluble<br />

Inorganic<br />

Phosphate<br />

Phosphatases<br />

Proteins<br />

Mycorrhiza<br />

Fungi<br />

Protozoans<br />

Proteases<br />

Peptidases<br />

Microbial<br />

mineralization<br />

H2S<br />

Excretion<br />

Phosphate<br />

Desmineralization<br />

NH3 +<br />

Plant<br />

Root<br />

NO3 -<br />

Nutrient Uptake<br />

N2<br />

Nitrogen<br />

fixation<br />

Aminoacids<br />

Nitrogen<br />

Desmineralization<br />

Sulfur<br />

mineralization<br />

Nitrification<br />

Sulfate<br />

Reduction<br />

Nutrient Uptake<br />

Organic<br />

Phosphate<br />

Feeding<br />

Bacteria<br />

Organic Acids<br />

Nitric Acid<br />

Nutrient Uptake<br />

Fig. 10. The interaction among functional groups of microorganisms in the carbon,<br />

nitrogen, phosphorus and sulphur cycles


4 Microorganisms on the Rhizosphere Microcosm 67<br />

the rhizosphere, a phenomenon known as rhizodeposition. These compounds,<br />

which are continuously released into the soil, constitute the main<br />

nutrient sources, maintain the microbiota, the fertility and participate in the<br />

maintenance of the soil structure.<br />

Microorganisms are classified into several categories according to the carbon<br />

and energy source used, but only some groups will be considered in this<br />

chapter. The heterotrophic microorganisms can use glucose or amino acids as<br />

carbon sources. Glucose can be obtained from some macromolecules, such as<br />

cellulose and starch, which undergo lytic action by enzymes produced by the<br />

cellulose and starch-reducing microorganisms (Fig. 10).<br />

Proteins are degraded to amino acids by proteolytic organisms, which can<br />

use these compounds as carbon or nitrogen sources. On the other hand, sulphur<br />

amino acids such as cystine and cysteine can also be used to obtain sulphur<br />

which is used in the biosynthesis of other compounds necessary for cell<br />

metabolism. The amino acids can also be used by the cell without lysis of the<br />

molecule, as many microorganism species are not able to biosynthesise all the<br />

amino acids required by the cell.<br />

Protozoa, such as amoebas, ciliates and flagellates, are organisms which<br />

have the function of immobilising and mineralising the nitrogen in the rhizosphere<br />

system. Bacteria are their main nutrient source, and they obtain<br />

nitrogen and other nutrients for their metabolism from them. Some of these<br />

nitrogen compounds are released into the soil as inorganic NH 3 + and can be<br />

absorbed by the root or by other microorganism groups such as nitrifiers, sulphate<br />

reducers or oxidisers or phosphate solubilisers. Biological nitrogen fixation<br />

is very important in the introduction of NH 3 + molecules into the rhizosphere<br />

(free-living N fixers) or in the <strong>plant</strong> (symbiotic N fixers). These fixed<br />

molecules can be transformed in NO 3 – or used in the biosynthesis of amino<br />

acids that will form the cell proteins when polymerised. Sulphur amino acids<br />

may be synthesised from SO 4 2– obtained by the oxidation of S by sulphur cycle<br />

bacteria. NO 3 – and NH3 + can be used in amino acid biosynthesis and also as<br />

final receptors of electrons for some groups of facultative anaerobic bacteria.<br />

Phosphate exists in the soil mainly in the soluble inorganic form. Several<br />

solubilisation mechanisms have been described and many microorganisms<br />

produce compounds which can solubilise phosphates. The nitrogen cycle<br />

functional group, the nitrifiers, produces NO 3 – that can form nitric acid. The<br />

sulphur cycle functional group can produce SO 4 2– that can form H2SO 4 or<br />

reduce it to H 2S, which will also solubilise insoluble inorganic phosphate. In<br />

the degradation of sulphur amino acids, proteolytic microorganisms release<br />

H 2S or CO 2, which can form carbonic acid. Both molecules can also solubilise<br />

inorganic phosphate. The carbon cycle microorganisms form CO 2 and<br />

organic acids as end products of their catabolism, and both compounds are<br />

responsible for pH reduction and inorganic phosphate solubilisation.<br />

Soluble inorganic phosphate is absorbed mainly by the mycorrhizal fungi<br />

that transport these molecules to the <strong>plant</strong>, which in turn transform them into


68<br />

Galdino Andrade<br />

organic phosphate. This phosphate is deposited in the soil by rhizodeposition<br />

or absorbed in the organic form by heterotrophic microorganisms which take<br />

part in the nitrogen, carbon or sulphur cycles (Fig. 10).<br />

References and Selected Reading<br />

Amora-Lazcano E, Azcón R (1997) Response of sulfur cycling microorganisms to arbuscular<br />

mycorrhizal fungi in the rhizosphere of maize. Appl Soil Ecol 6:217–222<br />

Amora-Lazcano E, Vázquez MM, Azcón R (1998) Response of nitrogen-transforming<br />

microorganisms to arbuscular mycorrhiza fungi. Biol Fertil Soils. 27:65–70<br />

Andrade G, Linderman RG, Bethlenfalvay GJ (1998) Bacterial associations with the mycorrhizosphere<br />

and hyphosphere of the arbuscular mycorrhizal fungus Glomus<br />

mosseae. Plant Soil 202:79–87<br />

Andrade G, Mihara KL, Linderman RG, Bethlenfalvay GJ (1997) Bacteria from rhizosphere<br />

and hyphosphere soils of different arbuscular mycorrhizal fungi. Plant Soil<br />

192:71–79<br />

Andrews JH (1984) Relevance of r and k theory to the ecology of <strong>plant</strong> pathogens. In:<br />

Klug MJ, Reddy CA (eds) Current perspectives in microbial ecology. American Society<br />

for Microbiology, Washington, pp 1–7<br />

Atlas RM, Bartha R (eds) (1993) Microbial ecology: fundamentals and applications, 3rd<br />

edn. The Benjamin/Cummings Publishing Company, California, 563 pp<br />

Brussaard L (1998) Soil fauna, guilds, functional groups and ecosystem processes. Appl<br />

Soil Ecol 9:123–135<br />

Griffiths BS, Ritz K, Wheatley RE (1997) In: Insan H, Ranger A (eds) Microbial communities:<br />

functional versus structural approaches. Springer, Berlin Heidelberg New York,<br />

pp 1–10<br />

Hayman DS (1982) Influence of soils and fertility on activity and survival of vesiculararbuscular<br />

mycorrhizal fungi. Phytopathology 72:1119–1125<br />

Juma NG (1993) Interactions between soil structure/texture, soil biota/soil organic matter<br />

and crop production. Geoderma 57:3–30<br />

Linderman RG (1988) Mycorrhizal interactions with the rhizosphere microflora. The<br />

mycorrhizosphere effect. Phytopathology 78:366–371<br />

Madigan TM, Martinko JM, Parker J (eds) (2000) Microbial ecology. In: Brock biology of<br />

microorganisms, 9th edn, Prentice Hall, New Jersey, pp 642–719<br />

Marschner H (ed) (1995) The soil–root interface (rhizosphere) in relation to mineral<br />

nutrition. Mineral nutrition of higher <strong>plant</strong>s, 2nd edn. Academic Press, London, pp<br />

537–595<br />

Oswald ET, Ferchau HA (1968) Bacterial associations of coniferous mycorrhizae. Plant<br />

Soil 28:187–192<br />

Paul EA, Clark FE (eds) (1989) Carbon cycling and soil organic mater. In: Soil <strong>microbiology</strong><br />

and biochemistry. Academic Press, San Diego, pp 93–116<br />

Stout JD, Heal OW (1967) Protozoa. In: Burgues A, Raw F (eds) Soil biology. Academic<br />

Press, New York, pp 149–195<br />

Sylvia DM, Fuhrman JJ, Hartel PG, Zuberer DA (eds) (1998) Principles and applications<br />

of soil <strong>microbiology</strong>. Prentice Hall, Englewood Cliffs, pp 346–367<br />

Tinker PB (1984) The role of microorganisms in mediating and facilitating the uptake of<br />

<strong>plant</strong> nutrients from soil. Plant Soil 76:77–91<br />

Toro M,Azcón R, Barea JM (1998) The use of isotopic dilution techniques to evaluate the<br />

interactive effects of Rhizobium genotype, mycorrhizal fungi, phosphate-solubilizing


4 Microorganisms on the Rhizosphere Microcosm 69<br />

rhizobacteria and rock phosphate on nitrogen and phosphorus acquisition by Medicago<br />

sativa. New Phytol 138:265–273<br />

Vosátka M, Gryndler M (1999) Treatment with culture fractions from Pseudomonas<br />

putida modifies the development of Glomus fistolosum mycorrhiza and response of<br />

potato and maize <strong>plant</strong>s to inoculation. Appl Soil Ecol 11:245–251


5 Diversity and Functions of Soil Microflora in<br />

Development of Plants<br />

Ramesh Chander Kuhad, David Manohar Kothamasi,<br />

K. K. Tripathi and Ajay Singh<br />

1 Introduction<br />

Soil is a dynamic and complex system consisting of living organisms interacting<br />

with inorganic mineral particles and organic matter. A wide range of<br />

functions is performed by soil that directly or indirectly sustains the world’s<br />

human population. Soil plays a vital role in food production, as a reservoir<br />

for water and filter for pollutants. Soils store almost twice as much carbon<br />

as the atmosphere does and are important links in the natural cycle that<br />

determines atmospheric carbon dioxide level (O’Donnell and Görres 1999).<br />

Soils sustain an immense diversity of microbes, which exceeds that of<br />

eukaryotic organisms (Torsvik and Øvreås 2002). Microorganisms exist in<br />

every conceivable place on earth, even in extreme environments. One gram<br />

of soil may harbor up to 10 billion microorganisms of possibly thousands of<br />

different species.<br />

It is widely accepted that the extent of microbial diversity has not been adequately<br />

explored. Some bacteriologists believe that about 100,000 to 1 billion<br />

bacterial species actually exist in the earth environment and only about 4000<br />

species have been described (Staley 1997). Mycologists estimate that there are<br />

more than 1.5 million species of fungi of which only 72,000 species have been<br />

isolated or described (Hawksworth 1997). Microorganisms can exist either in<br />

an active or in a dormant yet persistent form. The ratio of viable counts to<br />

direct counts reflects the ratio between the numbers of the active (dividing)<br />

cells and the quiescent cells, and most bacteria in soil are in the latter form<br />

(Hattori et al. 1997). The tropics are considered to be richer in microbial<br />

diversity than boreal or temperate environments (Hunter-Cevera 1998). Some<br />

microbiologists believe that there is an equal amount of microbial diversity in<br />

the deserts.Actinomycetes with motile spores appear to be widely distributed<br />

in littoral zones and arid environments.<br />

Analysis of microbial functional diversity is important when considering<br />

the ability of the ecosystem to respond to changing environmental conditions,<br />

links between ecosystem processes and functional diversity and the<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


72<br />

Ramesh Chander Kuhad et al.<br />

need to conserve the microbial gene pool (Prosser 2002). Fortunately, with the<br />

development of advanced molecular in situ methods and improved cultivation<br />

procedures, more accurate estimates of the microbial functional diversity<br />

on earth can be predicted and their role in the soil ecosystem can be thoroughly<br />

evaluated. In this chapter, the interaction and functional diversity of<br />

microorganisms in the soil environment related to <strong>plant</strong> growth and development<br />

is discussed.<br />

2 Functional Diversity of Soil Microflora<br />

The microbial functional diversity encompasses a range of activities and has<br />

been assumed to influence ecosystem stability, productivity and resilience<br />

towards stress and disturbances. Typically, microorganisms decrease with<br />

depth in the soil profile, as do the <strong>plant</strong> roots and soil organic matter. Differences<br />

in microbial community structures reflect the ability of microorganisms<br />

to respond to specific environmental controls and substrates (Paul and<br />

Clark 1998). For example, the arbuscular mycorrhizal fungus, Glomus,occurs<br />

worldwide on a variety of agricultural <strong>plant</strong>s. Examination of the crop rotations<br />

shows that strains of this fungus change with the type and nutrition of<br />

the host crop. The fluorescent pseudomonads are attracted to <strong>plant</strong> roots and<br />

show genetic and physiological divergence between soil and <strong>plant</strong> <strong>surface</strong>s.<br />

While Penicillium is abundant in temperate and cold climates, Aspergillus<br />

predominates in warm areas. Cyanobacteria are commonly found in neutral<br />

to alkaline soils, but rarely under acidic conditions. Depending on the preferred<br />

metabolites present in the soil, nitrogen-fixing, sulfur- and hydrogenoxidizing<br />

and nitrifying bacteria are often found in addition to the denitrifiers,<br />

sulfate-reducers and methanogens. Various microbial processes in soil,<br />

which directly or indirectly influence <strong>plant</strong> development, are shown in<br />

Table 1.<br />

Microbiologists are continually learning that microbial function in the<br />

ecosystem is as diverse as the microbes themselves. In studying functional<br />

relationships between agricultural <strong>plant</strong>s and microbes, Shen (1997) reported<br />

that Pseudomonas and Bacillus spp. enable <strong>plant</strong>s to remain healthy and help<br />

improve growth yields. Microbially digested organic waste enhances <strong>plant</strong><br />

growth and improves soil structure and nutrients (Shen 1997). Denitrifying<br />

bacteria can utilize nitrous oxides (NO x) as the terminal electron acceptor.<br />

Many denitrifiers produce NO x reductase and can metabolize NO x in aerobic<br />

and anaerobic conditions (Stepanov and Korpelal 1997).<br />

Soil comprises a variety of microhabitats with different physicochemical<br />

gradients and discontinuous environmental conditions. Microbes adapt to the<br />

microhabitat and live together in consortia with more or less clear boundaries,<br />

interacting with each other and with other parts of the soil biota (Yin et<br />

al. 2000; Tiedje et al. 2001). Competitive interactions are also thought to be a


5 Diversity and Functions of Soil Microflora in Development of Plants 73<br />

Table 1. Major processes of soil microflora influencing <strong>plant</strong> growth<br />

Microbial process Examples of microbes<br />

Organic matter decomposition Trichoderma, Fusarium, Bacillus, Streptomyces,<br />

Clostridium<br />

Symbiotic nitrogen fixation Rhizobium, Bradyrhizobium, Frankia, Anabaena<br />

Nonsymbiotic nitrogen fixation Azotobacter, Beijerinckia, Aerobacter, Chlorobium,<br />

Nostoc<br />

Nitrogen mineralization Bacillus, Pseudomonas, Serratia<br />

Nitrification Nitrobacter, Nitrosomonas<br />

Denitrification Achromobacter, Pseudomonas<br />

Phosphate solubilization Azotobacter, Enterobacter, Bacillus, Aspergillus,<br />

Penicillium, Rhizoctonia, Trichoderma<br />

Sulfur transformation Desulfovibrio, Thiobacillus<br />

Iron transformation Ferribacterium, Leptothrix<br />

Phytohormone production Azotobacter, Azospirillum, Pseudomonas,<br />

Rhizobium, Bacillus, Flavobacterium, Actinomyces,<br />

Nocardia, Fusarium, Gibberella, Aletrnaria,<br />

Penicillium<br />

Siderophore production Neurospora, Trichoderma, Agaricus, Fusarium,<br />

Penicillium, ericoid mycorrhizal fungi, Nocardia,<br />

Pseudomonas, Bacillus, Aeromaonas, Erwinia<br />

Biotic control Pseudomonas, Bacillus, Strepetomyces<br />

key factor controlling microbial community structure and diversity. The<br />

impact of soil structure and spatial isolation on microbial diversity and community<br />

structure has been clearly demonstrated (Staley 1997; Pankhurst et al.<br />

2002). More than 80 % of the bacteria were found located in micropores of stable<br />

soil microaggregates (2–20 mm) in soils subjected to different fertilization<br />

treatments (Ranjard and Richaume 2001). Such microhabitats offer most<br />

favorable conditions for microbial growth with respect to water and substrate<br />

availability, gas diffusion and protection against predation. Soil structure and<br />

water regime influence competitive interactions by causing spatial isolation<br />

within communities. A high diversity in soil with high spatial isolation may<br />

also have been caused by a higher heterogeneity of carbon resources in the<br />

soil. Particle size and other factors like pH and type and amount of available<br />

organic compound may highly impact microbial diversity and community<br />

structure (De Fede et al. 2001). Soil microbes are also subjected to considerable<br />

seasonal fluctuations in environmental conditions such as temperature,<br />

water content, and nutrient availability (Smit et al. 2001).<br />

Catabolic diversity has been used to investigate the effect of stress and the<br />

disturbance on the soil biodiversity. The catabolic response profile (CRP), a


74<br />

Ramesh Chander Kuhad et al.<br />

measure of short-term substrate-induced respiration, has been used to calculate<br />

the diversity and catabolic functions expressed in situ (Degens et al.<br />

2001). When soils from long-term managed environments were subjected to<br />

stress and disturbances, microbial communities with low catabolic evenness<br />

(crop fields) were less resistant to stress and disturbance than were communities<br />

with high catabolic evenness (pasture). After a major disturbance<br />

(landslides, volcanic eruptions, etc.), marked changes in catabolic functional<br />

diversity has been reported in developing soil ecosystems (Schipper et al.<br />

2001).<br />

Most members of the soil biota are organotrophs. The major source of carbon<br />

input for soil organisms are the <strong>plant</strong> roots and organic residues contributed<br />

during and following <strong>plant</strong> growth. The proportion of nitrogen, carbon<br />

and other organic matter changes with both <strong>plant</strong> types and landscape,<br />

which in turn, alter microbial mass, activity and diversity (Paul and Clark<br />

1998).<br />

Microorganisms play an essential role in functioning and sustainability of<br />

all natural ecosystems including biogeochemical cycling of nutrients and<br />

biodegradation. Most soils are exposed to fluctuating environmental conditions<br />

and the high diversity of organic substrate is likely to have a positive<br />

effect on the function. Interactions between different trophic levels were elucidated<br />

in a simple ecosystem model in which primary producers (<strong>plant</strong>s) and<br />

decomposers (microorganisms) were linked through cycling of a limiting<br />

nutrient factor for the primary producers (Loreau 2001). The model predicts<br />

that microbial diversity has a positive effect on nutrient cycling efficiency, and<br />

contributes to increased ecosystem processes. However, in interacting microbial<br />

consortia, a small linear change in diversity may result in nonlinear<br />

changes in the process, therefore relationship between microbial diversity<br />

and soil processes may not necessarily be linear.<br />

Biochemical quality of the substrate and the physical availability of those<br />

components to the degradative microorganisms are key determinants of the<br />

rate of decomposition processes in soils, and reflects a number of interacting<br />

components (Bending et al. 2002). In the case of crop residues, nitrogen content<br />

and structural polymers such as lignin interact to control microbial<br />

nitrogen mineralization-immobilization processes during decomposition.<br />

The types of nutritional substrates available are different in soils with varying<br />

soil organic matter quality, and directly affect the microbial communities<br />

active in the soil. Native soil organic matter content may also significantly<br />

affect the enzyme diversity, which is found greater in high organic matter soil.<br />

Organic acids, such as malate, citrate and oxalate, have also been proposed to<br />

be involved in many rhizospheric processes, including nutrient acquisition,<br />

metal detoxification, alleviation of anaerobic stress in roots, mineral weathering<br />

and pathogen attraction (Jones 1998).<br />

The ecological relevance of the community structure for the function of<br />

systems is the main reason to study the microbial diversity. There is no single


5 Diversity and Functions of Soil Microflora in Development of Plants 75<br />

technique available today that can reveal the entire diversity of a microbial<br />

community. Several approaches are available for assessment of microbial<br />

diversity (Bridge and Spooner 2001; Dahllöf 2002; Prosser 2002). Time-consuming<br />

cultivation-based assessment of microbial diversity has been widely<br />

used (Torsvik et al. 1996).With advanced methods, identification can be accelerated<br />

by automated methods, e.g., Biolog; phospholipid fatty acid (PLFA)<br />

profiling, fatty acid methyl ester profiling (FAME), DNA-hybridization and reassociation.<br />

However, potential limitations of this approach are widely<br />

accepted. Separation of biomass from particulate material varies between<br />

species, and, with growth form (spore, cells, and mycelia), introduces bias. It is<br />

almost impossible to design growth media and cultivation conditions that are<br />

suitable for all members of the microbial community. The approach of identification<br />

using traditional methods, based on phenotypic characteristics, is<br />

also limited for analysis of diversity in complex environments, such as soil<br />

when quantification of the diversity is required.<br />

The importance and need to study the vast biodiversity in different environments<br />

has stimulated the development of molecular methods for cultureindependent<br />

study of microbial communities. These methods have employed<br />

a combination of analysis of genes and microscopy. Analysis of 16S rRNA<br />

genes is now widely used for the analysis of bacterial populations and analysis<br />

of 18S rRNA genes and internal transcribed spacer (ITS) regions are<br />

increasingly used for fungal population analysis (Hunter-Cevera 1998; Bridge<br />

and Spooner 2001; Torsvik and Øvreås 2002). Ribosomal RNA genes are ideal<br />

for this purpose because they possess regions with sequences conserved<br />

between all bacteria or fungi, facilitating alignment of sequences when making<br />

comparisons, while other regions exhibit different degrees of variation,<br />

enabling distinction between different groups. These differences provide the<br />

basis for a phylogenic taxonomy and enable quantification of evolutionary<br />

differences between different groups. Polymerase chain reaction (PCR)-based<br />

fingerprinting techniques provide a rapid analysis of changes in whole community<br />

structure with high resolution. These fingerprinting techniques, such<br />

as denaturind gradient gel electrophoresis (DGGE), amplified rDNA restriction<br />

analysis (ARDRA), terminal restriction fragment length polymorphism<br />

(T-RFLP) and ribosomal intergenic spacer analysis (RISA), provide information<br />

on the species composition, and can be used to compare common species<br />

present in samples. Sequence information can also be used to design and construct<br />

fluorescent-labelled oligonucleotide probes specific for particular<br />

microbial groups using fluorescence in situ hybridization (FISH technique).<br />

For a comprehensive description and discussion on potential and limitations<br />

of various molecular approaches, excellent reviews by Bridge and Spooner<br />

(2001), Kozdroj and van Elsas (2001), Dahllöf (2002), Prosser (2002) and<br />

Torsvik and Øvreås (2002) may be consulted.


76<br />

Ramesh Chander Kuhad et al.<br />

3 Role of Soil Microflora in Plant Development<br />

3.1 Mycorrhiza<br />

Fungi, which form a symbiotic association with <strong>plant</strong> roots, are referred to as<br />

mycorrhizal fungi and the association itself is called as mycorrhiza. There are<br />

five broad groups of mycorrhiza: the ectomycorrhizae, the arbuscular mycorrhizae,<br />

the ericaceous mycorrhizae, the ectendomycorrhizae, and the orchidaceous<br />

mycorrhizae (Bagyaraj and Varma 1995; Hodge 2000). The most common<br />

mycorrhizal association found in cultivated crop <strong>plant</strong>s throughout the<br />

world is the arbuscular mycorrhizal (AM) fungi. Ectomycorrhiza (EM),<br />

formed by fungi belonging to basidiomycetes and ascomycetes, are commonly<br />

associated with temperate trees, whereas ericoid mycorrhiza are found in the<br />

<strong>plant</strong>s from the family Ericaceae and <strong>plant</strong> communities at high latitude and<br />

altitude (Perotto et al. 2002; Koide and Dickie 2002). Orchid mycorrhizae are<br />

associated with orchids. The AM and ectendomycorrhizal fungi are more<br />

prevalent in the tropics and arid/semiarid regions. AM, the most prevalent<br />

<strong>plant</strong>-fungus association, comprise about 150 species, belonging to the order<br />

Glomales of Zygomycotina (Simon 1996; Myrold 2000).<br />

Most angiosperm, gymnosperm, fern and bryophyte families form mycorrhizae.<br />

It is believed that <strong>plant</strong>s growing in aquatic, water logged and saline<br />

habitats usually do not form mycorrhizae. However, AM colonization in the<br />

mangrove <strong>plant</strong>s of the Great Nicobar Islands in India has been reported in<br />

the past. Among the monocots, Cyperaceae and Juncaceae often do not form<br />

mycorrhizal associations. In the dicots, Brassicaceae, Chenopodiaceae, Proteaceae,<br />

Restionaceae, Zygophylaceae, Lecythidaceae, Sapotaceae and all families<br />

of Centrospermae do not form mycorrhizae. Families rich in glucosinolates<br />

predominantly lack mycorrhizae because of the inhibitory action on<br />

fungal growth (Vierheilig et al. 2000).<br />

Mycorrhizae form the connecting link between the biotic and geochemical<br />

portions of the ecosystem ( Miller and Jastrow 1994). Mycorrhizae aid the<br />

<strong>plant</strong> in better growth by assisting it in absorbing useful nutrients from the<br />

soil, in the competition between <strong>plant</strong>s and in increasing the diversity of a<br />

given area (Koide and Dickie 2002; Perotto et al. 2002). Owing to their role in<br />

nutrient cycling, mycorrhizae keep more nutrients in the biomass and,<br />

thereby increase the productivity of the ecosystem. Mycorrhizal links<br />

between seedlings and mature trees may help the seedlings in establishing<br />

themselves by providing them with the required nutrients.<br />

AM form hyphal links between <strong>plant</strong>s of different species which could be<br />

involved in the transfer of nutrients between <strong>plant</strong>s. At the <strong>plant</strong> community<br />

level, AM hyphae form a network – the wood-wide web that facilitates carbon<br />

exchange between the host and the symbiont, uptake of nutrients and their<br />

movement between <strong>plant</strong>s (Watkins et al. 1996; Fitter et al. 1998; Helgason et<br />

al. 1998; Sen 2000). AM are present in most soils and are generally not consid-


5 Diversity and Functions of Soil Microflora in Development of Plants 77<br />

ered to be host-specific. However, population sizes and species composition is<br />

highly variable and influenced by <strong>plant</strong> characteristics. A number of environmental<br />

factors such as temperature, soil pH, soil moisture, P and N levels,<br />

heavy metal concentration (Boddington and Dodd 1999), the presence of<br />

other microorganisms, application of fertilizers and soil salinity (Bationo et<br />

al. 2000) may affect population diversity and size.<br />

Mycorrhizae regulate <strong>plant</strong> communities by affecting competition, composition<br />

and succession (Kumar et al. 1999). In competition between <strong>plant</strong>s,<br />

mycorrhizae in the soil favor the growth of one species and are detrimental to<br />

other competing species. AM may regulate competition between <strong>plant</strong>s by<br />

making available to mycorrhizal <strong>plant</strong>s, the resources that are not available to<br />

nonmycorrhizal neighbors. AM symbiosis may also increase intraspecific<br />

competition (Facelli et al. 1999). As a result, density of individuals of a single<br />

species would be reduced, thereby allowing the co-existence of individuals of<br />

different species. This would lead to an increase in species diversity.<br />

Mycorrhizae govern species composition in communities by influencing<br />

<strong>plant</strong> fitness at the establishment phase and preventing nonmycorrhizal<br />

<strong>plant</strong>s from growing in soils colonized by them. This has a selective advantage<br />

for the fungus. Maintaining a high proportion of compatible host<br />

species at the expense of noncompatible species provides the fungus with an<br />

undisturbed carbon supply (Francis and Read 1994). Owing to their role in<br />

nutrient uptake, mycorrhizae may play an important part in determining the<br />

rate and direction of the process by influencing either the outcome of succession<br />

or by affecting the composition and diversity of species (Smith and<br />

Read 1997).<br />

The above pattern of succession seems to be true in temperate regions. In<br />

tropical countries like India, mycorrhizal <strong>plant</strong>s act as pioneer species. It has<br />

been reported that mycorrhizal species like Adhatoda vasica, Solanum xanthocarpum,<br />

Sporobolus sp. and Desmostachya sp. form the pioneer vegetation<br />

in alkaline wastelands (Janardhanan et al. 1994).<br />

The functioning of <strong>plant</strong> communities depends to a large extent on decomposition,<br />

which makes nutrient elements available to the <strong>plant</strong>s. Decomposition<br />

is essentially carried out by the soil biota (bacteria, fungi, nematodes,<br />

arthropods, annelids), which breaks down the litter and organic matter of the<br />

soil (Zhu and Ehrenfeld 1996). The external mycelium of both ectomycorrhiza<br />

and AM interact with these organisms. Some soil organisms have been found<br />

to feed on AM spores. By bringing about changes in the abundance and activity<br />

of decomposers, mycorrhizal fungi are believed to hasten the process of<br />

decomposition and thereby the nutrient cycling.<br />

An important role played by the fungal component in <strong>plant</strong> growth is the<br />

absorption of nutrients from the soil, making them available to the <strong>plant</strong>s<br />

(Hooker and Black 1995; Goicoechea et al. 2000). Nitrogen, phosphorous and<br />

potassium are the important nutrient elements required by <strong>plant</strong>s for their<br />

growth.AM assist in nutrient uptake by exploring the soil beyond the range of


78<br />

Ramesh Chander Kuhad et al.<br />

roots (Torrisi et al. 1999). Extra radical AM hyphae augment the uptake of<br />

nutrients from up to 12 cm away from the root <strong>surface</strong> (Cui and Caldwell<br />

1996).<br />

The network of hyphae may increase the availability of nutrients like N or<br />

P from locked sources by decomposing large organic molecules (George et al.<br />

1995). Mycorrhizal fungi are also known to develop bridges connecting the<br />

root with the surrounding soil particles to improve both nutrient acquisitions<br />

by the <strong>plant</strong> and soil structure (Varma 1995; Hodge 2000). Unlike N 2-fixing<br />

bacteria that function as biological fertilizers, AM fungi do not add P to the<br />

soil. They only improve its availability to the <strong>plant</strong>. There is evidence that<br />

phosphatase activity is higher in the rhizosphere around AM than in nonmycorrhizal<br />

roots. P uptake is enhanced with the increase in root colonization by<br />

mycorrhizae.A system of barter operates, the colonized <strong>plant</strong> provides photosynthate<br />

to the fungus, in return, its extraradical hypha makes more P available<br />

to the host (Merryweather and Fitter 1995). Plants rely more on AM when<br />

growing in soils deficient in P (Bationo et al. 2000). Depriving a <strong>plant</strong> in its<br />

natural environment of mycorrhizae on a long-term basis can also reduce P<br />

acquisition. Plants that are nonmycorrhizal invest more in their vegetative tissues<br />

like shoots and roots. In contrast, in mycorrhizal <strong>plant</strong>s, the functions of<br />

the roots are taken over by the AM hyphae, thereby permitting the host <strong>plant</strong><br />

to invest its resources in reproductive organs.<br />

Nitrogen occurs in the soil predominantly in the form of nitrate and<br />

ammonia, which is water-soluble and readily available for absorption. Studies<br />

with labelled N have revealed that the AM increases N uptake by <strong>plant</strong>s (Bijbijen<br />

et al. 1996; Faure et al. 1998; Mädder et al. 2000). AM fungal hyphae have<br />

been credited with the uptake and transfer of large amounts of N from the soil<br />

to the host (Johansen et al. 1996; Hodge et al. 2000). However, there is little reciprocal<br />

transfer of N from the <strong>plant</strong> to the fungi, which makes uptake and<br />

assimilation of N by the symbiont essential for its growth (Bijbijen et al. 1996).<br />

Since AM form underground hyphal links between <strong>plant</strong>s, N transfer between<br />

<strong>plant</strong>s by means of such links is possible. Using labelled 15 N, Frey and Schüepp<br />

(1993) demonstrated that N flows from Trifolium alexandrium to Zea mays<br />

via AM fungal network. AM are believed to enhance N 2-fixation by symbiotic<br />

legumes by increasing root and nodule biomass, N 2-fixation rates, root N<br />

absorption rates, and <strong>plant</strong> N and P content (Olesniewicz and Thomas 1999).<br />

Mycorrhizae have also been reported to be involved in the uptake of other<br />

micro- and macro-nutrients like K, S, Mg, Zn, Cu, Ca and Na (Díaz et al. 1996;<br />

Hodge 2000).<br />

Soil microorganisms, particularly saprophytic fungi affect the development<br />

and function of AM symbiosis. Fracchia et al. (2000) investigated the<br />

effect of the saprophytic fungus Fusarium oxysporum on AM colonization<br />

and <strong>plant</strong> dry matter was studied in greenhouse and field experiments using<br />

host <strong>plant</strong>s, maize, sorghum, lettuce, tomato, wheat, lentil and pea and AM<br />

fungi, Glomus mosseae, G. fasciculatum, G. intraradices, G. clarum and G.


5 Diversity and Functions of Soil Microflora in Development of Plants 79<br />

deserticola. The greatest <strong>plant</strong> growth and AM colonization responses in sterilized<br />

and nonsterilized soils was observed with pea, G. deserticola and<br />

sodium alginate pellets as carrier for F. oxysporum inoculum.Application of F.<br />

oxysporum increased shoot dry matter, N and P concentrations of pea and<br />

sorghum <strong>plant</strong>s and the level of AM fungi colonization.<br />

Piriformospora indica, a newly described axenically cultivable phytopromotional<br />

endosymbiont, which mimics the capabilities of AM fungi, was<br />

recently described by Varma et al. (1999) and Singh et al. (2000). The fungus<br />

has a broad host spectrum and inoculation with the fungus and application<br />

of culture filtrate promotes <strong>plant</strong> growth and biomass production. It mobilizes<br />

the insoluble phosphate and translocates the phosphorus to the host in<br />

an energy-dependent process. As a biological hardening agent of micropropagated<br />

<strong>plant</strong>s, it renders more than 90 % survival rate for laboratory to<br />

field transferred <strong>plant</strong>lets. Regenerative protoplasts of P. indica have been<br />

successfully isolated, which opens up the possibility of improving symbiosis<br />

by transgenic manipulation of the fungal component in a symbiosis-specific<br />

manner.<br />

In the ectomycorrhizal (EM) symbiosis between fungi and trees, the fungus<br />

completely ensheaths the tree roots and takes over water and mineral nutrient<br />

supply, while the <strong>plant</strong> supplies photosynthate (Wiemken and Boller 2002). N<br />

and P are the main elements limiting <strong>plant</strong> growth in terrestrial ecosystems.<br />

One of the great assets of the ectomycorrhizal symbiosis is its capability to<br />

short-circuit nutrient uptake from organic material to the symbiotic partner.<br />

In addition to mobilizing mineral nutrients from organic sources, EM fungi<br />

may also link <strong>plant</strong>s to rock directly though secretion of organic acids and<br />

solubilizing nutrients from the mineral part of soil. Many EM fungi also retain<br />

considerable saprotrophic potential, for example, production of lignindegrading<br />

enzymes, a quality that benefits the symbionts in the acquisition of<br />

nutrients from lignin-rich organic material.<br />

Sulfur nutrition of <strong>plant</strong>s is largely determined by sulfate uptake of the<br />

roots, the allocation of sulfate to the sites of sulfate reduction and assimilation,<br />

the reduction of sulfate to sulfide and its assimilation into reduced sulfur-containing<br />

amino acids and peptides and the allocation of reduced sulfur<br />

to growing tissues (Rennenberg 1999). EM colonization of oak and beech tree<br />

roots can alter the response of sulfate uptake to sulfate availability in the soil<br />

and enhance xylem transport of sulfate to the leaves. Simultaneously, sulfate<br />

reduction in the roots seems to be stimulated by EM association. These interactions<br />

between EM association and the processes involved in sulfur nutrition<br />

are required to provide sufficient amounts of reduced sulfur for<br />

increased protein synthesis that is used to enhance tree growth.<br />

Information on the diversity of ericoid mycorrhizal endophytes in the Ericaeae<br />

and Epacridaceae has been compiled over the years by several authors<br />

(Varma and Bonfante 1994; Read 1996; Bergero et al. 2000; Berch 2001; Perotto<br />

et al. 2002). Hymenoscyphus ericae and Oidiodendron sp. appear to be the


80<br />

Ramesh Chander Kuhad et al.<br />

dominant fungi in the diverse assemblages of symbionts colonizing the<br />

<strong>plant</strong>s. Unlike other mycorrhizal symbionts, where the fungal partner produces<br />

an extensive mycelial phase that grows from the host roots and act as an<br />

efficient nutrient collecting system, ericoid fungi produce little mycelial<br />

growth external to the root. It is now widely accepted that the major benefit<br />

conferred upon the ericaceous host <strong>plant</strong> by mycorrhizal infection is enzymatic<br />

degradation of organic nutrient sources in soil and transfer of much of<br />

the resulting products across the fungus–root interface (Cairney and Burke<br />

1998). Ericoid mycorrhizal fungi produce a range of extracellular enzymes<br />

including cellulases, hemicellulases, ligninases, pectinases, phosphatases, proteases<br />

and polyphenol oxidases which not only have the potential to mediate<br />

utilization of organic sources of nitrogen and phosphorus in soil, but also<br />

allow them to decompose the <strong>plant</strong> cell wall, facilitating access to mineral<br />

nutrients sequestered within the walls of moribund <strong>plant</strong> cells.<br />

Ericoid mycorrhizal fungi can interact with metals in the surrounding<br />

environment by releasing extracellular metabolites that can modify heavy<br />

metal bioavailability. Ericoid mycorrhizal symbiosis can reduce metal toxicity<br />

to the host, allowing <strong>plant</strong>s to survive in soils with potentially toxic amounts<br />

of soluble and insoluble metal species. In addition to metabolites, fungi can<br />

also respond to the presence of metals with the release of specific proteins in<br />

the surrounding medium. The mechanism of arsenic tolerance in ericoid<br />

mycorrhizal fungi has been investigated by Sharples et al. (2000). Arsenic<br />

enters the cell through the phosphate transporter, causing the fungi to<br />

enhance both phosphate and arsenate uptake.Active and specific efflux mechanisms<br />

are adopted by ericoid fungi from polluted sites to decrease cellular<br />

concentrations of arsenic while retaining phosphate.<br />

3.2 Actinorhiza<br />

Actinorhiza is the symbiotic association between the actinomycete Frankia<br />

and the roots of several nonleguminous woody angiosperms. The symbiosis<br />

is established when Frankiae infect roots and lead to the development of nodules<br />

that are active in N 2 fixation. Actinorhizal <strong>plant</strong>s are distributed among<br />

24 genera of 8 angiosperm families (Verghese et al. 1998). These <strong>plant</strong>s are<br />

neither related, nor do they share characters that would identify them as<br />

uniquely symbiotic. The large phylogenetic disparity in comparison to the<br />

symbiotic legumes suggests that relationship between angiosperms and<br />

Frankia occurred early in evolutionary time resulting in significant divergence<br />

since then.<br />

Morphological, physiological and cytochemical criteria are employed to<br />

assign strains to the genus Frankia (Lechevalier 1994; Maunuksela 2001). The<br />

morphological features used for taxonomic purposes include the formation<br />

of septate, branching hyphae, production of multilocular sporangia, presence


5 Diversity and Functions of Soil Microflora in Development of Plants 81<br />

of nonmotile spores in multilocular sporangia and the production of thickwalled,<br />

lipid encapsulated structures called vesicles – the seat of nitrogen fixation.<br />

On the basis of host specificity, Frankia isolates have been classified<br />

into four major groups: (1) Alnus–Myrica; (2) Casuarina–Myrica; (3) Myrica-<br />

Eleagnus; (4) members of Elagenceae.<br />

Actinorhizal genera have a worldwide distribution with a few exemptions.<br />

Africa, with the exception of Myrica species, is lacking in native actinorhiza.<br />

Actinorhizal genera can be characterized as inhabiting nutrient-poor sites in<br />

temperate regions. The Frankia-Alnus symbiosis is the most extensively studied<br />

actinorhiza. Alnus, Casuarina and Elaeagnus are the most widely distributed<br />

actinorhizal <strong>plant</strong>s largely due to the introduction by man to all the continents.<br />

Although the N 2 -fixing potential of Frankia-Alnus symbiosis may be<br />

high, the amount of nitrogen actually fixed is low because of unfavorable<br />

environmental conditions. Therefore, proper management practices that optimize<br />

efficiency of the nitrogen-fixing system are required (Dommergues<br />

1997).<br />

Frankia populations occur in three niches, the root nodules, the rhizosphere<br />

and the soil. In the soil, Frankia can be (1) a symbiont of actinorhizal<br />

<strong>plant</strong>s, (2) an associate of nonhost <strong>plant</strong>s or (3) a saprophyte. Although the<br />

biochemical and molecular events of the Frankia-actinorhizal <strong>plant</strong> symbiosis<br />

are not as well understood as the Rhizobium-legume symbiosis, there is a<br />

regulated series of events leading to this close association between Frankia,<br />

the compatible host <strong>plant</strong> and the subsequent formation of root nodules.<br />

Frankia infection can be through (1) root hair (Casuarinaceae and Myricaceae)<br />

or (2) through intercellular spaces of the root epidermis and root<br />

cortex (Elaeagnceae and Ceanothus). In Alnus, infection is initiated via root<br />

hairs, which become branched in response to Frankia contact (Maunuksela<br />

2001). The host cell produces wall-like material containing pectin, hemicellulose<br />

and encapsulates the Frankia hyphae within the host cells. Division of<br />

root cortical cells results in the formation of a prenodule. The actual nodule<br />

lobe originates in the pericycle and becomes infected by penetrating Frankia<br />

hyphae.<br />

Actinorhizal <strong>plant</strong>s are pioneer species that have the ability to colonize lownitrogen<br />

and disturbed sites such as fires, volcanic eruptions and flooding.<br />

They facilitate succession in the sites by soil solubilization and augmenting<br />

N 2-content. A well-developed actinorhizal <strong>plant</strong> root system favors soil-binding<br />

capacity, which improves the quality of impoverished soils and strongly<br />

supports the use of these <strong>plant</strong>s in land reclamation. Many actinorhizal <strong>plant</strong>s<br />

are also mycorrhizal and possess the ability to absorb other nutrients. As succession<br />

progresses, non N 2 -fixing <strong>plant</strong>s are able to replace the original actinorhizal<br />

pioneers. Myrica faya growing at a volcanic site in Hawaii was able to<br />

fix 18.5 kg N/ha/year and significantly increased the amount of available N 2 in<br />

soils under the <strong>plant</strong>s. Non N 2-fixing <strong>plant</strong>s growing in the vicinity of M. faya<br />

accumulated greater biomass in comparison to <strong>plant</strong>s growing at sites away


82<br />

Ramesh Chander Kuhad et al.<br />

from Myrica. This is indicative of the importance of actinorhizal <strong>plant</strong>s in the<br />

ecosystem development. Actinorhizal <strong>plant</strong>s are also used as intercrops for<br />

other tree species (Dommergues 1997).<br />

3.3 Plant Growth-Promoting Rhizobacteria<br />

The rhizosphere is the region of soil surrounding the roots that is subject to<br />

influence by the root and rhizobacteria are <strong>plant</strong>-associated bacteria that are<br />

able to colonize and persist on roots (Subba Rao 1999). Several genera of bacteria<br />

such as Arthrobacter, Agrobacterium, Azotobacter, Burkholderia, Cellulomonas,<br />

Micrococcus, Flavobacterium, Mycobacterium, Pseudomonas and<br />

others have been reported to be present in the rhizosphere (see chap. 12, this<br />

vol.). It has been demonstrated that the metabolic activities of bacteria associated<br />

with the rhizosphere are different from those of the nonrhizosphere<br />

soils. Electron and direct microscopy has revealed that up to 10 % of the root<br />

<strong>surface</strong> is colonized by microorganisms in a random fashion depending on<br />

the presence of soil organic matter. Some strains of <strong>plant</strong> growth-promoting<br />

rhizobacteria (PGPR) can effectively colonize <strong>plant</strong> roots and protect <strong>plant</strong>s<br />

from diseases caused by a variety of root pathogens and growth promotion of<br />

<strong>plant</strong>s through formation of <strong>plant</strong> growth hormones. Considerable progress<br />

has been made using molecular techniques to elucidate the important microbial<br />

factors or genetic traits involved in the PGPR-stimulated <strong>plant</strong> growth<br />

and in the suppression of fungal root diseases (Glick and Bashan 1997;<br />

Kumari and Srivastava 1999; Bloemberg and Lugtenberg 2001; Zehnder et al.<br />

2001). Several genera of allelopathic nonpathogenic bacteria have been identified<br />

and characterized which produce <strong>plant</strong> growth-inhibiting allelochemicals<br />

(Barazani and Friedman 2001). Allelochemicals like phytoxins,<br />

geldanamycin, nigericin and hydanthocidin have been isolated from Streptomyces<br />

viridochromogenes.<br />

PGPR can affect <strong>plant</strong> growth either directly or indirectly. The direct<br />

effect of PGPR include providing the host <strong>plant</strong>s with fixed nitrogen, P and<br />

Fe solubilized from the soil and phytohormones that are synthesized by the<br />

bacteria (Glick 1995). The indirect effect on <strong>plant</strong> growth occurs when PGPR<br />

reduces or prevents the harmful effects of one or more phytopathogenic<br />

organisms. PGPR effective in biocontrol produce a variety of substances<br />

including antibiotics, siderophores and a variety of enzymes (chitinase, protease,<br />

lipase, b-1,3-glucanase etc.) to limit the damage to <strong>plant</strong>s by phytopathogens.<br />

PGPR have also been reported to reduce heavy metal toxicity<br />

in <strong>plant</strong>s (Burd et al. 2000).<br />

Symbiotic nitrogen fixation has long been considered to be an excellent<br />

replacement of N fertilization. The most efficient nitrogen fixers are strains of<br />

Rhizobium, Sinorhizobium, Mesorhizobium, Bradirhizobium and Azorhizobium,<br />

which form a host-specific symbiosis with leguminous <strong>plant</strong>s (Paul and


5 Diversity and Functions of Soil Microflora in Development of Plants 83<br />

Clark 1998; Subba Rao 1999). The genes involved in nitrogen fixation, nitrogen<br />

assimilation and regulation in various bacteria have been studied extensively<br />

(Glick and Bashan 1997; Bloemberg and Lugtenberg 2001; Rengel 2002).<br />

Several of the nif and fix genes, involved in N 2-fixation, have been characterized<br />

in different nitrogen fixers. Most of the organism contains similar nitrogenase<br />

complexes. Increased efficacy of N 2 -fixation can be achieved by selecting<br />

and manipulating the best combination of host genotype and bacteria.<br />

Improvement in the symbiotic relationship in suboptimal environmental situations<br />

related to soil-borne or environmental stress is also important to<br />

improve N 2-fixation.<br />

Free-living N 2-fixing rhizobacteria are capable of fixing atmospheric nitrogen.<br />

The aerobic, free-living bacteria that utilize organic substrates as a source<br />

of energy include Azotobacter, found in neutral and alkaline soils. Members of<br />

the same family Beijerinckia and Derxia have a broader pH range and are<br />

more often found in acidic soils in the tropics. Azospirillum, Acetobacter,<br />

Herbaspirillum and Azoarcus have frequently been found associated with<br />

grasses (Steenhoudt and Vanderleyden 2000). Azotobacter and Beijerinckia<br />

require aerobic conditions for the production of energy required for N 2 fixation.<br />

However, in these organisms as well as other diazotrophs, the activity of<br />

nitrogenase is inhibited by O 2 and special mechanisms for the protection of<br />

nitrogenase are present. Facultative microaerophilic organisms such as<br />

Azospirillum, Klebsiella and Bacillus produce energy in the form of ATP by<br />

oxidative pathways in an environment where nitrogenase does not need to be<br />

as well protected from O 2. The amount of N 2 fixed by free-living diazotrophs<br />

such as Azotobacter and Pseudomonas is generally a few kilograms per<br />

hectare (Paul and Clark 1998). Nitrogen-fixing microorganisms in the waterlogged<br />

rice fields may contribute 40–50 kg per hectare which is a cumulative<br />

effect of free-living as well as symbiotic organisms such as blue-green algae,<br />

Azotobacter, Azospirillum, Rhizobium, Beizerinckia, Clostridium, Desulfovibrio<br />

and Pseudomonas (Subba Rao 1999).<br />

Soil amendments and artificial inoculation of beneficial rhizobacteria can<br />

induce changes in rhizosphere microflora (Bashan 1998; Bai et al. 2002). Rhizosphere<br />

nitrogen fixation could be enhanced by incorporation of N 2-fixing<br />

capacity into common rhizosphere. The large scale application of PGPR in<br />

agriculture is attractive as it substantially reduces the use of chemical fertilizers<br />

and pesticides. A growing number of PGPR are being marketed, and at<br />

present, biofertilizer application accounts for approximately 65 % of the N<br />

supply to crops worldwide (Bloemberg and Lugtenberg 2001). Integrated<br />

approaches have been applied with a combination of AM fungi or biocontrol<br />

fungi like Trichoderma and PGPR for the beneficial <strong>plant</strong> growth and disease<br />

control effects (Valdenegro et al. 2001; Elliot and Broschat 2002). Recently<br />

focus has also been directed towards the development and use of rhizobacteria<br />

as biocontrol agents to combat fungal diseases (Naseby et al. 2001; Unge<br />

and Jansson 2001).


84<br />

Ramesh Chander Kuhad et al.<br />

3.4 Phosphate-Solubilizing Microorganisms<br />

After nitrogen, phosphorus is the major <strong>plant</strong> growth-limiting nutrient,<br />

though P is abundant in soils in both inorganic and organic forms. Most of the<br />

mineral nutrients in soil solution are present in millimolar amounts, however,<br />

P is present only in micromolar (up to 10 mm) amounts. Low level of availability<br />

of P is due to high reactivity of soluble P with Ca, Fe and Al (Gyaneshwar<br />

et al. 2002). Calcium phosphates are the predominant form of P in calcareous<br />

soils, whereas inorganic P in acidic soil is associated with Fe and Al<br />

compounds. In soils with high organic matter, organic P may make up as<br />

much as 50 % of the total soluble P available in soil. Phosphate-solubilizing<br />

microorganisms (PSM) are ubiquitous in soils and play an important role in<br />

supplying P to <strong>plant</strong>s in a sustainable manner. Although a lot of laboratory<br />

work on phosphate solubilization has been done, the results of field trials<br />

were highly variable (Nahas 1996).<br />

In spite of the importance of PSM in agriculture, the detailed biochemical<br />

and molecular mechanisms of P solubilization is not known. Mineral P solubilizing<br />

ability of microbes could be linked to specific genes which may be<br />

present in even non P-solubilizing bacteria (Goldstein 1995). The ability to<br />

solubilize the mineral–phosphate complexes has been attributed to the ability<br />

of PSM to reduce the pH of the surroundings by releasing organic acids such<br />

as acetate, lactate, oxalate, tartarate, succinate, citrate, gluconate etc. (Kim et al.<br />

1998; Ezawa et al. 2002). These organic acids can either dissolve the mineral<br />

phosphate as a result of anion exchange or can chelate Fe or Al ions associated<br />

with the phosphate. However, acidification does not seem to be the only<br />

mechanism of P solubilization, as the ability to reduce pH in some cases does<br />

not correlate with the ability to solubilize mineral phosphates (Jones 1998;<br />

Gyaneshwar et al. 2002).<br />

Plants have been shown to benefit from the association with microorganisms<br />

under P-deficient conditions, either resulting from a better uptake of the<br />

available P or by accession of the nonavailable form of P-source.Various kinds<br />

of bacteria and fungi have been isolated and characterized for their ability to<br />

solubilize mineral phosphate complexes. Although P-solubilizing bacteria<br />

outnumber P-solubilizing fungi in soil, fungal isolates generally exhibit<br />

greater P-solubilizing ability than bacteria in both liquid and solid media<br />

(Goldstein 1995; Gyaneshwar et al. 2002). Phosphate-solubilizing strains of<br />

bacteria Enterobacter agglomerans (Kim et al. 1998) and Azotobacter chroococcum<br />

(Kumar and Narula 1999) have been isolated from wheat rhizosphere<br />

and characterized for solubilization of hydroxyapetite, tricalcium phosphate<br />

and Mussoorie rock phosphate in laboratory experiments. Nautiyal et al.<br />

(2000) described the isolation and characterization of four unidentified bacterial<br />

strains from the chickpea rhizosphere in alkaline soil. NBRI 2601 was<br />

the most efficient strain in terms of its capability to solubilize phosphorus in<br />

the presence of 10 % salt, pH 12 and 45 °C. Seed inoculation with an acid-tol-


5 Diversity and Functions of Soil Microflora in Development of Plants 85<br />

erant strain of Bacillus sp. significantly increased the vegetative and grain<br />

yield of fingermillet, maize, amaranth, buckwheat and french bean (Pal 1998).<br />

Although <strong>plant</strong>s inoculated with PSM exhibit increased growth and P contents<br />

in laboratory studies, large variations have been found in the effectiveness<br />

of inoculations in field conditions.<br />

Phosphate solubilizing fungi and their role in <strong>plant</strong> nutrition and growth<br />

have been extensively studied. Among the known fungal genera are<br />

Aspergillus (Goenadi et al. 2000; Narsian and Patel 2000), Penicillium<br />

(Whitelaw et al. 1999; Reyes et al. 2001), Rhizoctonia (Jacobs et al. 2002) and<br />

Cyathus (Singal et al. 1991). Supplementation of A. niger cultivated on sugar<br />

beet waste material to soil significantly improved the growth rate and shoot P<br />

concentration of Trifolium repens (Vassilev et al. 1996). Reddy et al. (2002)<br />

reported the biosolubilization of different rock phosphates by three isolates of<br />

A. tubingensis for the first time. Altomare et al. (1999) investigated the capability<br />

of biocontrol fungus Trichoderma harzianum to solubilize MnO 2, metallic<br />

zinc and rock phosphate and discussed its possible role in <strong>plant</strong> growth.<br />

Application of encapsulated fungal or bacterial cell systems for effective use<br />

as soil microbial inoculants in P solubilization and <strong>plant</strong> nutrition has been<br />

discussed in detail by Vassilev et al. (2001).<br />

Nodule formation in legumes is often limited by the availability of P (Subba<br />

Rao 1999). While there are only a few reports on P solubilization by Rhizobium<br />

(Chabot et al. 1996), the improvement in the efficiency of N 2-fixation in<br />

legumes has been demonstrated by supplementation of P in alfalfa, clover,<br />

cow pea and pigeon pea (Al-Niemi et al. 1997). In chickpea and barley growing<br />

in soils treated with insoluble phosphate and inoculated with Mesorhizobium<br />

mediterraneum, the P content increased by 100 and 125 %, respectively<br />

(Peix et al. 2001). The dry matter, N, K, Ca and Mg contents in both <strong>plant</strong>s also<br />

increased significantly. A coculture inoculum of Rhizobium meliloti and a<br />

phosphate-solubilizing fungus, Penicilium bilalii increased the P uptake of<br />

several field crops (Rice et al. 1995). Co-inoculations of AM fungi with PSM<br />

have shown positive effects on <strong>plant</strong> growth and crop yield (Toro et al. 1997;<br />

Ezawa et al. 2002). Beneficial effects of enriching vermicompost by nitrogenfixing<br />

and phosphate-solubilizing bacteria have also been demonstrated<br />

(Kumar and Singh 2001).<br />

3.5 Lignocellulolytic Microorganisms<br />

The high cellulose and lignin contents of <strong>plant</strong> residue incorporated into soil<br />

emphasize the importance of lignocellulolytic microorganisms in the mineralization<br />

processes in soil (Kuzyakov and Domanski 2000). The chemical<br />

composition of the entire <strong>plant</strong> residues, their decomposition and biochemical<br />

transformations in the soil during humification has been investigated in<br />

detail (Paul and Clark 1998). The importance of microbial biomass and extra-


86<br />

Ramesh Chander Kuhad et al.<br />

cellular lignocellulolytic enzyme activity in the assessment of soil quality is<br />

established by the essential role of soil microbes in nutrient cycling within<br />

agricultural ecosystems (Christensen and Johnston 1997). During the microbial<br />

degradation and humification of <strong>plant</strong> residues, about 80 % of the residual<br />

carbon is released to the atmosphere as CO 2 (Omar 1994). The amendment<br />

of infertile or saline soils with <strong>plant</strong> residues and their subsequent<br />

degradation by cellulolytic soil microflora with a concomitant increase in CO 2<br />

could increase soil aeration, improve its structure and also increase soil fertility.<br />

The activities of cellulolytic microbes affect the availability of energy and<br />

specific nutrients to a group of organisms deficient in hydrolytic enzyme<br />

activities (Jensen and Nybroe 1999).<br />

Soils managed with organic inputs generally have larger and more active<br />

microbial populations than those managed with mineral fertilizers (Badr El-<br />

Din et al. 2000). Reincorporation of organic matter into the soil improves soil<br />

fertility, enhances microbial growth and buffers the soil environment from<br />

sudden changes. There are many types of agroindustrial organic refuse which<br />

can be transformed and applied to soil as crop amendments, such as compost,<br />

thus reducing the need for chemical fertilizers. During the composting<br />

process, the organic substrate present in the agricultural wastes is mainly<br />

transformed oxidatively into a stabilized organic matter. The slow transformation<br />

of lignocellulosic material results in the formation of humic substances.<br />

Several researchers have established a positive correlation between<br />

the amount of humic substances and promotion of <strong>plant</strong> growth. Application<br />

of different combinations of coir with peat and vermiculate significantly<br />

increased the growth of tomato trans<strong>plant</strong>s with respect to root dry weight,<br />

stem diameter and leaf area (Arenas et al. 2002).<br />

Straw incorporation could also be beneficial in enhancing symbiotic nitrogen<br />

fixation and crop growth (Abd-Alla and Omar 1998). In nonsymbiotic<br />

nitrogen fixation studies in the laboratory and in the field, a significant<br />

increase in nitrogenase activity associated with the breakdown of straw after<br />

inoculation with various combinations of cellulolytic fungi and bacteria has<br />

been reported (Halsall and Gibson 1991; Chapman et al. 1992). Application of<br />

wheat straw with cellulolytic fungi, Trichoderma harzianum significantly<br />

enhanced growth, nodulation, nodule efficiency and increased the concentration<br />

of Ca, Mg and K in the shoots and roots of fenugreek <strong>plant</strong>s grown in<br />

saline soil (Abd-Alla and Omar 1998). The increase in dry matter production<br />

and nitrogen content was due to improved N 2 fixation reflected by enhanced<br />

formation and growth of nodules as well as nitrogenase activity.<br />

Inoculation of straw with lignocellulolytic organisms offers potential for<br />

manipulating and improving the composting of cellulosic waste (Verstraete<br />

and Top 1999; Hart et al. 2002). Composts produced using this method provide<br />

a more sustainable approach to agriculture, enabling subsistence farmers<br />

to utilize their agricultural waste products as a means to improve soil quality.<br />

Saprophytic lignin-decomposing basidiomycetes isolated from <strong>plant</strong> litter


5 Diversity and Functions of Soil Microflora in Development of Plants 87<br />

were found to play an important role in soil aggregation and stabilization<br />

(Caesar-Ton That and Cochran 2000). The basidiomycete produced large<br />

quantities of extracellular water-insoluble and heat-resistant materials that<br />

bind soil particles into aggregates.<br />

Differences in the chemical properties of the organic matter from highly<br />

lignocellulosic compost after incubation with two lignocellulolytic microorganisms<br />

were studied by Requena et al. (1996). Inoculation with Trichoderma<br />

viride and Bacillus sp. enhanced degradation processes and the degree of<br />

organic matter humification. Both degradation-humification pathways beneficially<br />

affected the lettuce growth demonstrating that inoculation with lignocellulolytic<br />

microbes may be a useful tool to improve agronomic properties of<br />

lignocellulosic wastes by modifying the chemical structure and properties of<br />

their organic matter. Rajbanshi et al. (1998) found significant positive effects<br />

of seeding material (substrates with a high number of degradative microbes)<br />

on total organic carbon and organic matter contents of grass straw-leaf mix.<br />

Temporal changes in soil moisture, soil temperature, and carbon input<br />

from crop roots, rhizosphere products (root exudate, mucilage, sloughed cells<br />

etc.), and crop residues can have a large effect on soil microbial activity<br />

(Jensen et al. 1997; Ritz et al. 1997). Crop growth often stimulates an increase<br />

in the size of microbial biomass during the growing season and after harvest.<br />

Enzyme activity displays different temporal patterns of the various soil<br />

enzymes. Some cellulases are closely related to inputs of fresh organic materials,<br />

<strong>plant</strong> growth and <strong>plant</strong> residues, while others appear to be more sensitive<br />

to soil temperature and moisture.<br />

Due to their dynamic nature, soil microbial biomass and soil enzymes<br />

respond quickly to changes in organic matter input. In a field experiment<br />

after 8 years of cultivation with low- or high-organic matter input, pronounced<br />

and constant increase in endocellulase and b-glucosidase activities<br />

and variable increase in microbial biomass carbon and cellobiohydrolase<br />

activity was observed over the sampling period (Debosz et al. 1999). Temporal<br />

variations in endocellulase activity showed a different pattern from those for<br />

b-glucosidase activity, with highest activity in the autumn/winter and early<br />

summer samplings. On all sampling dates, endocellulase activity in the higher<br />

organic matter was about 30 % higher than in the low organic matter treatments.<br />

Specific organic amendments such as mulched straw has been reported to<br />

influence soil suppression of <strong>plant</strong> diseases (Knudsen et al. 1999). Many fungi,<br />

known as antagonists to <strong>plant</strong> pathogens, e.g., Trichoderma sp., produce a<br />

wide range of cellulolytic enzymes which are believed to be associated with<br />

their antagonistic abilities. Rasmussen et al. (2002) investigated the relationship<br />

between soil cellulolytic activity and suppression of seedling blight of<br />

barley caused by Fusarium culmorum in arable soils. A bioassay for disease<br />

suppression in test soils indicated that the samples from 6 to 13-cm depth<br />

exhibited positive correlation between soil suppressiveness and the activities


88<br />

Ramesh Chander Kuhad et al.<br />

of b-glucosidase and cellobiohydrolase, where soil representing the highest<br />

disease suppression had the highest activity. Furthermore, soil suppressiveness,<br />

as well as the enzyme activity significantly correlated with the soil content<br />

of total C and N.<br />

4 Plant Growth Promoting Substances Produced by Soil<br />

Microbes<br />

The ability of soil microorganisms to produce various metabolites stimulating<br />

<strong>plant</strong> growth is considered to be one of the most important factors in soil<br />

fertility (Frankenberger and Arshad 1995; Paul and Clark 1998; Subba Rao<br />

1999). Some PGPR control the damage to <strong>plant</strong>s from <strong>plant</strong> pathogens by a<br />

number of different mechanisms including physical displacement and outcompeting<br />

the phytopathogen, secretion of siderophores to prevent<br />

pathogens in the immediate vicinity from proliferating, production of<br />

enzymes, antibiotics and a variety of small molecules that inhibit the phytopathogen<br />

and stimulation of systemic resistance in <strong>plant</strong>s (Glick and<br />

Bashan 1997). Microbially produced antibiotics have a potential role in indirectly<br />

promoting <strong>plant</strong> growth by controlling <strong>plant</strong> diseases (Kumari and Srivastava<br />

1999). Two prominent antifungal antibiotics are griseofulvin, a metabolic<br />

product of Penicillium griseofulvum and aureofungin, a metabolic<br />

product of Streptoverticillium cinnamomeum.<br />

Soil microorganisms produce a variety of phytohormones such as auxins,<br />

gibberellins, cytokinins, ethylene and abscisic acid.Auxin production is widespread<br />

among many soil and rhizosphere microorganisms (fungi,bacteria and<br />

actinomycetes) and algae (Martens and Frankenberger 1993). Tryptophan is<br />

considered the physiological precursor of auxin for both <strong>plant</strong> and soil<br />

microbes. A number of indole compounds and phenylacetic derivatives have<br />

been reported with auxin activity. Indole-3-acetic acid (IAA) is considered the<br />

most physiologically active auxin in <strong>plant</strong>s. Auxins are known to affect cell<br />

enlargement involving cell wall extensibility. Plant growth responses also<br />

include root and shoot dry weights, root/stem elongation and root/shoot<br />

ratios.Species of Agrobacterium,Azospirillum,Pseudomonas,Rhizobium,Ustilago,<br />

Gymnosporangium, Rhizopus and Synchytrium produce IAA in pure cultures<br />

or in association with higher <strong>plant</strong>s (Subba Rao 1999).<br />

Gibberellins (GA) are an important group of <strong>plant</strong> hormones that are<br />

diterpenoid acids. The involvement of GA in almost all phases of <strong>plant</strong> growth<br />

and development, starting from germination to senescence is well known.<br />

However, the most prominent physiological effect of GA is in shoot elongation.<br />

Some other <strong>plant</strong> growth related functions of GA include overcoming<br />

dormancy and dwarfism in <strong>plant</strong>s, inducing flowering in some photoperiodically<br />

sensitive and other low temperature-dependent <strong>plant</strong>s, and contributing<br />

to fruit setting. Several soil microbes are known to produce gibberellins or


5 Diversity and Functions of Soil Microflora in Development of Plants 89<br />

gibberellin-like substances (Kumar and Lonsane 1989; Steenhoudt and Vanderleyden<br />

2000). The common bacterial genera are Arthrobacter, Azotobacter,<br />

Azospirillum, Pseudomonas, Rhizobium, Bacillus, Brevibacterium and Flavobacterium.<br />

Actinomyces and Nocardia are the important actinomycetes and<br />

Fusarium, Gibberella, Aletrnaria, Aspergillus, Penicillium and Rhizopus are<br />

known fungi.<br />

Cytokinins, N 6 -substituted aminopurines, regulate cell division and differentiation<br />

in certain <strong>plant</strong> tissues. Cytokinins play an important role in nodule<br />

development and formation. Along with auxins, cytokinins stimulate mature<br />

root cells to undergo polyploid mitosis. Symbiotic N 2-fixing bacteria, Rhizobium,<br />

free-living N 2-fixing bacteria Azospirillum and Azotobacter,and mycorrhizal<br />

fungus, Rhizopogon roseolus are known to produce cytokinins in the<br />

rhizosphere along with other growth-promoting substances (Nieto and<br />

Frankenberger 1989). Other bacteria that produce cytokinins or cytokininlike<br />

substances include Agrobacterium, Bacillus, Paenibacillus and Pseudomonas<br />

(Timmusk et al. 1999).<br />

Ethylene (C 2H 4) is the only phytohormone that is a gas under physiological<br />

temperature and pressure. Ethylene is considered to be a promoter of senescence<br />

and an inhibitor of growth and elongation. It can also promote flowering,<br />

fruit ripening and stimulate cell elongation in certain <strong>plant</strong>s (Elsgaard<br />

2001). Bacterial species of Aeromonas, Citrobacter, Arthrobacter, Erwinia, Serratia,<br />

Klebsiela and Streptomyces, and fungal species of Acremonium,<br />

Alternaria, Mucor, Fusarium, Pythium, Neurospora and Candida are capable<br />

of producing ethylene (Subba Rao 1999).<br />

Abscisic acid (ABA) is generally involved in deceleration or cessation of<br />

<strong>plant</strong> growth.ABA is active in regulating abscission of young leaves and fruits,<br />

dormancy of buds and seeds, and ripening of fruit. ABA production in two<br />

bacterial species, Azospirillum brasilense and Rhizobium spp. and several phytopathogenic<br />

fungi such as Cercospora, Fusarium, Cladsporium, Monilia, Pestatoria<br />

and Verticillium has been demonstrated (Frankenberger and Arshad<br />

1995; Paul and Clark 1998).<br />

Siderophores are low molecular weight (


90<br />

Ramesh Chander Kuhad et al.<br />

in solubility and transport of iron, hydroxamate siderophores are also<br />

involved in iron storage.<br />

Based on the chemical nature of their coordination sites, microbial<br />

siderophores are classified as hydroxamates, catecholates, carboxylates and<br />

mixed type. Hydroxamates are produced both by bacteria and fungi. In most<br />

fungi, a mixture of siderophores is produced which varies depending on cultivation<br />

conditions. Aspergilli produce ferricrocin accompanied by fusarinines,<br />

while certain penicillia produce ferrichrome accompanied by coprogen.<br />

Similar observations have been made with Neurospora, Gliocladium,<br />

Trichoderma and Agaricus bisporus (Neilands and Leong 1986). Fusarinines<br />

(fusigens) produced by species of Fusarium and Penicillium are linear and<br />

cyclic hydroxamic acids joined by ester bonds. Ericoid mycorrhizal fungi produce<br />

ferrichrome and fusarinines.<br />

Varieties of bacterial hydroxamates are known. Ferrioxamine, produced by<br />

actinomycetes, Nocardia and Pseudomonas stutzeri, is a cyclic trihydroxamate.<br />

Citrate hydroxamates are characterized by the presence of two hydroxamates<br />

and one citrate group as ligand, it is a linear citratehydroxamic acid<br />

obtained from Klebsiella pneumonia and several enteric bacteria. Catecholate<br />

siderophores are generally less diverse than the hydroxamates, and are conjugated<br />

to amino acids or polyamine backbones. Species of Bacillus, Aeromaonas<br />

and Erwinia are known to produce catecholate siderophores. Carboxylate<br />

(complexone) siderophores are produced by Rhizopus microsporus,<br />

Rhizobium meliloti and Staphylococcus hycius. Pyoverdines, the mixed types<br />

form a wide class of mixed siderophores showing a great variety of structures.<br />

Some strains of fluorescent pseudomonads produce hydroxamate siderophores<br />

(ferribactin) in addition to pyoverdine siderophores.<br />

The <strong>plant</strong> growth-promoting rhizobacteria (PGPR) owe their <strong>plant</strong> growth<br />

promoting activity to their stronger siderophores with higher stability constants<br />

that outgrow the other bacterial population in competition for iron and<br />

finally displace them from the root <strong>surface</strong>. The siderophore-producing PGPR<br />

have become important in the biological control of <strong>plant</strong> pathogens (Glick<br />

and Bashan 1997).<br />

5 Conclusions<br />

Microorganisms play an essential role in the functioning and sustainability of<br />

soil ecosystems including biogeochemical cycling of nutrients and biodegradation.<br />

Recent advances in soil community analysis using molecular and biochemical<br />

approaches have helped us understand the enormous microbial<br />

diversity and their functional significance in nutrient recycling in soil and<br />

<strong>plant</strong> development. Soil diversity exceeds that of aquatic environments and<br />

provides a great resource for the biological exploitation of novel organisms,<br />

processes and products. Microbes isolated from soil and developed as biofer-


5 Diversity and Functions of Soil Microflora in Development of Plants 91<br />

tilizers or inoculants play an important role in enhancing <strong>plant</strong> growth<br />

enhancing efficiency of biological nitrogen fixation, availability of P, trace elements<br />

such as Fe and Zn, and production of <strong>plant</strong> growth substances. The<br />

development of better screening procedures and understanding the genetic<br />

basis of rhizosphere competence will help in developing novel microbial inoculants<br />

that will be better suited to survive and perform their desirable function<br />

in a natural environment.As we explore the soil microbial diversity more,<br />

we must remember that the microbes evolve more quickly than we can study<br />

them, providing an ever-increasing diversity of function, not only in agriculture,<br />

but also for industrial applications.<br />

Acknowledgements. The authors thank Mr. Manoj Kumar for the preparation of the<br />

manuscript.<br />

References and Selected Reading<br />

Abd-Alla MH, Omar SA (1998) Wheat straw and cellulolytic fungi application increases<br />

nodulation, nodule efficiency and growth of fenugreek (Trigonella foenum-graceum<br />

L.) grown in saline soil. Biol Fertil Soil 26:58–65<br />

Al-Niemi TS, Kahn ML, McDermott TR (1997) P metabolism in the Rhizobium tropicibean<br />

symbiosis. Plant Physiol 113:1233–1242<br />

Altomare C, Norvell WA, Bjoerkman T, Harman GE (1999) Solubilization of phosphates<br />

and micronutrients by the <strong>plant</strong> growth-promoting and biocontrol fungus Trichoderma<br />

harzianum Rifai 1295–22. Appl Environ Microbiol 65:2926–2933<br />

Arenas M,Vavrina CS, Cornell JA, Hanlon EA, Hochmuth GJ (2002) Coir as an alternative<br />

to peat in media for tomato trans<strong>plant</strong> production. Hort Science 37:309–312<br />

Badr El-Din SMS,Attia M,Abo-Sedera SA (2000) Field assessment of composts produced<br />

by highly effective cellulolytic microorganisms. Biol Fertil Soils 32:35–40<br />

Bagyaraj DJ (1984) Biological interactions with VA mycorrhizal fungi. In: Powell CL, Bagyaraj<br />

DJ (eds) VA Mycorrhiza. CRC Press, Boca Raton, pp 131–153<br />

Bagyaraj DJ, Varma A (1995) Interaction between arbuscular mycorrhizal fungi and<br />

<strong>plant</strong>s. Their importance in sustainable agriculture in arid and semiarid tropics. Adv<br />

Microb Ecol 14:119–142<br />

Bai Y, D’Aoust F, Smith DL, Driscoll BT (2002) Isolation of <strong>plant</strong> growth promoting Bacillus<br />

strains from soybean root nodules. Can J Microbiol 48:230–238<br />

Barazani O, Friedman J (2001) Allelopathic bacteria and their impact on higher <strong>plant</strong>s.<br />

Crit Rev Microbiol 27:41–55<br />

Bashan Y (1998) Inoculants of <strong>plant</strong> growth-promoting bacteria for use in agriculture.<br />

Biotechnol Adv 16:729–770<br />

Bationo A, Wani SP, Bielders CL, Vlek PLG, Mokwunye AU (2000) Crop residue and fertilizer<br />

management to improve soil organic carbon content, soil quality and productivity<br />

in the desert margins of West Africa. In: Lal R, Kimble JM, Steward BA (eds)<br />

Advances in soil science. Global climate change and tropical ecosystems. CRC Press,<br />

Washington, DC, pp 117–145<br />

Bending GD, Turner MK, Jones JE (2002) Interactions between crop residue and soil<br />

organic matter quality and the functional diversity of soil microbial communities.<br />

Soil Biol Biochem 34:1073–1082


92<br />

Ramesh Chander Kuhad et al.<br />

Berch SM (2001) Molecular diversity and phylogeny of ericoid mycorrhizal fungi. Plant<br />

Soil 244:55–66<br />

Berch SM, Allen TR, Berbee ML (2002) Molecular detection, community structure and<br />

phylogeny of ericoid mycorrhizal fungi. Plant Soil 244:55–66<br />

Bergero R, Perotto S, Girlanda M,Vidano G, Luppi AM (2000) Ericoid mycorrhizal fungi<br />

are common root associates of a Mediterranean ectomycorrhizal <strong>plant</strong> Quercus ilex.<br />

Mol Ecol 9:1639–1650<br />

Bijbijen JN, Urquiaga S, Ismaili M, Alves BJR, Boddey RM (1996) Effect of arbuscular<br />

mycorrhizae on uptake of nitrogen by Brachiaria arrecta and Sorghum vulgare from<br />

soils labelled for several years with 15 N. New Phytol 133:487–494<br />

Bloemberg GV, Lugtenberg BJJ (2001) Molecular basis of <strong>plant</strong> growth promotion and<br />

biocontrol by rhizobacteria. Curr Opin Plant Biol 4:343–350<br />

Boddington CL, Dodd JC (1999) Evidence that differences in phosphate metabolism in<br />

mycorrhizae formed by species of Glomus and Gigaspora might be related to their life<br />

cycle strategies. New Phytol 142:531–538<br />

Bridge P, Spooner B (2001) Soil fungi: diversity and detection. Plant Soil 232:147–154<br />

Burd GI, Dixon DG, Glick BR (2000) Plant growth-promoting bacteria that decrease<br />

heavy metal toxicity in <strong>plant</strong>s. Can J Microbiol 46:237–245<br />

Caesar-TonThat TC, Cochran VL (2000) Soil aggregate stabilization by a saprophytic<br />

lignin-decomposing basidiomycete fungus. I. Microbiological aspects. Biol Fertil Soil<br />

32:374–380<br />

Cairney JWG, Burke RM (1998) Extracellular enzyme activities of the ericoid mycorrhizal<br />

endophyte Hymenoscyphus ericae (Read) Korf and Kernen: their likely roles in<br />

decomposition of dead <strong>plant</strong> tissue in soil. Plant Soil 205:181–192<br />

Chabot R, Antoun H, Cesas MP (1996) Growth promotion of maize and lettuce by phosphate<br />

solubilizing Rhizobium leguminosarum biovar phaseoli. Plant Soil 184:311–321<br />

Chapman SJ,Veal DA, Lynch JM (1992) Effect of oxygen concentration on dinitrogen fixation<br />

and volatile fatty acid production by Clostridium butyricum growing in association<br />

with fungi on cellulose and on wheat straw. J Appl Bacteriol 72:9–15<br />

Christensen BT, Johnston AE (1997) Soil organic matter and soil quality lessons learned<br />

from long-term experiments at Askov and Rothamsted. In: Gregorich EG, Carter MR<br />

(eds) Soil quality for crop production and ecosystem health. Elsevier, Amsterdam<br />

Cui M, Caldwell MM (1996) Facilitation of <strong>plant</strong> phosphate acquisition by arbuscular<br />

mycorrhizae from enriched to soil patches. I. Roots and hyphae exploiting the same<br />

volume. New Phytol 133:453–460<br />

Dahllöf I (2002) Molecular community analysis of microbial diversity. Curr Opin<br />

Biotechnol 13:213–217<br />

Debosz K, Rasmussen PH, Pedersen AR (1999) Temporal variations in microbial biomass<br />

C and cellulolytic enzyme activity in arable soils: effects of organic matter input.Appl<br />

Soil Ecol 13:209–218<br />

De Fede KL, Pannaccione DG, Sexstone AJ (2001) Characterisation of dilution enrichment<br />

cultures obtained from size-fractionated soil bacteria by BIOLOG (R) community<br />

level physiological profiles and restriction analysis of 16S rRNA genes. Soil Biol<br />

Biochem 33:1555–1562<br />

Degens BP (1998) Microbial functional diversity can be influenced by the addition of<br />

simple organic substrates to soil. Soil Biol Biochem 30:1981–1988<br />

Degens BP, Scipper LA. Sparling GP, Duncan LC (2001) Is the microbial community in a<br />

soil with reduced catabolic diversity less resistant to stress or disturbance? Soil Biol<br />

Biochem 33:1143–1153<br />

Diaz G, Azcon-Aguilar C, Honrubia M (1996) Influence of arbuscular mycorrhizae on<br />

heavy metal (Zn and Pb) uptake and growth of Lygeum spartum and Anthyllis cytisoides.<br />

Plant Soil 180:241–249


5 Diversity and Functions of Soil Microflora in Development of Plants 93<br />

Dommergues YR (1997) Contribution of actinorhizal <strong>plant</strong>s to tropical soil productivity<br />

and rehabilitation. Soil Biol Biochem 29:931–941<br />

Elliott ML, Broschat TK (2002) Effects of a microbial inoculant on <strong>plant</strong> growth and rhizosphere<br />

bacterial populations of container-grown <strong>plant</strong>s. Hort Technol 12:222–225<br />

Elsgaard L (2001) Ethylene turnover in soil, litter and sediment. Soil Biol Biochem<br />

33:249–252<br />

Ezawa T, Smith SE, Smith FA (2002) P metabolism and transport in AM fungi. Plant Soil<br />

244:221–230<br />

Facelli E, Facelli JM, Smith SE, Mclaughlin MJ (1999) Interactive effects of arbuscular<br />

mycorrhizal symbiosis intraspecific competition and resource availability on Trifolium<br />

subterraneum cv. Mt. Barker. New Phytol 141:535–547<br />

Faure S, Cliquet J-B, Thephany G, Boucaud J (1998) Nitrogen assimilation in Lolium<br />

perenne colonized by the arbuscular mycorrhizal fungus Glomus fasciculatum. New<br />

Phytol 138:411–417<br />

Fitter AH, Graves JD, Watkins NK, Robinson D, Scrimgeour C (1998) Carbon transfer<br />

between <strong>plant</strong>s and its control in networks of arbuscular mycorrhizas. Funct Ecol<br />

12:406–412<br />

Fortin JA, Becard G, Declerck S, Dalpe Y, St-Arnaud M, Coughlan AP, Piche Y (2002)<br />

Arbuscular mycorrhiza on root-organ cultures. Can J Bot 80:1–20<br />

Fracchia S, Garcia-Romera I, Godeas A, Ocampo JA (2000) Effect of the saprophytic fungus<br />

Fusarium oxysporum on arbuscular mycorrhizal colonization and growth of<br />

<strong>plant</strong>s in greenhouse and field trials. Plant Soil 223:175–184<br />

Francis R, Read DJ (1994) The contributions of mycorrhizal fungi to the determination<br />

of <strong>plant</strong> community structure. Plant Soil 159:11–25<br />

Frankenberger Jr WT, Arshad M (1995) Phytohormones in soils. Microbial production<br />

and function. Marcel Dekker, New York<br />

Frey B, Schüepp H (1993) A role of vesicular arbuscular (VA) mycorrhizal fungi in facilitating<br />

inter-<strong>plant</strong> N transfer. Soil Biol Biochem 25:651–658<br />

Glick BR (1995) The enhancement of <strong>plant</strong> growth by free-living bacteria. Can J Microbiol<br />

41:109–117<br />

Goenadi D, Siswanto H, Sugiarto Y (2000) Bioactivation of poorly soluble phosphate<br />

rocks with a phosphorus-solubilizing fungus. Soil Sci Soc Am J 64:927–932<br />

Goldstein AH (1995) Recent progress understanding the molecular genetics and biochemistry<br />

of calcium phosphate solubilization by gram negative bacteria. Biol Agric<br />

Hort 12:185–193<br />

George E, Marschner H, Jakobsen I (1995) Role for arbuscular mycorrhizal fungi in<br />

uptake of phosphorous and nitrogen from soil. Crit Rev Biotechnol 15:257–270<br />

Glick BR, Bashan Y (1997) Genetic manipulation of <strong>plant</strong> growth-promoting bacteria to<br />

enhance biocontrol of phytopathogens. Biotechnol Adv 15:353–378<br />

Goicoechea N, Antolin MC, Sanchez-Diaz M (2000) The role of <strong>plant</strong> size and nutrient<br />

concentrations in associations between Medicago, and Rhizobium and/or Glomus.<br />

Biol Plant 43:221–226<br />

Gyaneshwar P, Naresh Kumar G, Parekh LJ, Poole PS (2002) Role of soil microorganisms<br />

in improving P nutrition of <strong>plant</strong>s. Plant Soil 245:83–93<br />

Halsall DM, Gibson AH (1991) Nitrogenase activity (C 2 H 2 reduction) in straw-amended<br />

wheat belt soils in response to diazotroph inoculation. Soil Biol Biochem 23:987–998<br />

Hart TD, De Leij FAAM, Kinsey G, Kinsey G, Kelley J, Lynch JM (2002) Strategies for the<br />

isolation of cellulolytic fungi for composting of wheat straw. World J Microbiol<br />

Biotechnol 18:471–480<br />

Hattori T, Mitsui H, Haga H, Wakao N, Shikano S, Gorlach K, Kasahara Y, El-Beltagy A,<br />

Hattori R (1997) Advances in soil microbial ecology and the biodiversity Ant van<br />

Leeuwen Hoek 72:21–28


94<br />

Ramesh Chander Kuhad et al.<br />

Hawksworth DL (1997) The fascination of fungi: exploring fungal diversity. Mycologist<br />

11:18–22<br />

Helgason T, Daniell TJ, Husband R, Fitter AH,Young JPW (1998) Ploughing up the woodwide<br />

web. Nature 394:431<br />

Hodge A (2000) Microbial ecology of arbuscular mycorrhiza. FEMS Microbiol Ecol<br />

32:91–96<br />

Hodge A, Robinson D, Fitter AH (2000) An arbuscular mycorrhizal inoculum enhances<br />

root proliferation in, but not nitrogen capture from nutrient rich patches in soil. New<br />

Phytol 145:575–584<br />

Hooker JE, Black KE (1995) Arbuscular mycorrhizal fungi as a component of sustainable<br />

soil <strong>plant</strong> systems. Crit Rev Biotechnol 15:201–212<br />

Hunter-Cevera JC (1998) The value of microbial diversity. Curr Opin Microbiol<br />

1:278–285<br />

Jacobs H, Boswell GP, Ritz K, Davidson FA, Gadd GM (2002) Solubilization of calcium<br />

phosphate as a consequence of carbon translocation by Rhizobium solani. FEMS<br />

Microbiol Ecol 40:65–71<br />

Janardhanan KK, Abdul-Khaliq Naushin F, Ramaswamy K (1994) Vesicular arbusicular<br />

mycorrhiza in an alkaline usar land ecosystem. Curr Sci 67:465–469<br />

Jenesen SL, Mueller T, Magid J, Nielsen NE (1997) Temporal variations of C and N mineralization,<br />

microbial biomass and extractable organic pools in soil after oilseed rape<br />

straw incorporation in the field. Soil Biol Biochem 29:1043–1055<br />

Jensen LE, Nybroe O (1999) Nitrogen availability to Pseudomonas fluorescens DF57 is<br />

limited during decomposition of barley straw in bulk soil and in barley rhizosphere.<br />

Appl Environ Microbiol 65:4320–4328<br />

Johansen A, Finaly RD, Olsson PA (1996) Nitrogen metabolism of external hyphae of the<br />

arbuscular mycorrhizal fungus Glomus intaradices. New Phytol 133:705–712<br />

Jones DL (1998) Organic acids in the rhizosphere – a critical review. Plant Soil 205:25–44<br />

Kim KY, Jordan D, McDonald GA (1998) Enterobacter agglomerans, phosphate solubilizing<br />

bacteria and microbial activity in soil: effect of carbon sources. Soil Biol Biochem<br />

30:995–1003<br />

Knudsen IMB, Debosz K, Hockenhull J, Jensen DF, Emholt S (1999) Suppressiveness of<br />

organically and conventionally managed soils towards brown foot rot of barley. Appl<br />

Soil Ecol 12:61–72<br />

Koide RT, Dickie IA (2002) Effects of mycorrhizal fungi on <strong>plant</strong> populations. Plant Soil<br />

244:307–317<br />

Kozdroj J, van Elsas JD (2001) Structural diversity of microorganisms in chemically perturbed<br />

soil assessed by molecular and cytochemical approaches. J Microbiol Meth<br />

43:197–212<br />

Kumar PKR, Lonsane BK (1989) Microbial production of gibberellins: state of the art.<br />

Adv Appl Microbiol 34:29–139<br />

Kumar V, Narula N (1999) Solubilization of inorganic phosphates and growth emergence<br />

of wheat as affected by Azotobacter chroococcum mutants. Biol Fertil Soils 28:301–305<br />

Kumar A, Nivedita P, Upadhyaya RS (1999) VA mycorrhizae and revegetation of coal<br />

mine spoils: a review. Trop Ecol 40:1–10<br />

Kumar V, Singh KP (2001) Enriching vermicompost by nitrogen fixing and phosphate<br />

solubilizing bacteria. Biores Technol 76:173–175<br />

Kumari V, Srivastava JS (1999) Molecular and biochemical aspects of rhizobacterial ecology<br />

with emphasis on biological control. World J Microbiol Biotechnol 15:535–543<br />

Kuzyakov Y, Domanski G (2002) Carbon input by <strong>plant</strong>s into the soil. J Plant Nutr Soil Sci<br />

163:421–431<br />

Lechevalier MP (1994) Minireview: taxonomy of the genus Frankia (Actinomycetales).<br />

Int J Syst Bacteriol 44:1–8


5 Diversity and Functions of Soil Microflora in Development of Plants 95<br />

Loreau M (2001) Microbial diversity, producer-decomposer interactions and ecosystem<br />

processes: a theoretical model. Proc R Soc Biol Sci B 268:303–309<br />

Madder P,Vierheilig H, Boller T, Streitwolf-Engel B, Frey B, Christie P,Wiemken A. (2000)<br />

Transport of 15 N from a soil compartment separated by a polytetraflouroethylene<br />

membrane to <strong>plant</strong> roots via the hyphae of arbuscular mycorrhizal fungi. New Phytol<br />

146:155–161<br />

Martens DA, Frankenberger Jr WT (1993) Metabolism of tryptophan in soil. Soil Biol<br />

Biochem 25:1679–1687<br />

Maunuksela L (2001) Molecular and physiological characterization of rhizosphere bacteria<br />

and Frankia in forest soils devoid of actinorhizal <strong>plant</strong>s. Ph.D thesis. University<br />

of Helsinki, Helsinki, Finland<br />

Merryweather J, Fitter A (1995) Phosphorus and carbon budgets mycorrhizal contributions<br />

in Hyacinthoides nonscripta (L.) Chouard ex Rothm under natural conditions.<br />

New Phytol 129:619–627<br />

Miller RM, Jastrow JD (1994) Vesicular arbuscular mycorrhizae and <strong>plant</strong> biogeochemical<br />

cycling. In: Pfleger FL, Linderman RG (eds) Mycorrhizae and <strong>plant</strong> health, APS<br />

Press, St. Paul, USA, pp 189–212<br />

Myrold DD (2000) Microorganisms. In: Alexander DE, Fairbridge RW (eds) Encyclopedia<br />

of Environmental Science. Kluwer, Amsterdam, pp 409<br />

Nahas E (1996) Factors determining rock phosphate solubilization by microorganisms.<br />

World J Microbiol Biotechnol 112:567–572<br />

Narsian V, Patel HH (2000) Aspergillus aculeatus as a rock phosphate solubilizer. Soil Biol<br />

Biochem 32:559–565<br />

Naseby DC, Way JA, Bainton NJ, Lynch JM (2001) Biocontrol of Pythium in the pea rhizosphere<br />

by antifungal metabolite producing and non-producing Pseudomonas<br />

strains. J Appl Microbiol 90:421–429<br />

Nautiyal CS, Bhadauria S, Kumar P, Lal H, Mondal R, Verma D (2000) Stress induced<br />

phosphate solubilization in bacteria isolated from alkaline soils. FEMS Microbiol Lett<br />

182:291–296<br />

Neilands JB, Leong SA (1986) Siderophores in relation to <strong>plant</strong> growth and disease.Annu<br />

Rev Plant Physiol 37:187–208<br />

O’Donnell AG, Görres HE (1999) 16S rDNA methods in soil <strong>microbiology</strong>. Curr Opin<br />

Biotechnol 10:225–229<br />

Olesniewics KS, Thomas RB (1999) Effects of mycorrhizal colonization on biomass production<br />

and nitrogen fixation of black locust (Roina pseudoacacia) seedlings grown<br />

under elevated atmospheric carbon dioxide. New Phytol 142:133–140<br />

Omar SA (1994) Degradation and mineralization of wheat straw by some cellulolytic<br />

fungi in pure culture. Microbiol Res 149:157–161<br />

Pal SS (1998) Interaction of an acid tolerant strain of phosphate solubilizing bacteria<br />

with a few acid tolerant crops. Plant Soil 198:169–177<br />

Pankhurst CE, Pierret A, Hawke BG, Kirby JM (2002) Microbiological and chemical properties<br />

of soil associated with macrospores at different depths in a red-duplex soil in<br />

NSW Australia. Plant Soil 238:11–20<br />

Paul EA, Clark FE (1998) Soil <strong>microbiology</strong> and biochemistry, 2nd edn. Academic Press,<br />

San Diego<br />

Peix A, Rivas-Boyero AA, Mateos PF, Rodriguez-Barrueco C, Martinez-Molina E,<br />

Velazquez E (2001) Growth promotion of chickpea and barley by a phosphate solubilizing<br />

strain of Mesorhizobium and mediterraneum under growth chamber conditions.<br />

Soil Biol Biochem 33:103–110<br />

Perotto S, Girlanda M, Martino M (2002) Ericoid mycorrhizal fungi: some new perspectives<br />

on old acquaintances. Plant Soil 244:41–53


96<br />

Ramesh Chander Kuhad et al.<br />

Prosser JI (2002) Molecular and functional diversity in soil microorganisms. Plant Soil<br />

244:9–17<br />

Rajbanshi SS, Endo H, Sakamoto K, Inubushi K (1998) Stabilisation of chemical and biochemical<br />

characteristics of grass straw and leaf mix during in-vessel composting with<br />

and without seeding material. Soil Sci Plant Nutr 44:485–495<br />

Ranjard L, Richaume A (2001) Quantitative and qualitative microscale distribution of<br />

bacteria in soil. Res Microbiol 152:707–716<br />

Rasmussen PH, Knudsen IMB, Elmholt S, Jensen DF (2002) Relationship between soil<br />

cellulolytic activity and suppression of seedling blight of barley in arable soils. Appl<br />

Soil Ecol 19:91–96<br />

Read DJ (1996) The structure and function of the ericoid mycorrhizal root. Ann Bot<br />

77:365–374<br />

Reddy MS, Kumar S, Babita K, Reddy MS (2002) Biosolubilization of poorly soluble rock<br />

phosphates by Aspergillus tubingensis and Aspergillus niger. Biores Technol<br />

84:187–189<br />

Rennenberg H (1999) The significance of ectomycorrhizal fungi for sulfur nutrition of<br />

trees. Plant Soil 215:115–122<br />

Rengel Z (2002) Breeding for better symbiosis. Plant Soil 245:147–162<br />

Requena N, Azcon R, Baca MT (1996) Chemical changes in humic substances from compost<br />

due to incubation with lignocellulolytic microorganisms and effects on lettuce<br />

growth. Appl Microbiol Biotechnol 45:857–863<br />

Reyes I, Baziramakenga R, Bernier L,Antoun H (2001) Solubilization of phosphate rocks<br />

and minerals by a wild type strain and two UV-induced mutants of Penicillium rugulosum.<br />

Soil Biol Biochem 33:1741–1747<br />

Rice WA, Olsen PE, Legget ME (1995) Co-culture of Rhizobium meliloti and a phosphorus<br />

solubilizing fungus (Penicillium bilaii) in sterile peat. Soil Biol Biochem 27:<br />

703–705<br />

Ritz K, Wheatley RE, Griffiths BS (1997) Effects of animal manure application and crop<br />

<strong>plant</strong>s upon size and activity of soil microbial biomass under organically grown<br />

spring barley. Biol Fert Soils 24:372–377<br />

Schipper LA, Dehgens BP, Sparling GP, Duncan LC (2001) Changes in microbial heterotrophic<br />

diversity along five <strong>plant</strong> successional sequences. Soil Biol Biochem<br />

33:2093–2103<br />

Sen R (2000) Budgeting for the wood – wide web. New Phytol 145:161–165<br />

Sharples JM, Mehrag A, Chambers SM, Cairney JWG (2000) Mechanism of arsenate resistance<br />

in the ericoid mycorrhizal fungus Hymenoscyphus ericae. Plant Physiol<br />

124:1327–1334<br />

Shen D (1997) Microbial diversity and application of microbial products for agricultural<br />

purposes in China. Agric Ecosyst Environ 62:237–245<br />

Simon L (1996) Phylogeny of the Glomales: Deciphering the past to understand the present.<br />

New Phytol 133:95–101<br />

Singal R, Gupta R, Kuhad RC, Saxena RK (1991) Solubilization of inorganic phosphates<br />

by a Basidiomycetous fungus Cyathus. Ind J Microbiol 31:397–401<br />

Singh A, Sharma J, Rexer K-H, Varma A (2000) Plant productivity determinants beyond<br />

minerals, water and light: Piriformospora indica – a revolutionary <strong>plant</strong> growth-promoting<br />

fungus. Curr Sci 79:1548–1554<br />

Smit E, Leeflang P, Gommans S, van der Broek J, van MS, Werners K (2001) Diversity and<br />

seasonal fluctuations of the dominant members of the bacterial soil community in a<br />

wheat field as determined by cultivation and molecular methods. Appl Environ<br />

Microbiol 67:2284–2291<br />

Smith SE, Read DJ (1997) Growth and carbon economy of VA mycorrhizal <strong>plant</strong>s. In:<br />

Mycorrhizal symbiosis. 2nd edn. Academic Press, London, pp 105–125


5 Diversity and Functions of Soil Microflora in Development of Plants 97<br />

Staley JT (1997) Biodiversity: are microbial species threatened? Curr Opin Biotechnol<br />

8:340–345<br />

Steenhoudt O, Vanderleyden J (2000) Azospirrilum, a freeliving nitrogen fixing bacterium<br />

closely associated with grasses: genetic, biochemical and ecological aspects.<br />

FEMS Microbiol Rev 24:487–506<br />

Stepanov AL, Korpelal TK (1997) Microbial basis for the biotechnological removal of<br />

nitrogen oxides from flue gases. Biotechnol Appl Biochem 25:97–104<br />

Subba Rao NS (1999) Soil <strong>microbiology</strong>, 4th edn, Science Publishers, New Hampshire<br />

Suneja S, Lakshminarayana K (1999) Siderophore production of Azotobacter. In: Narula<br />

N (ed) Azotobacter in sustainable agriculture, CBS Publishers, New Delhi, pp 64–73<br />

Timmusk S, Nicander B, Granhall U, Tillberg E (1999) Cytokinin production by Paenibacillus<br />

polymyxa. Soil Biol Biochem 31:1847–1852<br />

Tiedje JM, Cho JC, Murray A, Travers D, Xia B, Zhou J (2001) Soil teeming with life: new<br />

frontiers for soil science. In: Rees RM, Ball BC, Campbell CD, Watson CA (eds) Sustainable<br />

management of soil organic matter, CAB International, London, pp 393–412<br />

Toro M,Azcon R, Barea J (1997) Improvement of arbuscular mycorrhiza development by<br />

inoculation of soil with phosphate solubilizing rhizobacteria to improve rock phosphate<br />

bioavailability (32P) and nutrient cycling. Appl Environ Microbiol 63:4408–<br />

4412<br />

Torrisi V, Pattinson GS, McGee PA (1999) Localized elongation of roots of cotton follows<br />

establishment of arbuscular mycorrhizas. New Phytol 142:103–112<br />

Torsvik V, Øvreås L (2002) Microbial diversity and function in soil: from genes to ecosystems.<br />

Curr Opin Microbiol 5:240–245<br />

Torsvik V, Sorheim R, Goksoyr J (1996) Total bacterial diversity in soil and sediment<br />

communities – a review. J Ind Mcirobiol 17:170–178<br />

Unge A, Jansson J (2001) Monitoring population size, activity, and distribution of gfpluxAB-tagged<br />

Pseudomonas fluorescence SBW25 during colonization of wheat.<br />

Microb Ecol 41:290–300<br />

Valdenegro M, Barea JM,Azcon R (2001) Influence of arbuscular-mycorrhizal fungi, Rhizobium<br />

meliloti strains and PGPR inoculation on the growth of Medicago arborea<br />

used as nodel legume for revegetation and biological reactivation in a semi-arid<br />

mediterranean area. Plant Growth Regul 34:233–240<br />

Varma A (1995) Ecophysiology of mycorrhizal fungi. In: Varma A, Hock B (eds) Mycorrhizae:<br />

structure, function, molecular biology and biotechnology, Springer-Verlag,<br />

Berlin Heidelberg New York, pp 561–592<br />

Varma A, Bonfante P (1994) Utilization of cell wall related carbohydrates by ericoid mycorrhizal<br />

endophytes. Symbiosis 16:301–313<br />

Varma A, Verma S, Sudha A, Sahay N, Butehorn B, Franken P (1999) Piriformospora<br />

indica, a cultivable <strong>plant</strong>-growth-promoting root endophyte. Appl Environ Microbiol<br />

65:2741–2744<br />

Vassilev N, Franco I,Vassileva M,Azcon R (1996) Improved <strong>plant</strong> growth with rock phosphate<br />

solubilized by Aspergillus niger grown on sugar beet waste. Biores Technol<br />

55:237–241<br />

Vassilev N,Vassileva M, Fenice M, Fedirici F (2001) Immobilized cell technology applied<br />

in solubilization of insoluble inorganic (rock) phosphates and P <strong>plant</strong> acquisition.<br />

Biores Technol 79:263–271<br />

Verghese SK, Sarma G, Chauhan V, Misra AK (1998) Optimising actinorhizal symbiosis<br />

using molecular markers. In: Rai B, Dkahr MS (eds) New trends in microbial ecology.<br />

NEH University, Shilong and International Society for Conservation of Natural<br />

Resources, BHU,Varanasi, India, pp 350–354<br />

Verstaete W, Top EM (1999) Soil clean up: lessons to remember. Int Biodeter Biodeg<br />

43:147–153


98<br />

Ramesh Chander Kuhad et al.<br />

Vierheilig H, Bennett R, Kiddle G, Kaldorf M, Ludwig-Muller J (2000) Differences in glucosionolate<br />

patterns and arbuscular mycorrhizal status of glucosionolate containing<br />

species. New Phytol 146:343–352<br />

Watkins NK, Fitter AH, Graves JD, Robinson D (1996) Carbon transfer between C3 and<br />

C4 <strong>plant</strong>s linked by a common mycorrhizal network, quantified using stable carbon<br />

isotopes. Soil Biol Biochem 28:471–477<br />

Whitelaw MA, Harden TJ, Helyar KR (1999) Phosphate solubilization in solution culture<br />

by the soil fungus Penicillium radicum. Soil Biol Biochem 31:655–665<br />

Wiemken V, Boller T (2002) Ectomycorrhiza: gene expression, metabolism and the<br />

wood-wide web. Curr Opin Plant Biol 5:1–7<br />

Yin B, Crowley D, Sparovek G, De Melo WJ, Borneman J (2000) Bacterial functional<br />

redundancy along a soil reclamation gradient. Appl Environ Microbiol 66:4361–4365<br />

Zehnder GW, Murphy JF, Sikora EJ, Kloepper JW (2001) Application of rhizobacteria for<br />

induced resistance. Eur J Plant Pathol 107:39–50<br />

Zhu W, Ehrenfeld JG (1996) The effects of mycorrhizal roots on litter decomposition, soil<br />

biota and nutrients in a spodsolic soil. Plant Soil 179: 109–118


6 Signalling in the Rhizobia–Legumes Symbiosis<br />

Dietrich Werner<br />

1 Introduction<br />

During the last few years, significant progress has been made in the understanding<br />

of signal production, signal perception and signal regulation in<br />

<strong>plant</strong>s, microorganisms and animals. In mammalian systems, the notch signal<br />

regulation has been studied intensively with a number of proteins involved in<br />

the signal transport and the proteolytic modifications of the notch signals<br />

(Fig. 1). The involvement of several organelles such as lysosomes, endoplasmic<br />

reticulum and the Golgi network points to interesting similarities and<br />

differences to the signalling across and through the symbiosome membrane<br />

(Werner 1992). Four different mammalian notch homologues have been identified<br />

(Baron et al. 2002). The integrin-adhesion-receptor signalling is another<br />

<strong>surface</strong> related crosstalk in multicellular organisms (Schwartz and Ginsberg<br />

2002). The cell adhesion involving integrins leads to a phosphorylation of different<br />

growth-factor receptors, including those for the fibroblast growth factor,<br />

the hepatocyte growth factor and the epidermal growth factor (Giancotti<br />

and Ruoslahti 1999).Very recently, a <strong>plant</strong> receptor-like kinase has been identified<br />

in the laboratory of Martin Parniske, The Sainsbury Laboratory, UK,<br />

which is required for the rhizobial legume symbiosis as well as for the arbuscular<br />

mycorrhiza symbiosis (Stracke et al. 2002). The SYMRK (symbiosis<br />

receptor-like kinase) genes have been studied and characterized in Lotus and<br />

in pea. The protein has a signal peptide, a transmembrane and an extracellular<br />

protein kinase domain. The SYMRK is part of a symbiotic signal transduction<br />

pathway with the perception of a microbial signal molecule, leading<br />

to a rapid symbiosis-related gene activation. In Medicago sativa,a “nodulation<br />

receptor kinase” NORK was identified with a predicted function in Nod-factor<br />

perception/transduction (Endre et al. 2002).<br />

Besides the short-distance signalling between microorganisms and <strong>plant</strong><br />

<strong>surface</strong>s, long-distance signalling also affects the <strong>plant</strong> partner of the interaction.<br />

Using mutants of Arabidopsis, the role for long-chain fatty acids in cellto-cell<br />

communication has been established and the <strong>plant</strong> hormones abscisic<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


100<br />

Dietrich Werner<br />

Fig. 1. Notch trafficking and signal regulation. Trafficking of Notch through the cell in<br />

conjunction with covalent modifications of Notch may confer potential points of regulation<br />

of Notch-signalling levels. Proteins known or suspected to affect particular steps in<br />

the transport process or the proteolytic modification of Notch are indicated in bold.<br />

Numbers indicate the potential decision points that could determine signal levels. 1<br />

Notch trafficking to the Golgi body may be subject to quality control and cargo selection<br />

by P24 family proteins. 2 Small GTPases such as Rab6 may regulate transport to trans-<br />

Golgi where Furin-dependent proteolysis occurs. 3 Notch transported to the cell <strong>surface</strong><br />

may undergo endocytosis (4), or 5 ligand-dependent activation at the cell <strong>surface</strong>. The<br />

latter is accompanied at least in some tissues by 6 trans-endocytosis of the Notch extracellular<br />

domain into the adjacent ligand-bearing cell. 7 The remaining membrane-tethered<br />

intracellular domain undergoes Presenilin-dependent cleavage releasing Notchintra<br />

for translocation to the nucleus leading to regulation to target gene expression. 8<br />

Accumulation of Notch-intra in the nucleus may be regulated by its ubiquitination and<br />

proteosome-dependent degradation (Baron et al. 2002)


acid, ethylene and jasmonates are involved in the long-distance signalling,<br />

regulating, e.g., the stomata number by environmental signals such as the CO 2<br />

partial pressure (Lake et al. 2002). Other long-distance signals, e.g., from roots<br />

to shoots, are transported in the xylem. This roots to shoots signalling has a<br />

lag time of only a few hours, persisting for several days (Jackson 2002).<br />

Abscisic acid (ABA) plays an important role in the signalling from the roots<br />

and the rhizosphere, e.g., in water stress for the regulation of stomata behavior<br />

(Wilkinson and Davies 2002). The signalling process includes the following<br />

steps: ABA sequestration in the root, ABA synthesis and catabolism in the<br />

root, the transfer of ABA across the root and into the xylem, the exchange of<br />

ABA from the xylem lumen to the xylem parenchyma in the shoot, the concentration<br />

of ABA in the leaf symplastic reservoir, the cleavage of ABA conjugates,<br />

the transfer of ABA from the leaf into the phloem and an assumed interaction<br />

between nitrate stress and the ABA signal.<br />

Moreover, in unicellular eukaryotic model organisms specific signal molecules<br />

have been identified, such as phosphatidylinositol (3,5) bisphosphate<br />

modified by the phosphatidylinositol 3-phosphate 5-kinase in Schizosacchromyces<br />

pombe and which are required to respond to nutritional starvation<br />

(Morishita et al. 2002). This pathway is also necessary for mating the<br />

pheromone signal in this yeast. The question arises if these compounds also<br />

play a role in microbes – <strong>plant</strong> <strong>surface</strong> signalling. The costs for biological<br />

signalling are an important aspect in evolution. New results indicate that<br />

there is a shift to signals with high cost and an underutilization of signals<br />

with low costs (de Polavieja 2002). The signal structure follows a generalized<br />

Boltzmann form, penalizing signals with high costs and a high sensitivity to<br />

errors. In this respect, costs are defined in metabolic costs, time costs or risk<br />

costs.<br />

In bacteria key components for major regulatory pathways have been identified<br />

such as the protein H-NS (Schröder and Wagner 2002). It is a small DNA<br />

binding protein, regulating a diverse range of genes such as for anaerobic<br />

growth phase activation, endochitinase, nitrate reductase, leucine responsive<br />

regulatory proteins, a proline/glycine betaine transport system, an activator<br />

for capsular polysaccharide synthesis and an invasion regulatory gene.<br />

2 The Signals from the Host Plants<br />

6 Signalling in the Rhizobia–Legumes Symbioses 101<br />

In principle, molecules released from <strong>plant</strong> <strong>surface</strong>s can be substrates and signals<br />

for microorganisms. Types and functions of root exudates in the rhizosphere<br />

have recently been reviewed by Brimecombe et al. (2001), Neumann<br />

and Römheld (2001) and Uren (2001). Their functions as signal molecules<br />

have been reviewed by Werner (2001) and Werner and Müller (2002).


102<br />

Dietrich Werner<br />

2.1 Phenylpropanoids: Simple Phenolics, Flavonoids and Isoflavonoids<br />

The basis of this biochemical pathway is the shikimate pathway, producing<br />

aromatic amino acids and several vitamins and co-factors. The phenylalanine-ammonia-lyase<br />

(PAL) produces cinnamate from phenylalanine, which is<br />

a precursor of a large number of phenolics, phenylpropanoids and flavonoids<br />

(Paiva 2000). Major cinnamate derivatives are para-coumaric acid, caffeic<br />

acid, ferulic acid and sinapic acid, leading to <strong>plant</strong> <strong>surface</strong> polymers such as<br />

suberin and lignin. The ortho-hydroxylation of cinnamate gives coumarate, a<br />

precursor of coumarin, which has a strong antimicrobial activity. Another<br />

derivate is salicylic acid, which has also antimicrobial activity and a function<br />

in signal transduction during <strong>plant</strong> pathogenic interactions. Especially well<br />

studied is the function of acetosyringone, inducing vir gene expression in<br />

Agrobacterium tumefaciens, with a broad range of host <strong>plant</strong>s. Dimers of<br />

phenylpropanoids form lignans such as the antifungal compound magnolol<br />

and the toxic compound podophyllotoxin. The biosynthesis of major classes<br />

of flavonoids and isoflavonoids are summarized in Fig. 2, according to Dixon<br />

and Steele (1999). Major enzymes involved are chalcone synthase, chalcone<br />

reductase, chalcone isomerase, flavone synthase I and II, flavonol synthase,<br />

isoflavone synthase and flavonoid 3¢-hydroxylase. The major nod genes<br />

inducing compounds in legumes are isoflavones such as daidzein (R1=H) and<br />

genistein (R1=OH). The pterocarpan at the end of this pathway includes phytoalexins<br />

such as medicarpin from alfalfa and glyceollin I from soybeans. In<br />

addition to the intensively studied effects of flavonoids and isoflavonoids in<br />

the interaction of <strong>plant</strong>s with microorganisms, the health-promoting effects<br />

in medical sciences are another intensively studied area. Genistein, for<br />

instance, has been shown in human cell lines to inhibit prostate tumor<br />

growth, stomach tumor growth and anti-angiogenesis (Rice-Evans and Miller<br />

1996; Hollman and Katan 1998). Genistein has also an effect in preventing<br />

bone-loss caused by deficiency of estrogens in female mice (Ishimi et al.<br />

1999).<br />

Fig. 2. The biosynthesis of the major classes of flavonoid derivates. The enzymes are:<br />

CHS chalcone synthase, CHR chalcone reductase, CHI chalcone isomerase, FSI flavone<br />

synthase I, FSII flavone synthase II, ‘IFS’ isoflavone synthase, consisting of 2-hydroxyisoflavanone<br />

synthase (2HIS) and 2-hydroxyisoflavanone dehydratase (2-HID), F3bH<br />

flavanone 3b hydroxylase, F3¢H flavonoid 3¢hydroxylase, F3¢5H flavonoid 3¢,5¢-hydroxy-


6 Signalling in the Rhizobia–Legumes Symbioses 103<br />

lase, DFR dihydroflavonol reductase, ANS anthocyanidin synthase, 3GT anthocyanidin<br />

3-glucosyltransferase, IOMT isoflavone O-methyltransferase, IFR isoflavone reductase,<br />

VR vestitone reductase, DMID 7,2¢-dihydroxy, 4¢-methoxyisoflavanol dehydratase.<br />

Enzymes in white are 2-oxoglutarate-dependent dioxygenases, in black bold are<br />

cytochrome P450s, and highlighted in grey are NADPH-dependent reductases. Simplifications<br />

include not discriminating between the 5-hydroxy (R1=OH) and 5-deoxy<br />

(R1=H) flavonoids and isoflavonoids, for which the loss of the 5-hydroxyl occurs<br />

because of the co-action of CHR with CHS, showing only the anthocyanin pathway leading<br />

to the compounds with a di-substituted B-ring (cyaniding derivatives). Parallel pathways<br />

function in the formation of anthocyanins with mono- and tri-substituted B-rings.<br />

In the latter, F3¢5¢H can act at the level of the dihydroflavonol with a mono-or di-substituted<br />

B-ring. The pathway to epicatechin from a dihydroflavonol is shown to follow two<br />

routes, both via leucocyanidin. It is unclear whether there is a specific form of DFR that<br />

functions only in condensed tannin biosynthesis. The 4¢-O-methylation of the B-ring of<br />

isolflavones occurs in alfalfa, pea and other legumes, but not in bean or soybean (Dixon<br />

and Steele 1999)


104<br />

Dietrich Werner<br />

2.2 Metabolization of Flavonoids and Isoflavonoids<br />

A degradation pathway for luteolin by Sinorhizobium meliloti and for<br />

daidzein by Bradyrhizobium japonicum is shown in Fig. 3. Identified metabolites<br />

from luteolin were caffeic acid, phoroglucinol, protocatechuic acid and<br />

phenylacetic acid. Daidzein, p-coumaric acid, p-hydroxybenzoic acid, phenylacetic<br />

acid and resorcinol were major metabolites. Umbeliferone has also<br />

been found to be produced from coumestrol (Cooper et al. 1995).<br />

Fig. 3. Proposed degradation pathway for luteolin by Sinorhizobium meliloti and for<br />

daidzein by Bradyrhizobium japonicum (Cooper et al. 1995)


6 Signalling in the Rhizobia–Legumes Symbioses 105<br />

Flavanone metabolites from animals and humans are summarized in<br />

Table 1 (Heilmann and Merfort 1998).A number of flavanes, flavanoles, trans-<br />

3-hydroxyflavanes, 6-hydroxyflavanones, 6-hydroxyflavanes, 4-hydroxyflavanes,<br />

3,6-dihydroxy-flavanes, 3,4-dihydroxy-flavanes and methoxyflavanes<br />

have been identified. The best studied flavane is catechin. The major metabolites<br />

of this compound in humans are 3-hydroxybenzoic acid, 3-hydroxyphenylpropionic<br />

acid, 3-hydroxyhippuric acid, 3,4-dihydroxyphenylbenzoic<br />

acid, 5-(3,4-dihydroxy)-valerianic acid and d-(3-hydroxy-4-methoxyphenyl)g-valerolactone.<br />

Metabolites excreted from mammalians can, in certain locations<br />

of the soil, therefore also significantly increase the concentrations of<br />

flavonoid and isoflavonoid metabolites which may affect the <strong>plant</strong>-microbe<br />

interactions. Sulfation of flavonoids and phenolic dietary compounds by<br />

cytosolic sulfotransferases has been studied in detail by Pai et al. (2001). The<br />

mechanisms for a chemoprotective action of these compounds is the inhibition<br />

of the bioactivation of carcinogens by the human cytosolic sulfotransferases.<br />

The ten known cytosolic sulfotransferases have a different substrate<br />

specificity, e.g., the isoform PST has a high activity with flavonoids, but no<br />

activity with isoflavonoids (Pai et al. 2001).<br />

A fluoroimmunoassay has been developed to detect small concentrations<br />

of daidzein and genistein as phytoestrogens in blood plasma. After synthesis<br />

of 4¢-O-carboxymethyl-daidzein and 4¢-O-carboxymethyl-genistein, these<br />

compounds were linked to bovine serum albumin and used to immunize rabbits.<br />

The antisera were cross-reactive with some isoflavonoids, but not with<br />

flavonoids (Wang et al. 2000). The assays could detect daidzein and genistein<br />

in the range between 1 and 370 nMol/l. The actual concentrations in the blood<br />

plasma were in the range between 4 and 7 nMol/l. The correlation coefficient<br />

between this fluoroimmunoassay and a reference method, using an isotope<br />

dilution gaschromatography mass spectrometry, was in the range of 0.95–<br />

0.99. Another method to detect low concentrations of estrogenic flavonoids<br />

was the development of a recombinant yeast strain in which the human estrogen<br />

receptor was stably integrated into the genome. The most active<br />

flavonoids in this assay were naringenin, apigenin, kaempferol, phloretin,<br />

Table 1. Metabolites from flavanones (Heilmann and Merfort 1998)<br />

Flavane-4-a-ol 4¢-hydroxyflavane<br />

Flavane-4-b-ol 4¢-hydroxyflavane-4-a-ol<br />

Trans-3-hydroxyflavanone 4¢- hydroxyflavane-4-b-ol<br />

Trans-3-hydroxyflavane-4-a-ol 3,6-dihydroxy-flavane-4-a-ol<br />

Trans-3-hydroxyflavane-4-b-ol 3,6-dihydroxy-flavane-4-b-ol<br />

Flavone-3-ol 3,4-dihydroxy-flavane-4-a-ol<br />

6-Hydroxyflavanone 3,4-dihydroxy-flavane-4-b-ol<br />

6-Hydroxyflavane-4-a-ol 4¢-hydroxy-3¢-methoxyflavane-4-a-ol<br />

6-Hydroxyflavane-4-b-ol 4¢-hydroxy-3¢-methoxyflavane-4-b-ol


106<br />

Dietrich Werner<br />

equol, genistein, daidzein and biochanin A. The main feature for the estrogenic<br />

activity in these compounds is a single hydroxyl group at the 4¢-position<br />

of the B-ring of the flavan nucleus. It must be pointed out that the estrogenic<br />

activity of these flavonoids is 4,000–4,000,000 times lower than that of 17bestradiol<br />

(Breinholt and Larsen 1998). In the Handbook of Flavonoids (Harborne<br />

1994), about 900 different isoflavones, chalcones, pterocarpans, Bflavonoids,<br />

rotenoids, isoflavanes and coumestrols are listed. Nevertheless,<br />

new flavonoids can be found by studying less known legumes. In Ulex airensis<br />

and Ulex europaeus three new isoflavonoids called ulexin C, ulexin D and<br />

7-O-methylisolupalbigenin could be isolated and characterized (Maximo et<br />

al. 2002).<br />

Another important group of <strong>plant</strong> signals involved in symbiosis and<br />

defense reactions are fatty acid-derived signals (Weber 2002). The best studied<br />

compound in this category is jasmonic acid and its volatile methyl ester.<br />

Almost 20 different jasmonate signalling mutants in Arabidopsis are known<br />

(Staswick et al. 1998; Thomma et al. 1998; Hilpert et al. 2001). With Arabidopsis<br />

it has also been shown that keto, hydroxy and hydroperoxy fatty acids such<br />

as ketodienoic fatty acid can accumulate in <strong>plant</strong>s after infection with<br />

Pseudomonas syringae. By infiltration of these compounds into Arabidopsis<br />

leaves, the gene encoding glutathione-S-transferase (GST1) has been demonstrated<br />

(Weber 2002). From a large number of flavonoids and isoflavonoids<br />

tested for their ability to inhibit the ascorbate-induced microsomal lipid peroxidation,<br />

kaempferol has been showed to have the highest activity of all<br />

flavonoids tested (Cos et al. 2001).<br />

2.3 Vitamins as Growth Factors and Signal Molecules<br />

On a molecular basis the best-studied system is the effect of biotin on growth<br />

and survival of Sinorhizobium meliloti. Already, nanomolar concentrations of<br />

biotin increase the colonization of alfalfa roots sevenfold and addition of<br />

avidin, a biotin-binding protein from eggs, reduces the colonization by a factor<br />

of 7. Biotin is a co-factor of carboxylation reactions and biotinylated carboxylases<br />

have been demonstrated in Rhizobium etli (Dunn 1996). From the<br />

biotin biosynthesis pathway, several genes such as bioA, bioC and bioH are<br />

apparently not functional and also for bioD, no homology genes have been<br />

found (Entcheva et al. 2001). On the other hand, components of a prokaryotic<br />

biotin transporter have been identified with bioMN, which are activated<br />

under biotin deficiency. Rhizobia have a very efficient uptake system for<br />

biotin. The kM values are 40 times lower than those for the transport system<br />

for E. coli. With biotin limitation the synthesis of PHB is significantly<br />

increased in Sinorhizobium meliloti and Rhizobium etli. Under biotin limitation<br />

the transcription rate of the gene, responsible for proteins of PHB degradation<br />

is down-regulated. Addition of biotin at nanomolar concentrations


increases the activity of the bdhA gene more than fivefold, as demonstrated<br />

for strains with a lac-Z-fusion (Streit et al. 1996; Hofman et al. 2000).<br />

Another very interesting signal molecule derived from a vitamin is<br />

lumichrome, which is produced from riboflavin and functions as a signal<br />

molecule in the rhizosphere (Philipps et al. 1999). Nanomolar concentrations<br />

of lumichrome around the roots increase the root respiration and also photosynthesis.<br />

This may be of general significance since a number of riboflavinproducing<br />

bacteria have been identified on roots in a significantly higher<br />

number than in the bulk soil (Strzelczyk and Rozycki 1985).<br />

3 Signals from the Microsymbionts<br />

3.1 Nod Factors<br />

6 Signalling in the Rhizobia–Legumes Symbioses 107<br />

A breakthrough in understanding infection, nodulation and host specificity<br />

in the Rhizobium-legume symbiosis was the identification of Nod factors<br />

such as lipochitooligosaccharides (LCOs) produced and excreted by more<br />

than 30 different nod, nol and noe genes and their corresponding proteins<br />

from the microsymbionts. The first identified Nod factors were from Rhizobium<br />

meliloti (Lerouge et al. 1990) and from Rhizobium leguminosarum bv.<br />

viciae (Spaink et al. 1991). The general structure of Nod factors are N-acetylglucosamine<br />

backbone with four or five GlcNAc residues with different substituents<br />

at nine different positions such as N-methyl, O-carbamyl, O-acetyl,<br />

O-sulfyl, a-linked fucosyl, 2-O-methylfucosyl, 4-O-acetyl-2-O-methylfucosyl,<br />

3-O-sulfate-2-O-methylfucosyl, ethyl, glyceryl, mannosyl and N-glycosyl<br />

groups. Another major residue variable is the fatty acid group attached to the<br />

nitrogen of the nonreducing end of the Nod factor. Fatty acids with 16–18 carbons<br />

and a different degree of unsaturation in different positions of the double<br />

bonds are mainly present. In addition, C18–C22 (w-1)-hydroxy fatty acids,<br />

Fig. 4. General structure of the Nod factors produced by rhizobia. The presence of substituents<br />

numbered R1–R9 is variable within various strains of rhizobia. In the absence<br />

of specific substituents, the R groups stand for hydrogen (R1), hydroxy (R2, R3, R4, R5,<br />

R6, R8, and R9), and acetyl (R7) (Spaink 2000)


108<br />

Dietrich Werner<br />

Table 2. Modifications of Nod factors (Modified from Spaink 2000 and Pacios-Bras et al.<br />

2002) a<br />

Bacterial strain Nodulated GlcNAc Special substituents c<br />

<strong>plant</strong> residues<br />

tribes (n) b<br />

S. meliloti Galegeae 3,4,5 R4:Ac, R5:S, C16:2, C16:3, C26(w-1)OH<br />

R. leguminosarum<br />

bv. Viciae RBL5560 Galegeae 3,4,5 R4:Ac, C18:4<br />

bv. Viciae TOM Galegeae 3,4,5 R4:Ac, R5:Ac, C18:4<br />

bv. Viciae A1 Galegeae 3,4,5 R4:Ac, R5:Ac, C18:4, C18:3<br />

bv. Trifolii ANU842 Galegeae 3,4,5 R4:AC, R5:Ac, R6:Et, C20:4, C20:3, C18:3<br />

R. galegae Galegeae 4,5 R4:Cb, R9:Ac, C18:2, C18:3, C20:2, C20:3<br />

M. huakuii Galegeae 3,4,5 R5:S, R7:G, C18:4<br />

M. loti<br />

E1R, NZP2235 Loteae 4,5 R1:Me, R3:Cb, R5:AcFuc<br />

NZP2037 Loteae<br />

Genisteae 4,5 R1:Me, R2:Cb, R3:Cb, R5:AcFuc<br />

NZP2213 Loteae 2,3,4,5 R1:Me, R3:Cb, R5:AcFuc, R9:Fuc<br />

B. aspalati bv. carnosa Crotalarieae 3,4,5 R1:Me, R3:Cb, R4:Cb<br />

B. japonicum USDA110 Phaseoleae 5 R5:MeFuc<br />

B. japonicum USDA135 Phaseoleae 5 R4:Ac, R5:MeFuc<br />

B. elkanii USDA61 Phaseoleae 4,5 R1:Me, R4:Ac, R3:Cb, R5:MeFuc, R6:Gro<br />

R. etli Phaseoleae 4,5 R1:Me, R3:Cb, R5:AcFuc<br />

R. etli KIM5S Phaseoleae 6 R1:Me, R2-R6:H, R7:acetyl, R8:H, ring<br />

5:acetyl or H<br />

R. tropici Phaseoleae, 4,5 R1:Me, R5:S, R6:Man<br />

Mimoseae<br />

S. fredii<br />

USDA257 23 Tribes 3, 4,5 R5:MeFuc<br />

NGR234 26 Tribes 4,5 R1:Me, R3:Cb, R4:Cb, R5:MeFuc/AcMe-<br />

Fuc/SmeFuc<br />

Rhizobium sp. GRH2 Acacieae 3,5,6 R1:Me, R5:S<br />

S. teranga bv. acaciae Acacieae 5 R1:Me, R3/4:Cb, R5:S<br />

Mesorhizobium ORS1001 Acacieae 5 R1:Me, R3/4:Cb, R5:S<br />

A. caulinodans Robinieae 4,5 R1:Me, R4:Cb, R5:Fuc, R8:Ara<br />

S. sahelii Robinieae 4,5 R1:Me, R3/4:Cb, R5:Fuc, R8:Ara<br />

S. teranga bv. sesbaniae Robinieae 4,5 R1:Me, R3/4:Cb, R5:Fuc, R8:Ara<br />

a For backbone structure, see Fig. 4<br />

b The underlined numbers of N-acetylglucosamine (GlcNAc) residues indicate the most abundant<br />

species<br />

c The indicated substituents do not always occur in all lipochitin oligosaccharides (LCOs) produced,<br />

leading to a mixture of LCOs, which do or do not contain all possible substituents.<br />

Abbreviations: Me, N-methyl; Cb, O-carbamyl; Ac, O-acetyl; S, O-sulfyl; Fuc, a-linked fucosyl;<br />

MeFuc, 2-O-methylfucosyl, AcMeFuc, 4-O-acetyl-2- O-methylfucosyl; SmeFuc, 3-O-sulfate-2-Omethylfucosyl;<br />

Et, ethyl, Gro, glyceryl, Man, mannosyl; G, N-glycolyl; FA, fatty acyl


which are perhaps intermediates in the synthesis of C23 (w-1) hydroxy fatty<br />

acyl groups in lipopolysaccharides, can be present in Nod factors of Sinorhizobium<br />

meliloti (Demont et al. 1994). Figure 4 and Table 2 summarize the<br />

large variations of Nod factors identified so far. A novel lipochitin oligosaccharide<br />

has recently been found in Rhizobium etli KIM5S (Pacios-Bras et al.<br />

2002). This is the first case where the major LCO contains six oligosaccharide<br />

residues and differs by this point from all other rhizobia analyzed so far. An<br />

additional specificity was that the chitin backbone was deacetylated in one or<br />

two of the GlcNAc moieties, although these were only minor compounds. The<br />

fatty acids of these Nod factors were C16:0, C16:1, C18:0, C18:1 and C17:1. In<br />

this respect, the fatty acids are much more variable than those of Rhizobium<br />

etli strain CE3. Moreover, the host range of strain KIM5S of Rhizobium etli<br />

was different from the Rhizobium type strain CE3, since it could not nodulate<br />

Lotus japonicus, although it did nodulate Siratro.In Sinorhizobium meliloti it<br />

has been shown that an enzymatic N-deacetylation of the Nod factors<br />

decreases their biological activity, but increases the stability in the rhizosphere<br />

(Staehelin et al. 2000).<br />

In all rhizobia the nodABC genes are essential for the synthesis of the core<br />

LCO: NodC synthesizes the chito-oligosaccharide backbone and nodB<br />

removes N-acetyl groups from the sugar at its nonreducing end.All other nod,<br />

nol and noe genes are responsible for the modifications of this general structure,<br />

as indicated in Table 2. NodD is a positive transcription regulator from<br />

the Lysr family and present in all rhizobia. In some rhizobial species such as<br />

Sinorhizobium meliloti, nodD genes are present in multiple forms and their<br />

proteins respond to different groups of flavonoids. NodG has the enzymatic<br />

activity of an 3-oxoacyl-acyl carrier protein reductase and is thereby homologous<br />

to FabG involved generally in fatty acid elongation (López-Lara and<br />

Geiger 2001).<br />

3.2 Cyclic Glucans<br />

6 Signalling in the Rhizobia–Legumes Symbioses 109<br />

Cyclic glucans in rhizobia are small molecules linked either by b-(1,2) glycosidic<br />

bonds with 17–40 units in Rhizobium and Sinorhizobium or by b-(1,3)<br />

and b-(1,6) glycosidic bonds in Bradyrhizobium japonicum. Dominant substituents<br />

can be either sn-1-phosphoglycerol (Breedveld and Miller 1998) or<br />

phosphocholine (Rolin et al. 1992). The function of the cyclic glucans in Rhizobium,<br />

Sinorhizobium and Bradyrhizobium is to protect against hypoosmotic<br />

conditions. Rhizobia also produce, however, large quantities of cyclic<br />

glucans in the endosymbiotic stage. A specific function during this stage is<br />

assumed to be an increase in the solubility of flavonoids and Nod factors<br />

(Morris et al. 1991; Schlaman et al. 1997). Another hypothesis is, that b-glucans<br />

play a decisive role in the suppression of the host <strong>plant</strong> defense response<br />

with rhizobia, compared to phytopathogenic bacteria.


110<br />

Dietrich Werner<br />

3.3 Lipopolysaccharides<br />

Lipopolysaccharides (LPS) of rhizobia have been studied in only a few species<br />

such as Rhizobium etli and Rhizobium trifolii. The structure contains three<br />

parts, the lipid A, the core chain and the repeat unit of the O-antigen chain.All<br />

three parts are very variable. Typical features of rhizobial LPS are the very<br />

long chain hydroxy fatty acids (Hollingsworth and Carlson 1989). The genes<br />

of the LPS core and O-antigen synthesis have been localized on a plasmid<br />

(Vinuesa et al. 1999). A mutation in a glycosyltransferase produced a rough<br />

colony phenotype with a disruption of the O-antigen biosynthesis.<br />

The LPS in rhizobia may be involved in the infection process (Brewin 1998;<br />

Kannenberg et al. 1998). Their function is perhaps not in the first stages of<br />

symbiosis development, but in the release of the bacteria from the infection<br />

thread, and the first steps of the symbiosome membrane development. For the<br />

LPS moreover, a function in the suppression of the host <strong>plant</strong> defense<br />

response has been assumed, comparable to the LPS functions in <strong>plant</strong><br />

pathogens (Schoonejans et al. 1987).<br />

3.4 Exopolysaccharides<br />

The exopolysaccharides (EPS) have been studied in detail by a large number<br />

of rhizobial strains (Becker and Pühler 1998; Becker et al. 1998; Van Workum<br />

and Kijne 1998). In Sinorhizobium meliloti two types of EPS forms could be<br />

discriminated, EPS I as a succinoglucan and EPS II as a galactoglucan with<br />

two size classes in each form, one with thousands of saccharide units and a<br />

low-molecular-weight class with only 8–40 saccharide units. All genes<br />

involved in the biosynthesis of the repeating units have been identified, especially<br />

in the laboratory of Alf Pühler.<br />

Exopolysaccharides play a major role in the primary stage of the infection<br />

of the host <strong>plant</strong>s. It has been suggested that EPS are involved in the suppression<br />

of a defense response by the host <strong>plant</strong>s and EPS mutants are eliciting a<br />

pronounced <strong>plant</strong> defense response (Parniske et al. 1994). There are linkages<br />

between the lipopolysaccharide and extracellular polysaccharide synthesis. A<br />

knockout of the dTDP-L-rhamnose synthase affects lipopolysaccharide and<br />

extracellular polysaccharide production, as shown for Azorhizobium caulinodans<br />

(Gao et al. 2001). The mutation affecting this gene induced only ineffective<br />

nodular structures on the host Sesbania rostrata, with no bacteroids<br />

and leghemoglobin present in the nodules. The bacteria were trapped in<br />

thick-walled infection threads.


6 Signalling in the Rhizobia–Legumes Symbioses 111<br />

4 Signal Perception and Molecular Biology of Nodule<br />

Initiation<br />

On the molecular and cellular level a large number of responses of legume<br />

roots to Nod factors (LCOs) are known (Table 3). In the epidermis and the<br />

root hairs, ion fluxes, plasma membrane depolarization and accumulation of<br />

calcium in the root hair tips have been observed within seconds. In the range<br />

of minutes; cytoskeleton modifications, root hair deformation and specific<br />

gene expression, e.g., for ENOD 12 are found as well as calcium 2+ spiking and<br />

phospholipase C and D activation. In the range of hours to days, formation of<br />

pre-infection threads and cell divisions in the nodule primordium can be<br />

observed, together with the expression of other early nodulins such as ENOD<br />

20. At the same time, in the vascular tissue, an inhibition of polar auxin transport<br />

and a specific gene expression of ENOD 40 follow. Several of these reactions<br />

are triggered by nanomolar concentrations of Nod factors.<br />

Table 3. Responses of legume roots to Nod factors (modified from Cullimore et al. 2000;<br />

Hartog et al. 2001; Hogg et al. 2002)<br />

Tissue Responses Rapidity of Concen- Tested<br />

response tration <strong>plant</strong> genera<br />

of Nod and species<br />

factors<br />

applied<br />

Epidermis Ion fluxes Seconds nM Medicago<br />

and root Plasma membrane Seconds nM Medicago<br />

hairs depolarization<br />

Increase in intracellular pH Seconds nM Medicago<br />

Accumulation of Ca 2+ in Seconds nM Medicago, Vigna<br />

root hair tip<br />

Ca 2+ spiking 10 min nM Medicago, Pisum<br />

Gene expression Minutes–hours fM–pM Medicago<br />

(e.g., ENOD12, RIP1)<br />

Root hair deformation Minutes–hours nM–µM Many<br />

Cytoskeleton modification Minutes–hours fM–pM Phaseolus, Vicia<br />

Phospholipase C and Minutes–hours nM Vicia sativa<br />

D activation<br />

Cortex Gene expression (e.g., ENOD20) Hours–days pM Medicago<br />

Formation of pre-infection Days nM–µM Vicia<br />

threads<br />

Cell division leading to nodule Days nM–µM Many<br />

primordium formation<br />

Competitive nodulation blocking Days nM Pisum<br />

(Cnb)<br />

Vasculature Inhibition of polar auxin Minutes Trifolium<br />

transport<br />

Gene expression (e.g., ENOD40) 24 h-days nM–µM Glycine, Vicia,<br />

Medicago


112<br />

Dietrich Werner<br />

There is increasing evidence that there is more than one LCO receptor<br />

responsible for these very different biochemical and structural phenotypes of<br />

Nod factor responses. In Medicago sativa a number of responses such as root<br />

hair deformation, membrane depolarization and ion fluxes require a sulfate<br />

group on the reducing sugar, whereas nonsulfated factors can elicit an<br />

increase in the cytosolic pH in root hairs (Felle et al. 1996). The presence of<br />

more than one LCO receptor is supported by different affinities with 4 nM for<br />

the Nod factor binding site NFBS2 and 86 nM for the binding site NFBS1<br />

(Gressent et al. 1999). Both receptors have a more than 100-fold higher affinity<br />

for Nod factors compared to chitin fragments. This means that they are<br />

different from the chitin fragment receptors in legumes and grasses (Stacey<br />

and Shibuya 1997). From Glycine and Dolichos a lectin nucleotype phosphohydrolase<br />

has been shown to have Nod factor-binding activity. It is plasma<br />

membrane located and may also have some functions in phosphate transport<br />

(Etzler et al. 1999; Thomas et al. 1999). With Medicago truncatula ENOD 12gene<br />

activation, it has been shown that heterotrimeric G proteins may be<br />

involved in the LCO signal transduction pathway (Pingret et al. 1998). This<br />

indicates an interesting relationship to signalling concepts in animal cells (see<br />

Sect. 1). The involvement of Nod factors in the different signalling pathways<br />

with G protein involvement was found by studying the phospholipase C activity<br />

in Medicago roots, by using the G protein activator mastoparan (Hartog et<br />

al. 2001). Similar to Nod factors, mastoparan produces root hair deformation<br />

in zone 1. It also increases the concentration of phosphatidic acid and diacylglycerol<br />

pyrophosphate four- to sixfold. The concentration of Nod factors also<br />

plays an important role.Addition of Nod factors to the cultivar Afghanistan in<br />

pea roots strongly inhibits nodulation (Hogg et al. 2002). The most obvious<br />

phenotype was the inhibition of infection thread initiation. The gene involved<br />

had been identified as sym2 A in this pea cultivar. Nod factors (LCOs) also have<br />

effects in nonlegume cells such as tobacco protoplasts (Röhrig et al. 1996).<br />

They activate the expression of the AX11 gene involved in auxin signalling.<br />

Auxin and LCOs are transduced in tobacco cells by different pathways at, or<br />

before, the AX11 transcription. The biochemical study on Nod factor integration<br />

into membranes revealed that they are rapidly transferred between<br />

membranes and from membrane vesicles to root hair cell walls (Goedhart et<br />

al. 1999). It was also shown that the Nod factors did not flip-flop between different<br />

membrane leaflets. Nod factors are present in buffers as monomers at a<br />

concentration effective in biological systems of around 10 nM, but when<br />

dioleoylphosphatidylcholine (DOPC) vesicles are added, the Nod factors<br />

associate with these vesicles. Our limited knowledge of the involvement of<br />

calcium spiking in the Ca 2+ signal pathway is obvious in the present models of<br />

calcium oscillations (Schuster et al. 2002). A general model involves six types<br />

of concentration variables: inositol-1,4,5-trisphosphate, cytoplasmic calcium,<br />

endoplasmic reticulum calcium and mitochondrial calcium, the occupied<br />

binding site of calcium buffers and the active IP 3-receptor calcium released


6 Signalling in the Rhizobia–Legumes Symbioses 113<br />

channel. The long search for the molecular identification of the gene responsible<br />

for the regulation of nodule number on the host <strong>plant</strong>s (supernodulation)<br />

was successful. Characterized were a receptor like kinase with the HAR<br />

1 gene (Krusell et al. 2002; Nishimura et al. 2002) and the GmNARK gene from<br />

soybeans, a CLAVATA 1 like receptor kinase (Searle et al. 2002). HAR 1 and<br />

NARK are the same genes from different species, as summarized by Downie<br />

and Parniske (2002).<br />

Besides Nod factors, rhizobia also excrete other components relevant for<br />

the symbiosis development, e.g., type III secretion systems (TTSSs). Rhizobial<br />

TTSS clusters contain an open reading frame, homologous to ysc and hrc<br />

genes (Bogdanove et al. 1996; Hueck 1998; Marie et al. 2001). Two proteins<br />

have been identified in Rhizobium NGR234, secreted in a TTSS dependent<br />

way: nolX and y4xL.In Bradyrhizobium japonicum a new two-component system,<br />

ElmS and ElmR, has recently been identified, coding for a putative regulator<br />

protein and a putative sensor histidine kinase with unknown functions<br />

(Mühlencoert and Müller 2002). In agreement with the results from cytological<br />

observations in the chapter of F. Dazzo (see Chap 27, this Vol.), the population<br />

density-dependent expression of Bradyrhizobium japonicum nodulation<br />

genes has been reported (Loh et al. 2002). Induction of nod genes is high<br />

at low culture densities and is repressed at high population densities. The<br />

expression of NolA and NodD2 was mediated by an extracellular factor<br />

excreted into the medium. Two rhizobia species with a very broad host range<br />

are Rhizobium strain NGR234 and Rhizobium fredii USDA257 (Pueppke and<br />

Broughton 1999). They nodulate a wide range of mimosoid legumes, especially<br />

Acacia species and also the nonlegume Parasponia andersonii. In a few<br />

cases, only Rhizobium fredii USDA257 could nodulate some host <strong>plant</strong>s such<br />

as Glycine max and Glycine soja. The most important result was that there is<br />

no relationship between the origin of the host <strong>plant</strong>s and the ability of the<br />

strains to nodulate specific host <strong>plant</strong> species. The strain NGR234 shows significant<br />

dynamics of the genome architecture (Mavingui et al. 2002) with a<br />

large-scale DNA rearrangement, cointegration and excision exist between the<br />

three replicons, the symbiotic plasmids, the megaplasmid and the chromosome.<br />

Going from the laboratory to the field under natural conditions, especially<br />

in agricultural soils, we must realize that there are not only a large number of<br />

soil, microbial and <strong>plant</strong> factors involved, but nowadays also a large number<br />

of agrochemicals present in small quantities on seeds and also on emerging<br />

roots (Johnsen et al. 2001). Pesticide effects on specific populations of soil<br />

bacteria have been demonstrated for Rhizobium species (Ramos and Ribeiro<br />

1993) and with nitrifying bacteria (Martinez-Toledo et al. 1992). The degradation<br />

of these pesticides by the microbial communities in soils is another relevant<br />

aspect, contributing to the complexity of effects of molecules on the <strong>plant</strong><br />

<strong>surface</strong> microbial interaction (Soulas and Lagacherie 2001). A large number<br />

of resistance genes in Rhizobium species is a strategy to deal with many


114<br />

Dietrich Werner<br />

antimicrobial factors concentrated around <strong>plant</strong> roots. Several multidrug<br />

efflux pumps have been identified, e.g., in Rhizobium etli (González-Pasayo<br />

and Martinez-Romero 2000).<br />

Every specific symbiotic interaction has also to take a look to other strategies<br />

of the symbiotic communication. There is increasing evidence that the<br />

genetic requirements in the symbiotic interaction, e.g., between different Rhizobium<br />

species, pathogens and arbuscular mycorrhiza fungi species with<br />

their respective host <strong>plant</strong>s, partially overlap (Parniske 2000). Different symbiotic<br />

and pathogenic interactions finally branch to very specific functions<br />

and nutrient exchanges. Common pathways and different aspects of symbiosis<br />

and defense developments are fascinating aspects of future research<br />

(Werner et al. 2002).<br />

Acknowledgements. I thank the European Union for support in the INCO-DEV Project<br />

ICA-CT-2001–10057, the JSPS, Japan, and Mrs Lucette Claudet for the excellent work for<br />

this article.<br />

References and Selected Reading<br />

Baron M, Aslam H, Flasza M, Fostier M, Higgs JE, Mazaleyrat SL, Wilkin MB (2002) Multiple<br />

levels of Notch signal regulation. Mol Membr Biol 19:27–38<br />

Becker A, Pühler A (1998) Specific amino acid substitutions in the proline-rich motif of<br />

the Rhizobium meliloti ExoP protein result in enhanced production of low-molecular-weight<br />

succinoglycan at the expense of high-molecular-weight succinoglycan. J<br />

Bacteriol 180:395–399<br />

Becker BU, Kosch K, Parniske M, Müller P (1998) Exopolysaccharide (EPS) synthesis in<br />

Bradyrhizobium japonicum: sequence, operon structure and mutational analysis of<br />

an exo gene cluster. Mol Gen Genet 259:161–171<br />

Bogdanove AJ, Beer SV, Bonas U, Boucher CA, Collmer A, Coplin DL, Cornelis GR, Huang<br />

H-C, Hutcheson SW, Panopoulos NJ et al. (1996) Unified nomenclature for broadly<br />

conserved hrp genes of phytopathogenic bacteria. Mol Microbiol 20:681–683<br />

Breedveld MW, Miller KJ (1998) Cell-<strong>surface</strong> b-glucans. In: Spaink HP, Kondorosi A,<br />

Hooykaas PJJ (eds) The Rhizobiaceae: molecular biology of model <strong>plant</strong>-associated<br />

bacteria. Kluwer, Dordrecht, pp 81–96<br />

Breinholt V, Larsen JC (1998) Detection of weak estrogenic flavonoids using a recombinant<br />

yeast strain and a modified MCF7 cell proliferation assay. Chem Res Toxicol<br />

11:622–629<br />

Brewin NJ (1998) Tissue and cell invasion by Rhizobium: the structure and development<br />

of infection threads and symbiosomes. In: Spaink HP, Kondorosi A, Hooykaas PJJ<br />

(eds) The Rhizobiaceae: molecular biology of model <strong>plant</strong>-associated bacteria.<br />

Kluwer, Dordrecht, pp 347–360<br />

Brimecombe MJ, De Leij FA, Lynch JM (2001) The effect of root exudates on rhizosphere<br />

microbial populations. In: Pinton R, Varanini Z, Nannipieri P (eds) The rhizosphere.<br />

Marcel Dekker, New York, pp 95–140<br />

Cooper JE, Rao JR, Eveaert E, De Cooman L (1995) Metabolism of flavonoids by rhizobia.<br />

In: Tikhonovich IA, Provorov NA, Romanov VI, Newton WE (eds) Nitrogen fixation:<br />

fundamentals and applications. Kluwer, Dordrecht, pp 287–292


6 Signalling in the Rhizobia–Legumes Symbioses 115<br />

Cos P, Calomme M, Sindambiwe JB, De Bruyne T, Cimanga K, Pieters L,Vanden Berghe D<br />

(2001) Cytotoxicity and lipid peroxidation-inhibiting activity of flavonoids. Planta<br />

Med 67:515–519<br />

Cullimore JV, Ranjeva R, Bono J-J (2001) Perception of lipo-chitooligosaccharidic Nod<br />

factors in legumes. Trends Plant Sci 6:24–30<br />

Demont N, Ardourel M, Maillet F, Promé D, Ferro M et al. (1994) The Rhizobium meliloti<br />

regulatory nodD3 and syrM genes control the synthesis of a particular class of nodulation<br />

factors N-acylated by (omega-1)-hydroxylated fatty acids. EMBO J 13:2139–<br />

2149<br />

Dixon RA, Steele CL (1999) Flavonoids and isoflavonoids – a gold mine for metabolic<br />

engineering. Trends Plant Sci 4:394–400<br />

Downie JA, Parniske M (2002) Fixation with regulation. Nature 420:369–370<br />

Dunn MF, Encarnación S, Araíza G, Vargas MC, Dávalos A, Peralta H, Mora Y, Mora J<br />

(1996) Pyruvate carboxylase from Rhizobium etli: Mutant characterization, nucleotide<br />

sequence, and physiological role. J Bacteriol 178:5960–5970<br />

Endre G, Kereszt A, Kevei Z, Mihacea S, Kalo P, Kiss GB (2002) A receptor kinase gene<br />

regulating symbiotic nodule development. Nature 417:962–966<br />

Entcheva P, Liebl W, Johann A, Hartsch T, Streit WR (2001) Direct cloning from enrichment<br />

cultures, a reliable strategy for the isolation of complete operons and genes from<br />

microbial consortia. Appl Environ Microbiol 67:89–99<br />

Etzler ME, Kalsi G, Ewing NN, Roberts NJ, Day RB, Murphy JB (1999) A Nod factor binding<br />

lection with apyrase activity from legume roots. Proc Natl Acad Sci USA<br />

96:5856–5861<br />

Felle HH, Kondorosi E, Kondorosi A, Schultze M (1996) Rapid alkalinization in alfalfa<br />

root hairs in response to rhizobial lipochitooligosaccharide signals. Plant J<br />

10:295–301<br />

Gao M, D’Haeze W, dee Rycke R,Wolucka B, Holsters M (2001) Knockout of an azorhizobial<br />

dTDP-L-rhamnose synthase affects lipopolysaccharide and extracellular polysaccharide<br />

production and disables symbiosis with Sesbania rostrata. Mol Plant<br />

Microbe Interact 7:857–866<br />

Giancotti FG, Ruoslahti E (1999) Integrin signaling. Science 285:1028–1033<br />

Goedhart J, Röhrig H, Hink MA, van Hoek A, Visser AJWG, Bisseling T, Gadella Jr TWJ<br />

(1999) Nod factors integrate spontaneously in biomembranes and transfer rapidly<br />

between membranes and to root hairs, but transbilayer flip-flop does not occur. Biochemistry<br />

38:10898–10907<br />

González-Pasayo R, Martinez-Romero E (2000) Multiresistance genes of Rhizobium etli<br />

CFN42. Mol Plant Microbe Interact 13:572–577<br />

Gressent F, Drouillard S, Mantegazza N, Samain E, Geremia RA, Canut H, Niebel A,<br />

Driguez H, Ranjeva R, Cullimore J, Bono JJ (1999) Ligand specificity of a high-affinity<br />

binding site for lipochitooligosaccharidic Nod factors in Medicago cell suspension<br />

cultures. Proc Natl Acad Sci USA 96:4704–4709<br />

Harborne JB (ed) (1994) The flavonoids. Chapman and Hall, London, 621 pp<br />

Hartog M den, Musgrave A, Munnik T (2001) Nod factor-induced phosphatidic acid and<br />

diacylglycerol pyrophosphate formation: a role for phospholipase C and D in root<br />

hair deformation. Plant J 25:55–65<br />

Heilmann J, Merfort I (1998) Aktueller Kenntnisstand zum Metabolismus von Flavonoiden<br />

II. Resorption und Metabolismus von Flavonen, Flavanonen, Flavanen, Proanthocyanidinen<br />

und Isoflavonoiden. Pharmazie in unserer Zeit 27:173–183<br />

Hilpert B, Bohlmann H, op den Camp RO, Przybyla D, Miersch O, Buchala A, Apel K<br />

(2001) Isolation and characterization of signal transduction mutants of Arabidopsis<br />

thaliana that constitutively activate the octadecanoid pathway and form necrotic<br />

microlesions. Plant J 26:435–446


116<br />

Dietrich Werner<br />

Hofmann K, Heinz EB, Charles TC, Hoppert M, Liebl W, Streit WR (2000) Sinorhizobium<br />

meliloti strain 1021 bioS and bdhA gene transcription are both affected by biotin<br />

available in defined medium. FEMS Microbiol Lett 182:41–44<br />

Hogg B, Davies AE, Wilson KE, Bisseling T, Downie JA (2002) Competitive nodulation<br />

blocking of cv.Afghanistan pea is related to high levels of nodulation factors made by<br />

some strains of Rhizobium leguminosarum bv. viciae. Mol Plant Microbe Interact<br />

15:60–68<br />

Hollingsworth RI, Carlson RW (1989) 27-hydroxyoctacosanoic acid is a major structural<br />

fatty acyl component of the lipopolysaccharide of Rhizobium trifolii ANU 843. J Biol<br />

Chem 264:9300–9303<br />

Hollman PC, Katan MB (1998) Bioavailability and health effects of dietary flavonols in<br />

man. Arch Toxicol (Suppl) 20:237–248<br />

Hueck CJ (1998) Type III in protein secretion systems in bacterial pathogens of animals<br />

and <strong>plant</strong>s. Microbiol Mol Biol Rev 62:379–433<br />

Ishimi Y, Miyaura C, Ohmura M, Onoe Y, Sato T, Uchiyama Y, Ito M, Wang X, Suda T,<br />

Ikegami S (1999) Selective effects of genistein, a soybean isoflavone on b-lymphopoiesis<br />

and bone loss caused by estrogen deficiency. Endocrinology 140:1893–<br />

1900<br />

Jackson MB (2002) Long-distance signalling from roots to shoots assessed: the flooding<br />

story. J Exp Bot 53:175–181<br />

Johnsen K, Jacobsen CS, Torswik V, Sorensen J (2001) Pesticide effects on bacterial diversity<br />

in agricultural soils – a review. Biol Fertil Soils 33:443–453<br />

Kannenberg EL, Reuhs BL, Forsberg LS, Carlson RW (1998) Lipopolysaccharides and Kantigens.<br />

Their structures, biosynthesis and functions. In: Spaink HP, Kondorosi A,<br />

Hooykaas PJJ (eds) The Rhizobiaceae: molecular biology of model <strong>plant</strong>-associated<br />

bacteria. Kluwer Acad, Dordrecht, pp 120–154<br />

Krusell L, Madsen LH, Sato S, Aubert G, Genua A, Szczyglowski K, Duc G, Kaneko T,<br />

Tabata S, de Bruijn F, Pajuelo E, Sandal N, Stougaard J (2002) Shoot control of root<br />

development and nodulation is mediated by a receptor-like kinase. Nature 420:422–<br />

426<br />

Lake JA, Woodwart FI, Quick WP (2002) Long-distance CO 2 signalling in <strong>plant</strong>s. J Exp<br />

Biol 53:183–193<br />

Lerouge P, Roche P, Faucher C et al. (1990) Symbiotic host-specificity of Rhizobium<br />

meliloti is determined by a sulphated and acylated glucosamine oligosaccharide signal.<br />

Nature 344:781–784<br />

Loh J, Lohar DP, Andersen B, Stacey G (2002) A two-component regulator mediates population-density-dependent<br />

expression of the Bradyrhizobium japonicum nodulation<br />

genes. J Bacteriol 184:1759–1766<br />

López-Lara IM, Geiger O (2001) The nodulation protein NodG shows the enzymatic<br />

activity of an 3-Oxoacyl-acyl carrier protein reductase. Mol Plant-Microbe Interact<br />

14:349–357<br />

Marie C, Broughton WJ, Deakin WJ (2001) Rhizobium type III secretion systems: legume<br />

charmers or alarmers? Curr Opin Plant Biol 4:336–342<br />

Martinez-Toledo MV, Salmeron V, Gonzalez-Lopez J (1992) Effect of the insecticides<br />

pyrimifosmethyl and chlorpyrifos on soil microflora in an agricultural loam. Plant<br />

Soil 147:25–30<br />

Mavingui P, Flores M, Guo X, Dávila G, Perret X, Broughton WJ, Palacios R (2002)<br />

Dynamics of genome architecture in Rhizobium sp. strain NGR 234. J Bacteriol<br />

184:171–176<br />

Maximo P, Lourenço A, Feio SS, Roseiro JC (2002) Flavonoids from Ulex airensis and<br />

Ulex europaeus spp. europaeus. J Nat Prod 65:175–178<br />

Morishita M, Morimoto F, Kitamura K, Koga T, Fukui Y, Maekawa H, Yamashita I, Shimoda<br />

C (2002) Phosphatidylinositol 3-phosphate 5-kinase is required for the cellular


6 Signalling in the Rhizobia–Legumes Symbioses 117<br />

response to nutritional starvation and mating pheromone signals in Schizosaccharomyces<br />

pombe. Genes Cells 7:199–215<br />

Morris VJ, Brownsey GJ, Chilvers GR, Harris JE, Gunning AP, Stevens BJH (1991) Possible<br />

biological roles for Rhizobium leguminosarum extracellular polysaccharide and<br />

cyclic glucans in bacteria-<strong>plant</strong> interactions for nitrogen-fixing bacteria. Food<br />

Hydrocoll 5:185–188<br />

Mühlencoert E, Müller P (2002) A novel two-component system of Bradyrhizobium<br />

japonicum: ElmS and ElmR are encoded in diverse orientations. DNA Sequence<br />

13:93–102<br />

Neumann G, Römheld V (2001) The release of root exudates as affected by the <strong>plant</strong>’s<br />

physiological status. In: Pinton R, Varanini Z, Nannipieri P (eds) The Rhizosphere.<br />

Marcel Dekker, New York, pp 41–94<br />

Nishimura R, Hayashi M,Wu G-J, Kouchi H, Imaizumi-Anraku H, Murakami Y, Kawasaki<br />

S, Akao S, Ohmori M, Nagasawa M, Harada K, Kawaguchi M (2002) HAR1 mediates<br />

systemic regulation of symbiotic organ development. Nature 420:426–429<br />

Pacios-Bras C, van der Burgt YEM, Deelder AM, Vinuesa P, Werner D, Spaink HP (2002)<br />

Novel lipochitin oligosaccharide structures produced by Rhizobium etli KIM5s. Carbohydr<br />

Res 337:1193–1202<br />

Pai TG, Suiko M, Sakakibara Y, Liu M-C (2001) Sulfation of flavonoids and other phenolic<br />

dietary compounds by the human cytosolic sulfotransferases. Biochem Biophys<br />

Res Com 285:1175–1179<br />

Paiva NL (2000) An introduction to the biosynthesis of chemicals used in <strong>plant</strong>-microbe<br />

communication. J Plant Growth Regul 19:131–143<br />

Parniske M (2000) Intracellular accommodation of microbes by <strong>plant</strong>s: a common developmental<br />

program for symbiosis and disease? Curr Opin Plant Biol 3:320–328<br />

Parniske M, Schmidt PE, Kosch K, Müller P (1994) Plant defense responses of host <strong>plant</strong>s<br />

with determinate nodules induced by EPS-defective exoB mutants of Bradyrhizobium<br />

japonicum. Mol Plant Microbe Interact 7:631–638<br />

Phillips DA, Joseph CM,Yang GP, Martínez-Romero E, Sanborn JR,Volpin H (1999) Identification<br />

of lumichrome as a Sinorhizobium enhancer of alfalfa root respiration and<br />

shoot growth. Proc Natl Acad Sci USA 96:12275–12280<br />

Pingret JL, Journet EP, Barker DG (1998) Rhizobium Nod factor signalling: evidence for<br />

a G protein-mediated transduction mechanism. Plant Cell 10:659–672<br />

Polavieja GG de (2002) Errors drive the evolution of biological signalling to costly codes.<br />

J Theor Biol 214:657–664<br />

Pueppke SG, Broughton WJ (1999) Rhizobium sp. Strain NGR234 and R. fredii USDA257<br />

share exceptionally broad, nested host ranges. Mol Plant Microbe Interact 12:293–318<br />

Ramos MLG, Ribeiro WOJ (1993) Effect of fungicides on survival of Rhizobium on seeds<br />

and the nodulation of bean (Phaseolus vulgaris L.) Plant Soil 152:145–150<br />

Rice-Evans CA, Miller NJ (1996) Antioxidant activities of flavonoids as bioactive components<br />

of food. Biochem Soc Trans 24:790–794<br />

Röhrig H, Schmidt J, Walden R, Czaja I, Lubenow H, Wieneke U, Schell J, John M (1996)<br />

Convergent pathways for lipochitooligosaccharide and auxin signaling in tobacco<br />

cells. Proc Natl Acad Sci USA 93:13389–13392<br />

Rolin DB, Pfeffer PE, Osman SF, Szwergold BS, Kappler F, Benesi AJ (1992) Structural<br />

studies of a phosphocholine substituted b-(1,3); (1,6) macrocyclic glucan from<br />

Bradyrhizobium japonicum USDA 110. Biochim Biophys Acta 1116:215–225<br />

Schlaman HRM, Gisel AA, Quaedvlieg NEM, Bloemberg GV, Lugtenberg BJJ et al. (1997)<br />

Chitin oligosaccharides can induce cortical cell division in roots of Vicia sativa when<br />

delivered by ballistic microtargeting. Development 124:4887–4893<br />

Schoonejans E, Expert D, Toussaint A (1987) Characterization and virulence properties<br />

of Erwinia chrysanthemi lipopolysaccharide-defective. øEC2-resistant mutants. J<br />

Bacteriol 169:4011–4017


118<br />

Dietrich Werner<br />

Schröder O, Wagner R (2002) The bacterial regulatory protein H-NS – a versatile modulator<br />

of nucleic acid structures. Biol Chem 383:945–960<br />

Schuster S, Marhl M, Höfer T (2002) Modelling of simple and complex calcium oscillations.<br />

From single-cell responses to intercellular signalling. Eur J Biochem 269:1333–<br />

355<br />

Schwartz MA, Ginsberg MH (2002) Networks and crosstalk: integrin signalling spreads.<br />

Nature Cell Biol 4:E65-E68<br />

Searle IR, Men AT, Laniya TS, Buzas DM, Searle IR, Men AE, Laniya TS, Buzas DM, Iturbe-<br />

Ormaetxe I, Carroll BJ, Gresshoff PM (2003) Long-distance signaling in nodulation<br />

directed by a CLAVATA1-like receptor kinase. Science 3299:109–112<br />

Soulas G, Lagacherie B (2001) Modelling of microbial degradation of pesticides in soils.<br />

Biol Fertil Soils 33:551–557<br />

Spaink HP (2000) Root nodulation and infection factors produced by rhizobial bacteria.<br />

Ann Rev Microbiol. 54:257–288<br />

Spaink HP, Sheeley DM, van Brussel AAN et al. (1991) A novel highly unsaturated fatty<br />

acid moiety of lipooligosaccharide signals determines host specificity of Rhizobium.<br />

Nature 354:125–130<br />

Stacey G, Shibuya N (1997) Chitin recognition in rice and legumes. Plant Soil<br />

194:161–169<br />

Staehelin C, Schultze M, Tokuyasu K, Poinsot V, Promé J-C, Kondorosi E, Kondorosi A<br />

(2000) N-deacetylation of Sinorhizobium meliloti Nod factors increases their stability<br />

in the Medicago sativa rhizosphere and decreases their biological activity. Mol Plant<br />

Microbe Interact 13:72–79<br />

Staswick PE, Yuen GY, Lehman CC (1998) Jasmonate signaling mutants of Arabidopsis<br />

are susceptible to the soil fungus Pythium irregulare. Plant J 15:747–754<br />

Stracke S, Kistner C,Yoshida S, Mulder L, Sato S, Kaneko T, Tabata S, Sandal N, Stougaard<br />

J, Szczyglowski K, Parniske M (2002) A <strong>plant</strong> receptor-like kinase required for both<br />

bacterial and fungal symbiosis. Nature 417:959–962<br />

Streit WR, Joseph CM, Phillips DA (1996) Biotin and other water-soluble vitamins are<br />

key growth factors for alfalfa rhizosphere colonization by Rhizobium meliloti 1021.<br />

Molec Plant-Microbe Interact 9:330–338<br />

Strzelczyk E, Rozycki H (1985) Production of B-group vitamins by bacteria isolated from<br />

soil, rhizosphere, and mycorrhizosphere of pine (Pinus sylvestris L.). Zbl Mikrobiol<br />

140:293–301<br />

Thomas C, Sun Y, Naus K, Lloyd A, Roux S (1999) Apyrase functions in <strong>plant</strong> phosphate<br />

nutrition and mobilizes phosphate from extracellular ATP. Plant Physiol 119:543–551<br />

Thomma B, Eggermont K, Penninckx IAMA, Mauch-Mani B,Vogelsang R, Cammue BPA,<br />

Broekaert WF (1998) Separate jasmonate-dependent and salicylate-dependent<br />

defense-response pathways in Arabidopsis are essential for resistance to distinct<br />

microbial pathogens. Proc Natl Acad Sci USA 95:15107–15111<br />

Uren NC (2001) Types, amounts, and possible functions of compounds released into the<br />

rhizosphere by soil-grown <strong>plant</strong>s. In: Pinton R,Varanini Z, Nannipieri P (eds) The rhizosphere.<br />

Marcel Dekker, New York, pp 19–40<br />

Van Workum WAT, Kijne JW (1998) Biosynthesis of rhizobial exopolysaccharides and<br />

their role in the root nodule symbiosis of leguminous <strong>plant</strong>s. In: Romeo JT, Downum<br />

KR, Verpoorte R (eds.) Phytochemical signals and <strong>plant</strong>-microbe interactions.<br />

Plenum, New York, pp 139–166<br />

Vinuesa P, Reuhs BL, Breton C, Werner D (1999) Identification of a plasmid-borne locus<br />

in Rhizobium. etli KIM5s involved in lipopolysaccharide O-chain synthesis and<br />

nodulation of Phaseolus vulgaris. J Bacteriol 181:5606–5614<br />

Wang GJ, Lapcik O, Hampl R, Uehara M, Al-Maharik N, Stumpf K, Mikola H, Wähälä K,<br />

Adlercreutz H (2000) Time-resolved fluoroimmunoassay of plasma daidzein and<br />

genistein. Steroids 65:339–348


6 Signalling in the Rhizobia–Legumes Symbioses 119<br />

Weber H (2002) Fatty acid-derived signals in <strong>plant</strong>s. Trends Plant Sci 7:217–224<br />

Werner D (1992) Symbiosis of <strong>plant</strong>s and microbes. Chapman and Hall, London, 389 pp<br />

Werner D (2001) Organic signals between <strong>plant</strong>s and microorganisms. In: Pinton R,<br />

Varanini Z, Nannipieri P (eds) The rhizosphere. Marcel Dekker, New York, pp 197–222<br />

Werner D, Müller P (2003) Communication and efficiency in the symbiotic signal<br />

exchange. In: Heldmaier G, Werner D (eds) Environmental signal processing and<br />

adaptation. Springer, Berlin Heidelberg New York, pp 9–38<br />

Werner D, Barea JM, Brewin NJ, Cooper JE, Katinakis P, Lindström K, O’Gara F, Spaink<br />

HP, Truchet G, Müller P (2002) Symbiosis and defence in the interaction of <strong>plant</strong>s with<br />

microorganisms. Symbiosis 32:83–104<br />

Wilkinson S, Davies WJ (2002) ABA-based chemical signalling: the co-ordination of<br />

responses to stress in <strong>plant</strong>s. Plant Cell Environ 25:195–210


7 The Functional Groups of Micro-organisms Used<br />

as Bio-indicator on Soil Disturbance Caused by<br />

Biotech Products such as Bacillus thuringiensis and<br />

Bt Transgenic Plants<br />

Galdino Andrade<br />

1 Introduction<br />

Insects are usually controlled with insecticides. Of the insecticides 5 % are<br />

biological, and more than 90 % of biological insecticides are based on Bacillus<br />

thuringiensis (Bt; Sanchis 2000). The use of bio-insecticides has increased<br />

because of the growing need to obtain better quality food and to protect the<br />

environment, but very little is known about the impact these organisms have<br />

on the environment and mainly on the soil functional microorganism<br />

groups.<br />

Due to the efficiency of bio-insecticides based on B. thuringiensis,the gene<br />

which produces the bio-insecticide crystal was introduced into <strong>plant</strong>s to produce<br />

Bt-transgenic <strong>plant</strong>s. Transformed tobacco using the Ti plasmodium<br />

from Agrobacterium tumefasciens was obtained in the 1980s. Later, the electroporation<br />

and bombardment or bio-ballistic of embryos method, which is<br />

more efficient for transformation of a greater number of <strong>plant</strong> species with<br />

the cry B. thuringiensis gene, was used (Peferoen 1997). The second generation<br />

of Bt-transgenic <strong>plant</strong>s is presently obtained with the introduction of at<br />

least two cry genes in the <strong>plant</strong> genome, and there are already more than 20<br />

species of transgenic <strong>plant</strong>s of economic importance being used in a few<br />

countries (Sanchis 2000).<br />

Although transgenic <strong>plant</strong>s have been produced and sown for two decades,<br />

there is little information about their environmental impact. Currently proposed<br />

<strong>plant</strong> gene products will probably have less impact on soil ecosystems<br />

than some familiar and accepted practices. However, some transgenic <strong>plant</strong><br />

products may have measurable adverse effects on soil organisms that will<br />

have to be monitored for some years after widespread introduction (Tomlin<br />

1994).<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


122<br />

Galdino Andrade<br />

Some studies have assessed B. thuringiensis spore and vegetative cell survival<br />

in the soil. The soil permanence of the protein crystal released by Bttransgenic<br />

maize root exudates has also been assessed (Saxena et al. 1999).<br />

The analysis of the soil stability of B. thuringiensis indicated that the bacterium<br />

was not active, and that the number of cells inoculated either in the<br />

vegetative or in the spore form decreased rapidly a few hours after inoculation.<br />

However, the soil can be considered its natural deposit, as the spores are<br />

released into the soil after the death of an insect and persist in this form for<br />

several years until they find another host insect. The B. thuringiensis toxin<br />

produced by Bt-maize is transferred to the soil by root exudates, pollen and<br />

other <strong>plant</strong> parts. Tapp et al. (1995a) reported that the B. thuringiensis crystal<br />

is adsorbed by the clay minerals in the soil, and thus may be protected from<br />

biodegradation action of hydrolytic enzymes such as proteases, and may<br />

remain active in the soil for several months.<br />

Pesticides have protocols to evaluate non-target effects where many organisms<br />

are used as biological indicators. Similar test protocols could be<br />

extended to <strong>plant</strong> bio-insecticide manufacturers. The soil biological process is<br />

poorly understood, and care should be taken to prevent further negative<br />

impacts on soil ecosystems from genetically modified organisms. Although<br />

these might be non-target events, all effects of Bt-<strong>plant</strong>s on the soil environment<br />

must be well understood.<br />

2 General Aspects of Bacillus thuringiensis<br />

The B. thuringiensis bacterium is a Gram positive rod, aerobic, chemoheterotrophic,<br />

with perithiquious flagella that sporulates when the environmental<br />

conditions are not favourable. The bio-insecticide protein is formed<br />

when the sporulation event is activated and cells should be isolated from soil<br />

or infected insects.<br />

Meadows (1993) suggested three hypotheses for the natural habitat of B.<br />

thuringiensis based on isolation studies. The bacterium could be an insect<br />

pathogen, a component of the normal flora of the phylosphere tree species, or<br />

a soil microorganism.<br />

According to the first hypothesis, the release of the crystal would be a strategy<br />

to kill the insect larvae, which would permit spore germination and vegetative<br />

cell multiplication.<br />

The second hypothesis was suggested by a study where great quantities of<br />

B. thuringiensis were found in tree species, and could be disseminated by the<br />

wind or rain, and the soil would only be a deposit where B. thuringiensis did<br />

not multiply (Smith and Couche 1991).<br />

These studies have shown that B. thuringiensis is widely distributed in soils<br />

throughout the world. Wide distribution even in localities which cannot be<br />

correlated with the presence of insects reinforces the hypothesis that the bac-


7 The Functional Groups of Micro-organisms and Biotech Products 123<br />

terium uses the soil as a natural habitat (Martin and Travers 1989). Soil survival<br />

studies showed that the number of inoculated vegetative cells decreases<br />

rapidly and only viable spores are found after 5 days. Vegetative cell multiplication<br />

was not observed (Villas-Bôas et al. 2000).<br />

Lereclus et al. (2000) suggested that during the sporulation phase the cry<br />

regulation gene has the function of producing high quantities of Cry protein<br />

to kill the insect, allowing the bacterium to complete its biological cycle,<br />

(spore germination – multiplication – sporulation – dispersion). Virulence<br />

factors, acting in unfavourable environmental conditions, enable the bacterium<br />

to damage and invade host tissues, obtaining ideal conditions for<br />

spore germination and cell multiplication.<br />

3 Survival in the Soil<br />

Knowledge of B. thuringiensis survival in the soil is important in the context<br />

of its use for biological control. It is also relevant in the study of the interactions<br />

with other soil microorganisms. The commercial B. thuringiensis formulas<br />

are composed of mixtures of spores and crystals, which are released in<br />

great quantities into the environment each year. The behaviour of these<br />

spores and crystals in the soil has been studied in the field, greenhouse and in<br />

sterilised and natural soils (Thomas et al. 2000; Villas Bôas et al. 2000). In<br />

these assessments, the spores were inoculated into the soil and their recovery<br />

was monitored.<br />

Some reports demonstrated that, initially, the number of colony spore<br />

forming units of B. thuringiensis declines rapidly. Vilas-Boas et al. (2000)<br />

observed that after 24 h only 20 % of the spores survived in sterilised<br />

soils.<br />

Addison (1993) observed that several factors could affect the survival of the<br />

spores, such as pH, moisture and nutrient availability. Spore viability seemed<br />

to be little influenced by soil type or temperature. The results on nutrient<br />

availability are controversial. Cell remains could be an extra source of nutrients<br />

for the native microbiota and inoculated bacteria, but B. thuringiensis is<br />

unable to use nutrients released by the lysis of inoculated dead cells (West et<br />

al. 1985).<br />

Competition with soil microbiota is one of the factors that most affects<br />

spore viability in the soil. In the soil microcosm, B. thuringiensis spores have<br />

a greater mortality index because of competition from other microorganisms<br />

(Pruett et al. 1980).<br />

The initial decline in the number of colony forming units after a 24-h permanence<br />

in the soil is about 80 %. The surviving cells rapidly produce spores,<br />

increasing the number of viable spores until the number of vegetative cells is<br />

matched. This means that at a given moment, the number of spores will equal<br />

that of the vegetative cells, and will remain stable for several months.


124<br />

Galdino Andrade<br />

Nutrient limitation is one of the main characteristics of soils (Edwards<br />

1993). Microorganisms develop strategies in this oligotrophic environment to<br />

capture nutrients and survive the environmental stress. Spore formation is<br />

one of the strategies of bacterium survival when the environmental conditions<br />

are unfavourable for growth. The spore can endure stress conditions for<br />

long periods of time.<br />

When inoculated into the soil, the spore number falls, after this initial fall,<br />

the number of cells stabilised due to vegetative cell sporulation. Petras and<br />

Casida (1985) observed an exceptional fall in the spore number during the<br />

first 2 weeks, but the number of viable spores stabilised in the third week.<br />

However, West et al. (1985) reported that spores of the B. thuringiensis var<br />

aizawai HD137 presented low mortality when inoculated into sterilised soil<br />

and persisted with little decrease in the initial number for 135 days.<br />

4 History of Bacillus thuringiensis-Transgenic Plants<br />

At the end of the 1980s, tobacco <strong>plant</strong>s were the first to receive the B.<br />

thuringiensis cry gene using the Agrobacterium transformation system. The<br />

Agrobacterium tumefasciens system was used in the transformation of several<br />

dicotyledon <strong>plant</strong>s. However, the electroporation and particle bombardment<br />

methods are more efficient for the transformation of monocotyledon and<br />

other dicotyledon <strong>plant</strong>s (Peferoen 1997).<br />

The first transgenic <strong>plant</strong>s obtained showed low expression of the complete<br />

gene for Cry protein production. From then onwards, only truncated genes<br />

were introduced which codify the toxic nucleus of the Cry protein, thereby<br />

increasing the expression in several <strong>plant</strong>s such as tobacco (Mazier et al.<br />

1997), sugar cane (Arencibia et al. 1997) and peanuts (Singsit et al. 1997). Bttransgenic<br />

potato, cotton and maize cultivation (Schnepf et al. 1998) began in<br />

1996 and, today, there are more than 20 transgenic <strong>plant</strong> species of agronomic<br />

interest on the market.<br />

Promoters, such as CaMV 35 s of the cauliflower mosaic virus and ubiquitinine-1<br />

from maize are being used to increase the expression of the cry gene to<br />

the required levels. In addition to this strategy, greater cry gene expression levels<br />

were obtained by altering the sequence of the gene to increase the cytosine<br />

and guanine content and enhance the expression level from 0.02 to 0.5 % of the<br />

<strong>plant</strong> soluble protein.However,the most pronounced expression level of the cry<br />

gene in <strong>plant</strong>s (3–5 % soluble protein) was obtained with the introduction of the<br />

unmodified gene in chloroplasts, a cell organelle which has the transcription<br />

and translation apparatus similar to that of prokaryotes (McBride et al.1995).<br />

New generations of transgenic <strong>plant</strong>s have been developed to express other<br />

types of cry genes or genes expressing proteins that could have insecticide<br />

action, such as protease inhibitors, lectin, kinases, cholesterol, oxidases,<br />

inhibitors of the a-amylase and Vip proteins (Sanchis 2000).


7 The Functional Groups of Micro-organisms and Biotech Products 125<br />

5 Persistence of the Protein Crystal in the Soil<br />

The release of the use of transgenic <strong>plant</strong>s with cry genes for agricultural pest<br />

control in the 1990s has raised a lot of controversy and concern in the scientific<br />

community. It is common sense that four factors must be very carefully<br />

assessed: (1) the potential of selection for insects resistant to the Cry proteins,<br />

(2) the persistence of the crystal released by the root exudates and lysis in the<br />

soil, (3) the non-selectivity towards other non-pathogenic insects, (4) the<br />

impact of the bio-insecticide crystal protein on the functional groups of soil<br />

microorganisms.<br />

Several studies on the persistence of the B. thuringiensis toxin released by<br />

transgenic <strong>plant</strong>s into the soil have shown that the toxin degradation is relatively<br />

quick during the first 45 days, and less than 25 % of the initial bio-activity<br />

is maintained after 120 days (Palm et al. 1994, 1996; Sims and Holden 1996;<br />

Sims and Ream 1997). On the other hand, Tapp et al. (1995a) showed that part<br />

of the insecticide activity of B. thuringiensis may be maintained because of<br />

the rapid adsorption and binding of the toxin in the soil clay minerals. Other<br />

authors reported that a substantial proportion of the Cry1Ab toxin released in<br />

the Bt-maize roots exudates could be detected and maintained their bioinsecticide<br />

activity for 234 days after release into the soil. This indicates that<br />

the protein crystal is very stable in the soil and is protected from microbial<br />

action due to adsorption by the soil clay (Saxena et al. 1999). Experimental<br />

results on adsorption of the protein to soil particles (Saxena and Stotzky<br />

2000) indicated that the cry1Ab gene coded toxin released by Bt-maize root<br />

exudates in sandy soil supplemented with montmorillonite and caullinite<br />

bound preferentially to clay minerals. This confirmed that adsorption to clay<br />

minerals is one of the main factors in the permanence and activity maintenance<br />

of the bio-insecticide crystals in the soil.<br />

Crystals were detected by the ELISA method (enzyme-linked immunosorbent<br />

assay). The permanence of the toxin was also determined in other soil<br />

types, with predominance of those with low organic matter content. Palm et<br />

al. (1994, 1996) observed a fall in the purified toxin concentration in the soil in<br />

the first 14 days after inoculation, with stabilisation after this period. In the<br />

case of transgenic <strong>plant</strong>-produced toxin, the fall in the soil concentration<br />

occurred for about 10 days and then remained stable. However, toxin production<br />

was continuous throughout the <strong>plant</strong> lifecycle with a consequent accumulation<br />

of bio-insecticide crystals in the soil. The lowest rate of recuperation<br />

after extraction was obtained in soil with a high quantity of organic matter,<br />

indicating that much of this protein may also be adsorbed by the soil organic<br />

matter.<br />

Saxena and Stotzky (2000) observed that the B. thuringiensis toxin<br />

expressed in Bt-transgenic maize was released into the rhizosphere through<br />

exudates and lysates and that much of the released crystal remained active<br />

for several months. Although Bt-transgenic <strong>plant</strong>s produce and accumulate


126<br />

Galdino Andrade<br />

significant quantities of bio-insecticide crystals in pollen, leaves and roots,<br />

their effect on the functional groups of soil microorganisms is little understood.<br />

The influence of the mineralogical composition of the soil on the stability<br />

of the bio-insecticide crystals has been reported by several authors. Tapp et al.<br />

(1995a) showed that the toxin is rapidly adsorbed or linked to clay minerals in<br />

the soil, remaining protected from degradation by soil microorganisms. In<br />

another study, Tapp et al. (1995b) showed that the toxin adsorbed by clay minerals<br />

becomes resistant to hydrolytic action of the enzymes produced by the<br />

soil microbiota. It has also been shown that soils with high levels of organic<br />

matter have a high protein crystal adsorption capacity (Palm et al. 1994). Sims<br />

et al. (1996) observed that B. thuringiensis var kurstaki Cry1Ab toxin present<br />

in Bt-transgenic maize tissues incorporated in the soil can be detected by<br />

bioassays with insects susceptible to the bio-insecticide action of the crystal.<br />

According to the results obtained by these authors, the bioassay allows the<br />

detection of smaller quantities of the protein (around 0.5 ng/ml in the diet)<br />

compared to the ELISA test (50.0 ng/g of soil). Sims et al. (1997), working with<br />

bioassays on B. thuringiensis var kurstaki transgenic cotton toxin inactivation<br />

in soils, observed that the toxin mean life in the soil ranges from 15 to 32 days,<br />

and less than 25 % of the initial activity remains after 120 days.<br />

6 Effect of Bacillus thuringiensis and Its Bio-insecticide<br />

Protein on Functional Soil Microorganism Assemblage<br />

Plant roots and their <strong>surface</strong>s constitute dynamic habitats densely colonised<br />

by soil-borne microbiota. The high microbial activity in these habitats is due<br />

to a flow of organic substances from the photosynthetic parts of the <strong>plant</strong>s to<br />

the roots (Olsson and Person 1999). This flow consists of low molecular<br />

weight organic substances (e.g. sugars, fatty acids and amino acids), as well as<br />

more complex substances (e.g. starch, cellulose and proteins). The chemical<br />

composition of this organic matter (the rhizodeposition) varies among <strong>plant</strong><br />

species and growth stages, and is affected by <strong>plant</strong> growth conditions (Curl<br />

and Truelove 1986). The functional groups of microorganisms of nitrogen,<br />

phosphorus and carbon cycling are important to the maintenance of nutrient<br />

turnover. These microorganisms interact with the <strong>plant</strong> roots, supply nutrients<br />

and participate actively in <strong>plant</strong> nutrition and growth (Andrade 1999).<br />

Mycorrhizal fungi are ubiquitous soil inhabitants and form a symbiotic<br />

relationship with the roots of most terrestrial <strong>plant</strong>s. When arbuscular mycorrhizae<br />

(AM) form, there are significant changes in the <strong>plant</strong> and root physiology.<br />

Photosynthetic rates increase and the nutritional status of the host tissues<br />

changes and thus, the quality and quantity of root exudates (Linderman<br />

1992). Altered exudation induces changes in the composition of microbial<br />

communities in the rhizosphere soil (Andrade et al. 1997) that may influence


7 The Functional Groups of Micro-organisms and Biotech Products 127<br />

formation and behaviour of rhizobia nodules. Such changes could influence<br />

the competition between rhizobia and other rhizobacteria. If bacteria selectively<br />

favoured in the rhizoplane enhanced rhizobia competitiveness, then<br />

nodulation would be favoured (Linderman 1992).<br />

In general, soil productivity and nutrient cycling are influenced by soil<br />

microbial populations. The relationships among functional groups of<br />

microorganisms of C, N and P cycling, and their influence on the <strong>plant</strong><br />

growth, are potential indicators to evaluate disturbance in the soil environment.<br />

A corresponding rise in the input of Bt and its toxins into soil systems can<br />

be expected with the increased use of B. thuringiensis-based insecticides,<br />

whether by direct spraying, in insect cadavers, or in transgenic <strong>plant</strong> material<br />

or microorganisms (Addison 1993). Very little attention has been paid to the<br />

effects that B. thuringiensis might have on the indigenous soil assemblages,<br />

and the information that is available is often confusing. Petras and Casida<br />

(1985) reported that endogenous soil bacteria, actinomycetes, fungi and<br />

nematodes increased moderately compared with the control when using a<br />

spore and crystal suspension of B. thurigiensis subsp. kurstaki isolated from<br />

Dipelr, a commercial preparation. Pruett et al. (1980) inoculated B. thuringiensis<br />

subsp. galleriae into clay soil and reported that bacterial populations<br />

increased 2 weeks after inoculation and were still increasing at the end of the<br />

135-day experiment.<br />

In contrast to the above studies, Atlavinyté et al. (1982) reported a decrease<br />

in indigenous soil microbiota when B. thuringiensis subsp. galleriae was inoculated<br />

into the soil. Bacterial numbers had decreased 50 % and actinomycetes<br />

by 90 % after 45 days, and in contrast, fungal populations had increased by<br />

300–500 % compared with the control.<br />

The influence of B. thuringiensis subsp. kurstaki and its protein on functional<br />

groups of soil assemblages was assessed for the first time in our laboratory,<br />

and we discuss our findings as follows.<br />

In non-sterile soil B. thuringiensis vegetative cells seemed to be unable to<br />

compete with the indigenous microorganisms in non-sterile soil. Under these<br />

conditions, the number of cells decreased drastically, sporulation occurred<br />

quickly and the number of spores was stable, approximately four log unit for<br />

at least 45 days. The cell number decrease was greater in non-sterile soil than<br />

in sterile soil conditions (Villas Bôas et al. 2000). The same results were found<br />

by Thomas et al. (2000). Their results suggested that, although the soils used<br />

were of different types and composition, B. thuringiensis apparently did not<br />

show biological activity after spores had been released into the environment<br />

and could persist for several years (Pruett et al. 1980; Pedersen et al. 1995).<br />

However, some species of Bacillus genera such as B. megaterium, B. subtilis, B<br />

cereus suppressed pathogen fungi and/or bacteria and saprophyte fungi populations<br />

in microcosm soil (Reddy and Rhae 1989; Halverson et al. 1993;<br />

Young et al. 1995; Kim et al. 1997). In many cases, the results showed a great


128<br />

Galdino Andrade<br />

decrease in the viable vegetative or spore form Bacillus units in the soil. Reddy<br />

and Rhae (1989) reported that a strain of B. subtilis introduced into an onion<br />

rhizosphere at a concentration of 7.2¥10 5 seed –1 could be recovered at only<br />

7¥10 3 <strong>plant</strong> –1 after 30 days, despite the decrease in numbers, the B. subtilis was<br />

effective in suppressing indigenous soil microbiota in the rhizosphere. Young<br />

et al. (1995) also observed that B. cereus survival was not influenced by developing<br />

wheat roots and the absence of a rhizosphere effect may be due to the<br />

fact that B. cereus was isolated originally from non-rhizosphere soil. The<br />

Bacillus spp. are often reported to be present in low numbers in the rhizosphere<br />

compared with other bacteria, such as fluorescent pseudomonas (Elliot<br />

Juhnke et al. 1987).<br />

Populations of C, P-cycling microorganisms and formed nodules changed<br />

during the <strong>plant</strong> growth period and were influenced by B. thuringiensis inoculation<br />

in soybean <strong>plant</strong>s. No differences were found on assemblages of bacteria<br />

and fungi in soil inoculated with B. thuringiensis, but time influenced the<br />

populations. The time corresponded to <strong>plant</strong> growth, AM root colonisation<br />

and nodule formation. Some physiological changes during <strong>plant</strong> growth,<br />

including C compounds released to the medium, influenced bacteria growth<br />

(Amora-Lazcano and Azcón 1997). AM colonisation and Bradyrhizobium<br />

japonicum nodulation normally decreased the amount of <strong>plant</strong> root-derived<br />

and organic matter available for heterotrophic bacteria and other soil<br />

microorganism growth by altering the root cell permeability, thus affecting<br />

exudation (Schwab et al. 1983). The carbon cycling microbiota populations<br />

also decreased their number of cells, possibly because of changes in C concentration<br />

in the rhizosphere. Negative correlation between symbiotic and<br />

cellulolytic, amylolytic and proteolytic microorganisms shows that carbon<br />

compounds from the root are important factors for their proliferation. Deleterious<br />

effects of AM roots on soil bacteria have also been observed, suggesting<br />

C competition (Marschner and Crowley 1996), although AM fungi and<br />

rhizobia do not consume C from the rhizosphere due to their symbiotic condition<br />

(Secilia and Bagyaraj 1987; Paulitz and Linderman 1989). Cellulolytic<br />

and amylolytic microorganisms decreased their cell number during the<br />

experiment, whereas proteolytic microorganisms increased their population<br />

the first time. This result suggested that this group had an extra supply of<br />

nutrients from inoculated crystal protein. The faster decrease in the proteolytic<br />

cell number after day 15 could be explained by the small amount of ICP<br />

free in the soil. Saxena et al. (1999) suggested that ICP binds rapidly and<br />

tightly to clays and humic acids and is protected against microbial degradation<br />

by being bound to soil particles. AM infection and nodule number<br />

increased in the time following the <strong>plant</strong> growth. The saprophyte fungi population<br />

decreased when the soil was inoculated with Cry- strain, and the same<br />

effect was observed in AM infection. In another experiment carried out under<br />

axenic conditions in Petri dishes, Cry– and Cry+ strains showed an inhibitory<br />

effect against the growth of some saprophytes fungi. This fungistasis effect


7 The Functional Groups of Micro-organisms and Biotech Products 129<br />

might be explained by degrading enzymes produced (Cody 1989) or another<br />

compound that would attack the fungus cell wall or inhibit the fungal growth.<br />

Probably, these enzymes and other metabolites are produced at a vegetative<br />

phase when B. thuringiensis multiplies, nevertheless, this does not happen in<br />

the soil (Thomas et al. 2000; Vilas Bôas et al. 2000). However, much of the cell<br />

contents were released during the cell/spores lysis after inoculation and could<br />

have a suppressor effect on soil microbiota.<br />

The B. thuringiensis effect on <strong>plant</strong> growth was observed only in <strong>plant</strong>s<br />

inoculated with Cry+ strain and ICP.Although the Cry+ strain inhibited mycorrhizal<br />

colonisation, <strong>plant</strong> growth was not affected, possibly because soil fertility<br />

status and nitrogen fixation were not affected by B. thuringiensis inoculum.<br />

The same results were found by Reddy and Rhae (1989) with B. subtilis<br />

and other rhizobacteria.<br />

AM fungi were suppressed by B. thuringiensis inoculum due to the use of<br />

spores as inoculum, but due to the fact that B. thuringiensis is found in low<br />

numbers in the rhizosphere, it is difficult to explain the inhibitory effect<br />

mechanism. Other authors (Andrade et al. 1995; Bethlenfalvay et al. 1997)<br />

found the same inhibitory effect by Bacillus spp. on mycorrhizae fungi<br />

colonising pea <strong>plant</strong>s. Some strains of Bacillus spp. can probably suppress the<br />

release of AM fungi and other soil microorganism cellular contents, but this<br />

subject needs more investigation to conclude the mechanisms involved.<br />

The present data provide evidence that B. thuringiensis inoculum does not<br />

produce an effect on <strong>plant</strong> growth when soil fertility is involved. However, B.<br />

thuringiensis var. Kurstaki HD1 demonstrated inhibitory effects on some<br />

functional groups of microorganisms that could be involved in deleterious<br />

effects in the field when the nutritional condition is oligotrophic. However,<br />

the cumulative effect of protein crystal was not evaluated. It should also be<br />

emphasised that the accumulative effect of the protein crystal due to successive<br />

cultivation of Bt-transgenic <strong>plant</strong>s has not yet been assessed, but some<br />

authors have suggested that there may be a deleterious effect on the microbiota<br />

(Saxena et al. 1999) and macrofauna (Donegan et al. 1997).<br />

The groups of soil functional microorganisms may be either positively or<br />

negatively affected by B. thuringiensis products, whether produced by bacteria<br />

or transgenic <strong>plant</strong>s. Up to now, the results obtained by microbial ecologists<br />

are still preliminary, and it is clear that exhaustive studies should be carried<br />

out before releasing these <strong>plant</strong>s into the environment. The dynamic of<br />

the functional groups of microorganisms in the presence of these <strong>plant</strong>s must<br />

be understood. In addition, the accumulative effect of the crystal on these<br />

microorganism groups should be assessed together with their subsequent<br />

effects on the bio-geochemical cycles. Confidence that Bt-<strong>plant</strong>s will not damage<br />

the environment when released for intense cultivation will be obtained<br />

after the positive or negative effects they may have on the environment are<br />

established.


130<br />

Galdino Andrade<br />

References and Selected Reading<br />

Addison JA (1993) Persistence and nontarget effects of Bacillus thuringiensis in soil: a<br />

review. Can J For Res 23:2329–2342<br />

Amora-Lazcano E, Azcón R (1997) Response of sulphur cycling microorganisms to<br />

arbuscular mycorrhizal fungi in the rhizosphere of maize. Appl Soil Ecol 6:217–222<br />

Andrade G (1999) Interacciones microbianas en la rizosfera. In: Siqueira JO, Moreira<br />

FMS, Lopes AS, Guilherme LR, Faquin V, Furtinni AE, Carvalho JG (eds) Soil fertility,<br />

soil biology and <strong>plant</strong> nutrition interrelationships. Brazilian Soil Science Society/<br />

Federal University of Lavras/Soil Science Department (SBCS/UFLA/DCS), Lavras,<br />

Brazil, pp 551–575<br />

Andrade G, Azcón R, Bethlenfalvay GJ (1995) Mycorrhizae in sustainable agriculture 1.<br />

An agrosystem affecting rhizobacterium modifies <strong>plant</strong> soil responses to a mycorrhizal<br />

fungus. Appl Soil Ecol 2:195–202<br />

Andrade G, Mihara KL, Linderman RG, Bethlenfalvay GJ (1997) Bacteria from rhizosphere<br />

and hyphosphere soils of different arbuscular mycorrhizal fungi. Plant Soil<br />

192;71–79<br />

Arencibia A,Vázquez RI, Prieto D, Téllez P, Carmona ER, Coego A, Hernández L, Selman-<br />

Housein G, De La Riva GA (1997) Transgenic sugarcane <strong>plant</strong>s resistant to stem borer<br />

attack. Mol Breed 3:247–255<br />

Bethlenfalvay GJ, Andrade G, Azcón-Aguilar C (1997) Mycorrhizae in sustainable agriculture.<br />

2. Plant and soil microorganisms in nodulated and nitrate fertilized peas. Biol<br />

Fertil Soils 24:164–168<br />

Cody RM (1989) Distribution of chitinase and chitibiose in Bacillus. Curr Microbiol<br />

19:201–205<br />

Curl EA, Truelove B (1986) The rhizosphere.Advances series in agricultural sciences, vol<br />

15, Springer, Berlin Heidelberg New York, pp 288<br />

Donegan KK, Seidler RJ, Fieland VJ, Schaller DL, Palm CJ, Ganio LM, Cardwell DM, Steinbergers<br />

Y (1997) Decomposition of genetically engineered tobacco under field conditions:<br />

persistence of the proteinase inhibitor I product and effects on soil microbial<br />

respiration and protozoa, nematode and microarthropod populations. J Appl Ecol<br />

34:767–777<br />

Elliot Juhnke M, Mathre DE, Sands DC (1987) Identification and characterization of rhizosphere-competent<br />

bacteria of wheat. Appl Environ Microbiol 53:2793–2799<br />

Halverson LJ, Clayton MK, Handelsman J (1993) Population biology of Bacillus cereus<br />

UW85 in the rhizosphere of field-grown soybeans. Soil Biol Biochem 25:485–493<br />

Kim DS, Cook RJ, Weller DM (1997) Bacillus sp. L324–92 for biological control of three<br />

root diseases of wheat grown with reduced tillage. Phytopathology 87:551–558<br />

Lereclus D, Agaisse H, Grandvalet C, Salamitou S, Gominet M (2000) Regulation of toxin<br />

and virulence gene transcription in Bacillus thuringiensis. Int J Med Microbiol<br />

290:295–299<br />

Linderman RG (1992) Vesicular-arbuscular mycorrhizae and soil microbial interactions.<br />

In: Bethlenfalvay GJ, Linderman RG (eds) Mycorrhizae in sustainable agriculture.<br />

ASA Special Publication, Madison, WI, pp 45–70<br />

Marschner P, Crowley DE (1996) Physiological activity of a bioluminescent Pseudomas<br />

fluorescens (strain 2–79) in the rhizosphere of mycorrhizal and non-mycorrhizal pepper<br />

(Capsicum annum L.). Soil Biol Biochem 18:191–196<br />

Martin PAW, Travers RS (1989) Worldwide abundance and distribution of Bacillus<br />

thuringiensis isolates. Appl Environ Microbiol 55:2437–2442<br />

Mazier M, Chaufaux J, Sanchis V, Lereclus D, Giband M, Tourneur J (1997) The cryIC gene<br />

from Bacillus thuringiensis provides protection against Spodoptera littoralis in young<br />

transgenic <strong>plant</strong>s. Plant Sci 127:179–190


7 The Functional Groups of Micro-organisms and Biotech Products 131<br />

McBride KE, Svab Z, Schaaf D J (1995) Amplification of a chimeric gene in chloroplasts<br />

leads to an extraordinary level of an insecticidal protein in tobacco. Bio/technology.<br />

13:362–365<br />

Meadows MP (1993) Bacillus thuringiensis in the environment: ecology and risk assessment<br />

(1993) In: Entwistle PF, Cory JS, Bailey MJ, Higgs S (eds) Bacillus thuringiensis<br />

an environmental biopesticide: theory and practice. Wiley, Chichester, pp193–220<br />

Olsson S, Person P (1999) The composition of bacterial population in soil fractions differing<br />

in their degree of adherence to barley roots. Appl Soil Ecol 12:205–215<br />

Palm CJ, Donegan K, Harris D, Seidler RJ (1994) Quantification in soil of Bacillus<br />

thuringiensis var kurstaki d-endotoxin from transgenic <strong>plant</strong>s. Mol Ecol 3:145–151<br />

Palm CJ, Schaller DL, Donegan KK, Seidler RJ (1996) Persistence in soil of transgenic<br />

<strong>plant</strong> produced Bacillus thuringiensis var kurstaki d-endotoxin. Can J Microbiol<br />

42:1258–1262<br />

Paulitz TC, Linderman RG (1989) Interactions between fluorescent pseudomonads and<br />

VA mycorrhizal fungi. New Phytol 113:37–45<br />

Pedersen JC, Damgaard PH, Eilenberg J, Hansen BM (1995) Dispersal of Bacillus<br />

thuringiensis var. kurstaki in an experimental cabbage field. Can J Microbiol 41:118–<br />

125<br />

Peferoen M (1997) Progress and prospects for field use of Bt genes in crops. Trends<br />

Biotechnol 15:173–177<br />

Petras SF, Casida Jr LE (1985) Survival of Bacillus thuringiensis spores in soil. Appl Environ<br />

Microbiol 50:1496–1501<br />

Pruett CJH, Burges HD, Wyborn CH (1980) Effect of exposure to soil on potency and<br />

spore viability of Bacillus thuringiensis. J Invert Pathol 35:168–174<br />

Reddy MS, Rhae JE (1989) Bacillus subtilis B-2 and selected onion rhizobacteria in onion<br />

seedling rhizospheres: effects on seedling growth and indigenous rhizosphere<br />

microflora. Soil Biol Biochem 21:379–383<br />

Sanchis V (2000) Biotechnological improvement of Bacillus thuringiensis for agricultural<br />

control of insect pests: benefits and ecological implications. In: Charles JF,<br />

Delecluse A, Nielsen-Leroux C (eds) Entomophatogenic bacteria: from laboratory to<br />

field application. Kluwer Academic, Berlin<br />

Saxena D, Stotzky G (2000) Insecticidal toxin from Bacillus thuringiensis is released from<br />

roots of transgenic Bt corn in vitro and in situ. FEMS Microbiol Ecol 33:35–39<br />

Saxena D, Flores S, Stotzky G (1999) Transgenic <strong>plant</strong>s; insecticidal toxin in root exudates<br />

from Bt corn. Nature 402:480<br />

Schnepf E, Crickmore N, Van Rie J, Lereclus D, Baum J, Feitelson J, Zeigler DR, Dean DH<br />

(1998) Bacillus thuringiensis and its pesticidal crystal proteins. Microbiol Mol Biol<br />

Rev 62:775–780<br />

Schwab SM, Menge JA, Leonard RT (1983) Quantitative and qualitative effects of phosphorus<br />

on extracts and exudates of sundangrass roots in relation to vesicular-arbuscular<br />

mycorrhiza formation. Plant Physiol 73:761–765<br />

Secilia J, Bagyaraj DJ (1987) Bacteria and actinomycetes associated with pot cultures of<br />

vesicular-arbuscular mycorrhizas. Can J Microbiol 33:1067–1073<br />

Sims SR, Holden LR (1996) Insect bioassay for determining soil degradation of Bacillus<br />

thuringiensis subsp. kurstaki CryIA(b) protein in corn tissue. Environ Entomol 25:<br />

659–664<br />

Sims SR, Ream JE (1997) Soil inactivation of the Bacillus thuringiensis subsp. kurstaki<br />

CryIIA insecticidal protein within transgenic cotton tissue: laboratory microcosm<br />

and field studies. J Agric Food Chem 45:1502–1505<br />

Singsit C, Adang MJ, Lynch RE, Anderson WF, Wang A, Cardineau G, Ozias-Akins P<br />

(1997) Expression of a Bacillus thuringiensis cryIA(c) gene in transgenic peanut<br />

<strong>plant</strong>s and its efficacy against lesser cornstalk borer. Transg Res 6:169–176


132<br />

Galdino Andrade<br />

Smith RA, Couche GA (1991) The philloplane as a source of Bacillus thuringiensis variants.<br />

Appl Environ Microbiol 57:311–331<br />

Tapp H, Stotzky G (1995a) Insecticidal activity of the toxins from Bacillus thuringiensis<br />

subspecies kurstaki and tenebrionis adsorbed and bound on pure and soil clays. Appl<br />

Environ Microbiol 61:1786–1790<br />

Tapp H, Stotzky G (1995b) Dot blot enzyme-linked immunosorbent assay for monitoring<br />

the fate of insecticidal toxins from Bacillus thuringiensis in soil. Appl Environ<br />

Microbiol 61:602–609<br />

Thomas DJI, Alun J, Morgan W, Whipps JM, Saunders JR (2000) Plasmid transfer<br />

between the Bacillus thuringiensis subspecies kurstaki and tenebrionis in laboratory<br />

culture and soil and in Lepidopteran and Coleopteran larvae.Appl Environ Microbiol<br />

118–124<br />

Tomlin AD (1994) Transgenic <strong>plant</strong> release: comments on the comparative effects of<br />

agriculture and foresty practices on soil fauna. Mol Biol 3:51–52<br />

Villas-Bôas LA, Villas-Bôas GFLT, Saridakis HO, Lemos MVF, Lereclus D, Arantes OMN<br />

(2000) Survival and conjugation of Bacillus thuringiensis in a soil microcosm. FEMS<br />

Microbiol Ecol 31:255–259<br />

West AW, Burges HD, Dixon TJ,Wyborn CH (1985) Survival of Bacillus thuringiensis and<br />

Bacillus cereus spore inocula in soil: effects of pH, moisture, nutrient availability and<br />

indigenous microorganisms. Soil Biol Biochem 17:657–665<br />

Young CS, Lethbridge G, Shaw LJ, Burns RG (1995) Survival of inoculated Bacillus cereus<br />

spores and vegetative cells in non-<strong>plant</strong>ed and rhizosphere soil. Soil Biol Biochem<br />

27:1017–1026


8 The Use of ACC Deaminase-Containing Plant<br />

Growth-Promoting Bacteria to Protect Plants Against<br />

the Deleterious Effects of Ethylene<br />

Bernard R. Glick and Donna M. Penrose<br />

1 Introduction<br />

Plant growth-promoting bacteria can affect <strong>plant</strong> growth and development in<br />

two different ways: indirectly or directly (Glick 1995; Glick et al. 1999). Indirect<br />

promotion of <strong>plant</strong> growth occurs when these bacteria decrease or prevent<br />

some of the deleterious effects of a phytopathogenic organism by any<br />

one or more of several different mechanisms. In general, bacteria can directly<br />

promote <strong>plant</strong> growth by providing the <strong>plant</strong> with a compound that is synthesized<br />

by the bacterium or facilitating the uptake of nutrients.<br />

There are several ways in which <strong>plant</strong> growth-promoting bacteria can<br />

directly facilitate the proliferation of their <strong>plant</strong> hosts. They may fix atmospheric<br />

nitrogen; produce siderophores which can solubilize and sequester<br />

iron and provide it to <strong>plant</strong>s; synthesize phytohormones, including auxins,<br />

cytokinins, and gibberellins which can enhance various stages of <strong>plant</strong><br />

growth; solubilize minerals such as phosphorus; and synthesize enzymes that<br />

can modulate <strong>plant</strong> growth and development (Brown 1974; Kloepper et al.<br />

1986, 1989; Davison 1988; Lambert and Joos 1989; Patten and Glick 1996; Glick<br />

et al. 1999). A particular bacterium may affect <strong>plant</strong> growth and development<br />

using any one, or more, of these mechanisms. Moreover, many <strong>plant</strong> growthpromoting<br />

bacteria possess several properties that enable them to facilitate<br />

<strong>plant</strong> growth and, of these, may utilize different ones at various times during<br />

the life cycle of the <strong>plant</strong>.<br />

The mechanism most often invoked to explain the various effects of <strong>plant</strong><br />

growth-promoting bacteria on <strong>plant</strong>s is the production of phytohormones,<br />

most notably auxin (Brown 1974; Tien et al. 1979; Patten and Glick 1996).<br />

Since <strong>plant</strong>s as well as <strong>plant</strong> growth-promoting bacteria can synthesize auxin,<br />

it is important when assessing the consequences of treating a <strong>plant</strong> with a<br />

<strong>plant</strong> growth-promoting bacterium, to distinguish between the bacterial<br />

stimulation of <strong>plant</strong> auxin synthesis and bacterial auxin synthesis (Gaudin et<br />

al. 1994). To complicate matters, the response of <strong>plant</strong>s to auxin-producing<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


134<br />

Bernard R. Glick and Donna M. Penrose<br />

bacteria may vary from one species of <strong>plant</strong> to another, as well as according to<br />

the age of the <strong>plant</strong>.<br />

2 Ethylene<br />

In higher <strong>plant</strong>s ethylene is produced from L-methionine via the intermediates,<br />

S-adenosyl-L-methionine (SAM) and 1-aminocyclopropane-1-carboxylic<br />

acid (ACC; Yang and Hoffman 1984). The enzymes involved in this<br />

metabolic sequence are SAM synthetase, which catalyzes the conversion of<br />

methionine to SAM (Giovanelli et al. 1980); ACC synthase, which is responsible<br />

for the hydrolysis of SAM to ACC and 5¢–methylthioadenosine (Kende<br />

1989) and ACC oxidase which further metabolizes ACC to ethylene, carbon<br />

dioxide, and cyanide (John 1991).<br />

Ethylene, which is produced in almost all <strong>plant</strong>s, mediates a range of <strong>plant</strong><br />

responses and developmental steps. Ethylene is involved in seed germination,<br />

tissue differentiation, formation of root and shoot primordia, root elongation,<br />

lateral bud development, flowering initiation, anthocyanin synthesis, flower<br />

opening and senescence, fruit ripening and degreening, production of volatile<br />

organic compounds responsible for aroma formation in fruits, storage product<br />

hydrolysis, leaf and fruit abscission, and the response of <strong>plant</strong>s to biotic<br />

and abiotic stress (Matoo and Suttle 1991; Abeles et al. 1992; Frankenberger<br />

and Arshad 1995). In some instances, ethylene is stimulatory while in others it<br />

is inhibitory.<br />

The term “stress ethylene” was coined by Abeles (1973) to describe the<br />

acceleration of ethylene biosynthesis associated with biological and environmental<br />

stresses, and pathogen attack (Morgan and Drew 1997). The increased<br />

level of ethylene formed in response to trauma inflicted by chemicals, temperature<br />

extremes, water stress, ultraviolet light, insect damage, disease, and<br />

mechanical wounding (Bestwick and Ferro 1998) can be both the cause of<br />

some of the symptoms of stress (e.g., onset of epinastic curvature and formation<br />

of arenchyma), and the inducer of responses which will enhance survival<br />

of the <strong>plant</strong> under adverse conditions (e.g., production of antibiotic enzymes<br />

and phytoalexins).<br />

Chemicals have been used to control ethylene levels in <strong>plant</strong>s. The application<br />

of compounds such as rhizobitoxin, an amino acid secreted by several<br />

strains of bacteria, and its synthetic analog, aminoethoxyvinylglycine (AVG),<br />

can inhibit ethylene biosynthesis; silver thiosulfate can inhibit ethylene action,<br />

and 2-chloroethylphosphoric acid (ethephon), regarded by some researchers<br />

as “liquid ethylene”, can release ethylene (Abeles et al. 1992). Sisler and Serek<br />

(1997) discovered that cyclopropenes can block ethylene perception and are<br />

potentially useful for extending the life span of cut flowers and the display life<br />

of potted <strong>plant</strong>s. In addition, tropolone compounds were isolated from wood<br />

by Mizutani et al. (1998). These compounds, which can inhibit the growth of


wood-rotting fungi, were shown to inhibit the biosynthesis of ethylene in<br />

excised peach pits.Many of these chemicals are potentially harmful to the environment:<br />

AVG and silver thiosulfate are highly toxic in food,and silver thiosulfate<br />

causes blackspotting in flowers (Bestwick and Ferro 1998).<br />

3 ACC Deaminase<br />

8 ACC Deaminase-Containing Plant Growth-Promoting Bacteria 135<br />

In 1978, an enzyme capable of degrading ACC was isolated from Pseudomonas<br />

sp. strain ACP, and from the yeast, Hansenula saturnus (Honma and Shimomura<br />

1978; Minami et al. 1998). Since then,ACC deaminase has been detected<br />

in the fungus, Penicillium citrinum (Honma 1993) and in a number of other<br />

bacterial strains (Klee and Kishore 1992; Jacobson et al. 1994, Glick et al. 1995;<br />

Campbell and Thomson 1996) all of which originated in the soil. Many of<br />

these microorganisms were identified by their ability to grow on minimal<br />

media containing ACC as its sole nitrogen source (Honma and Shimomura<br />

1978; Klee et al. 1991; Honma 1993; Jacobson et al. 1994; Glick et al. 1995;<br />

Campbell and Thomson 1996; Burd et al. 1998; Belimov et al. 2001).<br />

Enzymatic activity of ACC deaminase is assayed by monitoring the production<br />

of either ammonia or a-ketobutyrate, the products of ACC hydrolysis<br />

(Honma and Shimomura 1978). ACC deaminase has been found only in<br />

microorganisms, and there are no microorganisms that synthesize ethylene<br />

via ACC (Fukuda et al. 1993). However, there is strong evidence that the fungus,<br />

Penicillium citrinum, produces ACC from SAM via ACC synthase, one of<br />

the enzymes of <strong>plant</strong> ethylene biosynthesis, and degrades the ACC by ACC<br />

deaminase. It appears that the ACC, which accumulates in the intracellular<br />

spaces, can induce ACC deaminase (Jia et al. 2000).<br />

ACC deaminase has been purified to homogeneity from Pseudomonas sp.<br />

strain ACP (Honma and Shimomura 1978), Hansenula saturnus (Minami et<br />

al. 1998), Penicillium citrinum (Jia et al. 1999) and partially purified from<br />

Pseudomonas sp. strain 6G5 (Klee et al. 1991) and Pseudomonas putida<br />

GR12–2 (Jacobson et al. 1994); enzyme activity is localized exclusively in the<br />

cytoplasm (Jacobson et al. 1994). The molecular mass and form is similar for<br />

the bacterial ACC deaminases. The enzyme is a trimer (Honma 1985); the size<br />

of the holoenzyme is approximately 104–105 kDa (Honma and Shimomura<br />

1978; Honma 1985; Jacobson et al. 1994) and the subunit mass is approximately<br />

36.5 kDa (Honma and Shimomura 1978; Jacobson et al. 1994). Similar<br />

subunit sizes were predicted from nucleotide sequences of cloned ACC deaminase<br />

genes from Pseudomonas sp. strains ACP (Sheehy et al. 1991) and 6G5<br />

(Klee et al. 1991), and Enterobacter cloacae UW4 (Shah et al. 1997).<br />

The molecular mass of the holoenzymes and subunits from Hansenula saturnus<br />

(69 and 40 kDa, respectively) and Penicillium citrinum (68 and 41 kDa,<br />

respectively) suggests that these ACC deaminases are dimers (Minami et al.<br />

1998; Jia et al. 1999).


136<br />

Bernard R. Glick and Donna M. Penrose<br />

K m values for the binding of ACC by ACC deaminase have been estimated<br />

for enzyme extracts from 12 microorganisms at pH 8.5. These values ranged<br />

from 1.5 to 17.4 mM (Honma and Shimomura 1978; Klee and Kishore 1992;<br />

Honma 1993) indicating that the enzyme does not have a particularly high<br />

affinity for ACC (Glick et al. 1999).<br />

ACC deaminase activity has been induced in both Pseudomonas sp. strain<br />

ACP and Pseudomonas putida GR12–2 by ACC, at levels as low as 100 nM,<br />

(Honma and Shimomura 1978; Jacobson et al. 1994); both bacterial strains<br />

were grown on a rich medium and then transferred to a minimal medium<br />

containing ACC as its sole nitrogen source. The rate of induction, similar for<br />

the enzyme from the two bacterial sources, was relatively slow: complete<br />

induction required 8–10 h. Enzyme activity increased only approximately<br />

tenfold over the basal level of activity even when the concentration of ACC<br />

increased up to 10,000-fold.<br />

Pyridoxal phosphate is a tightly bound cofactor of ACC deaminase in the<br />

amount of approximately three moles of enzyme-bound pyridoxal phosphate<br />

per mole of enzyme, or one mole per subunit (Honma 1985).<br />

Genes encoding ACC deaminase have been cloned from a number of different<br />

soil bacteria including Pseudomonas sp. strains 6G5 and 3F2 (Klee et al.<br />

1991; Klee and Kishore 1992), Pseudomonas sp. strain 17 (Campbell and<br />

Thomson 1996) Pseudomonas sp. strain ACP (Sheehy et al. 1991) and Enterobacter<br />

cloacae strains CAL2 and UW4 (Glick et al. 1995; Shah et al. 1998);<br />

yeast, Hansenula saturnus (Minami et al. 1998); and fungus, Penicillium citrinum<br />

(Jia et al. 1999).<br />

The ACC deaminase genes from Pseudomonas sp. strains 6G5 and F17, and<br />

Enterobacter cloacae strains UW4 and CAL2 all have an ORF of 1014<br />

nucleotides that encodes a protein containing 338 amino acids with a calculated<br />

molecular weight of approximately 36.8 kDa (Klee et al. 1991; Campbell<br />

and Thomson 1996; Shah et al. 1998). The genes from these strains are highly<br />

homologous to each other: at the nucleotide level 6G5, F17, UW4 and CAL2<br />

are 85–95 % identical to each other (Campbell and Thomson 1996; Shah et al.<br />

1998) and most of the dissimilarities are in the wobble position (Shah et al.<br />

1998). However, the DNA sequences from strains UW4 and CAL2 show only<br />

about 74 % homology with the sequence of the ACC deaminase gene from<br />

Pseudomonas sp. strain ACP (Sheehy et al. 1991; Shah et al. 1998).<br />

Sequence data indicate that strain UW4 contains a DNA region similar to<br />

that of the anaerobic transcription regulator, FNR, (fumarate and nitrate regulator)<br />

at positions –39 to –49 (Grichko and Glick 2000). Moreover, the ACC<br />

deaminase gene promoter in strain UW4 is under the transcriptional control<br />

of a nearby gene that has a DNA sequence similar to a leucine-responsive regulatory<br />

protein (LRP) and the LRP-like protein is transcriptionally regulated<br />

by ACC (Grichko and Glick 2000; Li and Glick 2001).<br />

When a broad host range plasmid containing the ACC deaminase gene<br />

from Enterobacter cloacae UW4 was introduced into two non<strong>plant</strong> growth-


8 ACC Deaminase-Containing Plant Growth-Promoting Bacteria 137<br />

promoting bacteria, Pseudomonas putida ATCC 17399 and Pseudomonas fluorescens<br />

ATCC 17400, by conjugational transfer, the transconjugants acquired<br />

the ability to grow on minimal media using ACC as the sole source of nitrogen,<br />

and to promote the elongation of canola roots (Shah et al. 1998).<br />

In 1998, Glick et al. proposed a model in which <strong>plant</strong> growth-promoting<br />

bacteria can lower <strong>plant</strong> ethylene levels and in turn stimulate <strong>plant</strong> growth. In<br />

this model, the <strong>plant</strong> growth-promoting bacteria bind to the <strong>surface</strong> of either<br />

the seed or root of a developing <strong>plant</strong>; in response to tryptophan and other<br />

small molecules in the seed or root exudates (Whipps 1990), the <strong>plant</strong> growthpromoting<br />

bacteria synthesize and secrete indole acetic acid (IAA; Fallik et al.<br />

1994; Patten and Glick 1996), some of which is taken up by the <strong>plant</strong>. This IAA<br />

together with endogenous <strong>plant</strong> IAA, can stimulate <strong>plant</strong> cell proliferation,<br />

<strong>plant</strong> cell elongation or induce the activity of ACC synthase to convert SAM to<br />

ACC (Kende 1993).<br />

Much of the ACC produced by this latter reaction is exuded from seeds or<br />

<strong>plant</strong> roots along with other small molecules normally present in seed or root<br />

exudates (Penrose and Glick 2001). The ACC in the exudates may be taken up<br />

by the bacteria and subsequently hydrolyzed by the enzyme, ACC deaminase,<br />

to ammonia and a-ketobutyrate. The uptake and cleavage of ACC by <strong>plant</strong><br />

growth-promoting bacteria decreases the amount of ACC outside the <strong>plant</strong>.<br />

Increasing amounts of ACC are exuded by the <strong>plant</strong> in order to maintain the<br />

equilibrium between internal and external ACC levels.As a result of the activity<br />

of ACC deaminase, the presence of the bacteria induces the <strong>plant</strong> to synthesize<br />

more ACC than it would otherwise need and as well, stimulates the<br />

exudation of ACC from the <strong>plant</strong>.<br />

Thus, <strong>plant</strong> growth-promoting bacteria are supplied with a unique source<br />

of nitrogen in the form of ACC that enables them to proliferate under conditions<br />

in which other soil bacteria may not flourish. As a result of lowering the<br />

ACC level within the <strong>plant</strong>, either the endogenous level or the IAA-stimulated<br />

level, the amount of ethylene in the <strong>plant</strong> is also reduced.<br />

Plant growth-promoting bacteria that possess the enzyme ACC deaminase<br />

and are bound to seeds or roots of seedlings, can reduce the amount of <strong>plant</strong><br />

ethylene and the extent of its inhibition on root elongation. Thus, these <strong>plant</strong>s<br />

should have longer roots and possibly longer shoots as well, inasmuch as stem<br />

elongation is also inhibited by ethylene, except in flooding-resistant <strong>plant</strong>s<br />

(Abeles et al. 1992).<br />

3.1 Treatment of Plants with ACC Deaminase Containing Bacteria<br />

Consistent with the above mentioned model, ACC deaminase activity was<br />

completely lost and the ability to promote the elongation of canola roots<br />

under gnotobiotic conditions was greatly diminished when the ACC deaminase<br />

gene (acdS) from Enterobacter cloacae UW4 was replaced, by homolo-


138<br />

Bernard R. Glick and Donna M. Penrose<br />

gous recombination, with a version of the same gene that contained a tetracycline<br />

resistance gene inserted within the coding region (Li et al. 2000). Results<br />

of an earlier study showed that ACC deaminase mutants of Pseudomonas<br />

putida GR12–2 did not promote the elongation of canola roots (Glick et al.<br />

1994). However, in those experiments, the mutants were created by chemical<br />

mutagenesis, and as a result, one could never be certain that the mutations<br />

were within the ACC deaminase structural gene per se. In the experiments by<br />

Li et al. (2000),ACC deaminase function was specifically eliminated by replacing<br />

the functional gene with an inactive version in order to demonstrate that<br />

there is no ambiguity as to the nature of the ACC deaminase minus mutants.<br />

It has been observed that both Escherichia coli and two different non<strong>plant</strong><br />

growth-promoting pseudomonads acquired the ability to significantly promote<br />

root elongation after they were transformed with a broad-host-range<br />

plasmid carrying the Enterobacter cloacae UW4 ACC deaminase gene and its<br />

upstream transcriptional regulatory region (Shah et al. 1998). Moreover, elongation<br />

of canola roots following treatment of seeds with an ACC deaminasecontaining<br />

bacterium is invariably accompanied by a decrease in the level of<br />

ACC found inside the root (Penrose et al. 2001). These observations confirm<br />

the effectiveness of ACC deaminase in lowering ACC levels.<br />

As mentioned earlier, many <strong>plant</strong>s respond to biotic and abiotic stresses by<br />

synthesizing ethylene.Among these stresses is the presence of heavy metals in<br />

the environment. It has been reasoned that at least some of the inhibitory<br />

effect of heavy metals on <strong>plant</strong> growth is the consequence of the <strong>plant</strong> synthesizing<br />

excessive amounts of stress ethylene in response to the presence of the<br />

metal, especially during early seedling development. Prior to being <strong>plant</strong>ed in<br />

metal-contaminated soil, canola and tomato seeds were treated with a heavy<br />

metal-resistant bacterium that also contained ACC deaminase. Seeds inoculated<br />

with the bacterium, Kluyvera ascorbata, and then grown in the presence<br />

of high concentrations of nickel chloride were partially protected against<br />

nickel toxicity (Burd et al. 1998). The presence of this bacterium had no measurable<br />

influence on the amount of nickel accumulated per mg dry weight in<br />

either roots or shoots of canola <strong>plant</strong>s. Therefore, the bacterial <strong>plant</strong> growthpromoting<br />

effect in the presence of nickel was not attributable to a reduction<br />

of nickel uptake by seedlings. Rather, it reflects the ability of the bacterium to<br />

lower the level of stress ethylene caused by the nickel.<br />

Transgenic canola <strong>plant</strong>s that express Enterobacter cloacae UW4 ACC<br />

deaminase were tested for the ability to proliferate and accumulate metal in<br />

the presence of high levels of arsenate in the soil. In both the presence and<br />

absence of the <strong>plant</strong> growth-promoting bacterium, Enterobacter cloacae<br />

CAL2, the transgenic <strong>plant</strong>s grew significantly larger than nontransformed<br />

<strong>plant</strong>s (Nie et al. 2002).<br />

Flooding is a common biotic stress that affects many <strong>plant</strong>s, often several<br />

times during the same growing season. Plant roots suffer a lack of oxygen as<br />

a consequence of flooding, and this in turn causes deleterious effects such as


8 ACC Deaminase-Containing Plant Growth-Promoting Bacteria 139<br />

epinasty, leaf chlorosis, necrosis, and reduced fruit yield. Two of the ACC<br />

synthase genes, LE-ACS7 and LE-ACS2, are rapidly induced in the roots of<br />

flooded tomato <strong>plant</strong>s. Of these two genes, LE-ACS7 is expressed earliest<br />

after flooding and LE-ACS2 is expressed approximately 8 h after flooding;<br />

the gene, LE-ACS7 is also involved in the early wound response of tomato<br />

leaves (Shiu et al. 1998). Since ACC oxidase-catalyzed ethylene synthesis cannot<br />

occur in the anaerobic environment of flooded roots, ACC is transported<br />

into the aerobic shoots where is converted to ethylene (Bradford and Yang<br />

1980; Else and Jackson 1998). Treatment of tomato <strong>plant</strong>s with ACC deaminase-containing<br />

<strong>plant</strong> growth-promoting bacteria significantly decreases the<br />

damage suffered by these <strong>plant</strong>s – damage that is caused by the deleterious<br />

effects of ethylene which normally occurs as a consequence of flooding<br />

(Grichko and Glick 2001). These ACC deaminase-containing <strong>plant</strong> growthpromoting<br />

bacteria can act as a sink for ACC, thereby lowering the level of<br />

ethylene that can be formed in the shoots. The tomato <strong>plant</strong>s are thus “protected”<br />

against flooding.<br />

Many of the symptoms of a diseased <strong>plant</strong> arise as a direct result of the<br />

stress imposed by the infection. That is, much of the damage sustained by<br />

<strong>plant</strong>s infected with fungal phytopathogens occurs as a result of the response<br />

of the <strong>plant</strong> to the increased levels of stress ethylene (Van Loon 1984). It has<br />

also been observed that exogenous ethylene often increases the severity of a<br />

fungal infection and, as well, ethylene synthesis inhibitors significantly<br />

decrease the severity of a fungal infection. In a study with over 60 different<br />

cultivars and breeding lines of wheat, ethylene production increased as a<br />

result of infection with the fungal phytopathogen, Septoria nodorum, and<br />

was correlated with increased <strong>plant</strong> disease susceptibility (Hyodo 1991). The<br />

damage caused by the fungal phytopathogen, Alternaria, decreased in cotton<br />

<strong>plant</strong>s by treating them with chemical inhibitors of ethylene synthesis<br />

(Bashan 1994). The levels of both ethylene and disease severity decreased in<br />

melon <strong>plant</strong>s infected by the fungal phytopathogen, Fusarium oxysporum,<br />

following treatment of the <strong>plant</strong>s with ethylene inhibitors (Cohen et al.<br />

1986). Fungal disease development increased in both cucumber <strong>plant</strong>s<br />

infected with Colletotrichum lagenarium (Biles et al. 1990) and in tomato<br />

<strong>plant</strong>s infected with Verticillium dahliae (Cronshaw and Pegg 1976) when<br />

the <strong>plant</strong>s were pretreated with ethylene. Treatment with ethylene inhibitors<br />

decreased disease severity in roses, carnations, tomato, pepper, French-bean<br />

and cucumber infected with the fungus, Botrytis cinerea (Elad 1988 and<br />

1990).<br />

Several biocontrol strains were transformed with the Enterobacter cloacae<br />

UW4 ACC deaminase gene and the effect of the transformation was assessed<br />

by using the cucumber-Pythium ultimum system (Wang et al. 2000). The<br />

results of the experiments indicated that ACC deaminase-containing biocontrol<br />

bacterial strains were significantly more effective than biocontrol strains<br />

that lacked this enzyme. Moreover, transgenic tomato <strong>plant</strong>s that express ACC


140<br />

Bernard R. Glick and Donna M. Penrose<br />

deaminase are also protected, to a significant extent, against phytopathogenmediated<br />

damage from several different phytopathogens (Lund et al. 1998;<br />

Robison et al. 2001). In effect, ACC deaminase acts synergistically with other<br />

mechanisms of biocontrol, such as the production of antibiotics or pathogenesis-related<br />

proteins, to prevent phytopathogens from damaging <strong>plant</strong>s. As<br />

with other types of stress, it is assumed that ACC deaminase can act to prevent<br />

the accumulation of ACC that would otherwise occur as a result of environmental<br />

stress.<br />

Ethylene is also a key signal in the initiation of senescence of flowers in<br />

most <strong>plant</strong>s. For example, carnation flowers produce minute amounts of ethylene<br />

until there is an endogenous rise (climacteric burst) in the level of this<br />

phytohormone. This rise in endogenous ethylene concentration is responsible<br />

for flower senescence (Mol et al. 1995), which in carnations is characterized by<br />

in-rolling of their petals.<br />

However, ethylene does not cause senescence in all flower families, and<br />

even the features of senescence that are caused by ethylene differ from <strong>plant</strong><br />

to <strong>plant</strong> (Woltering and Van Doorn 1988): for example, Caryophyllaceae (e.g.,<br />

carnations) show ethylene-mediated wilting of their petals, whereas ethylene<br />

causes petal abscission in Rosaceae (e.g., roses), but does not cause any senescence<br />

of petals in Compositae (e.g., sunflowers).<br />

Since ACC is a key element in the senescence of flower petals, a reduction in<br />

endogenous ACC would lower the amount of ethylene synthesized by the<br />

flower and delay the senescence of the petals. Many cut flowers (e.g., carnations<br />

and lilies), sold commercially, are routinely treated with the ethylene<br />

inhibitor, silver thiosulfate, which in high concentrations is potentially phytotoxic<br />

and environmentally hazardous. However, the use of ACC deaminasecontaining<br />

<strong>plant</strong> growth-promoting bacteria could be an environmentally<br />

friendly method of lowering ACC levels in cut flowers. As a first step toward<br />

determining the feasibility of this suggestion, carnation petals were treated<br />

with ACC deaminase-containing <strong>plant</strong> growth-promoting bacteria; petal<br />

senescence was delayed by several days when compared with untreated flower<br />

petals (Nayani et al. 1998).<br />

4 Conclusions<br />

There are a large number of situations in which the manipulation of ACC<br />

deaminase genes could be used to improve agricultural/horticultural/silvicultural<br />

practice. Organisms containing these genes may find use in, among<br />

other things, promoting early root development from either seeds or cuttings,<br />

increasing the life of cut flowers, protecting <strong>plant</strong>s against a wide range of<br />

environmental stresses, facilitating the production of volatile organic compounds<br />

responsible for aroma formation and phytoremediation of contaminated<br />

soils.


8 ACC Deaminase-Containing Plant Growth-Promoting Bacteria 141<br />

Currently, many consumers worldwide are reluctant to embrace the use of<br />

genetically modified <strong>plant</strong>s as sources of foods. Thus, for the foreseeable<br />

future it may be advantageous to use either natural or genetically engineered<br />

<strong>plant</strong> growth-promoting bacteria as a means of lowering <strong>plant</strong> ethylene levels<br />

rather than genetically modifying the <strong>plant</strong> itself to achieve the same end.<br />

Moreover, given the large number of different <strong>plant</strong>s, the various cultivars of<br />

those <strong>plant</strong>s and the multiplicity of genes that would need to be introduced<br />

into <strong>plant</strong>s, it is not feasible to genetically engineer all <strong>plant</strong>s to be resistant to<br />

all types of pathogens and environmental stresses. Rather, it makes a lot of<br />

sense to engineer <strong>plant</strong> growth-promoting bacteria to do this job, and the first<br />

step in this direction could well be the introduction of appropriately regulated<br />

ACC deaminase genes.<br />

Acknowledgements. The work from our laboratory that is described here was supported<br />

by grants from the Natural Science and Engineering Research Council of Canada. We<br />

wish to acknowledge the role of numerous collaborators and students in the work<br />

described here including: Chunxia Wang, Geneviève Défago, Shimon Mayak, Varvara<br />

Grichko, Jiping Li, Mary Robison, Peter Pauls, Saleh Shah, Barbara Moffatt, Genrich Burd,<br />

Seema Nayani, Gina Holguin, Cheryl Patten, Chris Jacobson and Daniel Ovakim. Thanks<br />

are also due to Andrei Belimov for sharing his results prior to their publication.<br />

References and Selected Reading<br />

Abeles FB (1973) Ethylene in <strong>plant</strong> biology. Academic Press, New York, 302 pp<br />

Abeles FB, Morgan PW, Saltveit ME Jr (1992) Ethylene in <strong>plant</strong> biology, 2nd edn. Academic<br />

Press, San Diego<br />

Bashan Y (1994) Symptom expression and ethylene production in leaf blight of cotton<br />

caused by Alternaria macrospora and Alternaria alternata alone and combined. Can<br />

J Bot 72:1574–1579<br />

Belimov AI, Safronova, VI, Sergeyeva TA, Egorova TN, Matveyeva VA, Tsyganov VE,<br />

Borisov AY, Tikhonovich IA, Kluge C, Preisfeld A, Dietz K-J, Stepanok VV (2001) Characterization<br />

of <strong>plant</strong> growth promoting rhizobacteria isolated from polluted soils and<br />

containing 1-aminocyclopropane-1-carboxylate deaminase. Can J Microbiol 27:642–<br />

652<br />

Bestwick RK, Ferro AJ (1998) Reduced ethylene synthesis and delayed fruit ripening in<br />

transgenic tomatoes expressing S-adenosylmethionine hydrolase. US Patent No:<br />

5,723,746<br />

Biles CL, Abeles FB, Wilson CL (1990) The role of ethylene in anthracnose of cucumber,<br />

Cucumis sativus, caused by Colletotrichum lagenarium. Phytopathology 80732–736<br />

Bradford KJ,Yang SF (1980) Xylem transport of 1-aminocyclopropane-1-carboxylic acid,<br />

an ethylene precursor, in waterlogged tomato <strong>plant</strong>s. Plant Physiol 65:322–326<br />

Brown ME (1974) Seed and root bacterization. Annu Rev Phytopathol 12:181–197<br />

Burd GI, Dixon DG, Glick BR (1998) A <strong>plant</strong> growth–promoting bacterium that<br />

decreases nickel toxicity in seedlings. Appl Environ Microbiol 64:3663–3668<br />

Campbell BG, Thomson JA (1996) 1-Aminocyclopropane-1-carboxylate deaminase<br />

genes from Pseudomonas strains. FEMS Microbiol Lett 138:207–210


142<br />

Bernard R. Glick and Donna M. Penrose<br />

Cohen R, Riov J, Lisker N, Katan J (1986) Involvement of ethylene in herbicide-induced<br />

resistance to Fusarium oxysporum f. sp. melonis. Phytopathology 76:1281–1285<br />

Cronshaw DK, Pegg GF (1976) Ethylene as a toxin synergist in Verticillium wilt of<br />

tomato. Physiol Plant Pathol 9:33–38<br />

Davison J (1988) Plant beneficial bacteria. Bio/Technology 6:282–286<br />

Elad Y (1988) Involvement of ethylene in the disease caused by Botrytis cinerea on rose<br />

and carnation flowers and the possibility of control. Ann Appl Biol 113:589–598<br />

Elad Y (1990) Production of ethylene in tissues of tomato, pepper, French-bean and<br />

cucumber in response to infection by Botrytis cinerea. Physiol Mol Plant Pathol<br />

36:277–287<br />

Else MA, Jackson MB (1998) Transport of 1-aminocyclopropane-1-carboxylic acid<br />

(ACC) in the transpiration stream of tomato (Lycopersicon esculentum) in relation to<br />

foliar ethylene production and petiole epinasty. Aust J Plant Physiol 25:453–458<br />

Fallik E, Sarig S, Okon Y (1994) Morphology and physiology of <strong>plant</strong> roots associated<br />

with Azospirillum. In: Okon Y (ed) Azospirillum/<strong>plant</strong> associations. CRC Press, Boca<br />

Raton, pp 77–85<br />

Frankenberger WT Jr,Arshad M (1995) Phytohormones in soil. Marcel Dekker, New York<br />

Fukuda H, Ogawa T, Tanase S (1993) Ethylene production by microorganisms. Adv<br />

Microb Physiol 35:275–306<br />

Gaudin V, Vrain T, Jouanin L (1994) Bacterial genes modifying hormonal balances in<br />

<strong>plant</strong>s. Plant Physiol Biochem 32:11–29<br />

Giovanelli J, Mudd SH, Datko AH (1980) Sulfur amino acids in <strong>plant</strong>s. In: Miflin BJ (ed)<br />

Amino acids and derivatives, the biochemistry of <strong>plant</strong>s: a comprehensive treatise. vol<br />

5. Academic Press, New York, pp 453–505<br />

Glick BR (1995) The enhancement of <strong>plant</strong> growth by free-living bacteria. Can J Microbiol<br />

41:109–117<br />

Glick BR, Jacobson CB, Schwarze MK, Pasternak JJ (1994) 1-Aminocyclopropane-1-carboxylic<br />

acid deaminase mutants of the <strong>plant</strong> growth promoting rhizobacterium<br />

Pseudomonas putida GR12-2 do not stimulate canola root elongation. Can J Microbiol<br />

40:911–915<br />

Glick BR, Karaturovíc DM, Newell PC (1995) A novel procedure for rapid isolation of<br />

<strong>plant</strong> growth promoting pseudomonads. Can J Microbiol 41:533–536<br />

Glick BR, Penrose DM, Li J (1998) A model for the lowering of <strong>plant</strong> ethylene concentrations<br />

by <strong>plant</strong> growth-promoting bacteria. J Theor Biol 190:63–68<br />

Glick BR, Patten CL, Holguin G, Penrose DM (1999) Biochemical and genetic mechanisms<br />

used by <strong>plant</strong> growth-promoting bacteria. Imperial College Press, London<br />

Grichko VP, Glick BR (2000) Identification of DNA sequences that regulate the expression<br />

of the Enterobacter cloacae UW4 1-aminocyclopropane-1-carboxylate deaminase<br />

gene. Can J Microbiol 46:1159–1165<br />

Grichko VP, Glick BR (2001) Amelioration of flooding stress by ACC deaminase-containing<br />

<strong>plant</strong> growth-promoting bacteria. Plant Physiol Biochem 39:11–17<br />

Honma M (1985) Chemically reactive sulfhydryl groups of 1-aminocyclopropane-1-carboxylate<br />

deaminase. Agric Biol Chem 49:567–571<br />

Honma M (1993) Stereospecific reaction of 1-aminocyclopropane-1-carboxylate deaminase.<br />

In: Pech JC, Latché A, Balagué C (eds) Cellular and molecular aspects of the<br />

<strong>plant</strong> hormone ethylene. Kluwer, Dordrecht, pp 111–116<br />

Honma M, Shimomura T (1978) Metabolism of 1-aminocyclopropane-1-carboxylic acid.<br />

Agric Biol Chem 42:1825–1831<br />

Hyodo H (1991) Stress/wound ethylene. In: Mattoo AK, Suttle JC (eds) The <strong>plant</strong> hormone<br />

ethylene. CRC Press, Boca Raton, pp 65–80<br />

Jacobson CB, Pasternak JJ, Glick BR (1994) Partial purification and characterization of<br />

1–aminocyclopropane-1-carboxylate deaminase from the <strong>plant</strong> growth promoting<br />

rhizobacterium Pseudomonas putida GR12-2. Can J Microbiol 40:1019–1025


8 ACC Deaminase-Containing Plant Growth-Promoting Bacteria 143<br />

Jia Y-J, Kakuta Y, Sugawara M, Igarashi T, Oki N, Kisaki M, Shoji T, Kanetuna Y, Horita T,<br />

Matsui H, Honma M (1999) Synthesis and degradation of 1–aminocyclopropane-1carboxylic<br />

acid by Penicillium citrinum. Biosci Biotechnol Biochem 63:542–549<br />

Jia Y-J, Ito H, Matsui H, Honma M (2000) 1-Aminocyclopropane-1-carboxylate (ACC)<br />

deaminase induced by ACC synthesized and accumulated in Penicillium citrinum<br />

intracellular spaces. Biosci Biotechnol Biochem 64:299–305<br />

John P (1991) How <strong>plant</strong> molecular biologists revealed a surprising relationship between<br />

two enzymes, which took an enzyme out of a membrane where it was not located, and<br />

put it into the soluble phase where it could be studied. Plant Mol Biol Rep 9:192–194<br />

Kende H (1989) Enzymes of ethylene biosynthesis. Plant Physiol 91:1–4<br />

Kende H (1993) Ethylene biosynthesis. Annu Rev Plant Physiol Plant Mol Biol<br />

44:283–307<br />

Klee HJ, Kishore GM (1992) Control of fruit ripening and senescence in <strong>plant</strong>s. US Patent<br />

No: 5,702,933<br />

Klee HJ, Hayford MB, Kretzmer KA, Barry GF, Kishore GM (1991) Control of ethylene<br />

synthesis by expression of a bacterial enzyme in transgenic tomato <strong>plant</strong>s. Plant Cell<br />

3:1187–1193<br />

Kloepper JW, Scher FM, Laliberté M, Tipping B (1986) Emergence-promoting rhizobacteria:<br />

description and implications for agriculture. In: Swinburne TR (ed) Iron,<br />

siderophores, and <strong>plant</strong> disease. Plenum, New York, pp 155–164<br />

Kloepper JW, Lifshitz R, Zablotowicz RM (1989) Free-living bacterial inocula for<br />

enhancing crop productivity. Trends Biotechnol 7:39–43<br />

Lambert B, Joos H (1989) Fundamental aspects of rhizobacterial <strong>plant</strong> growth promotion<br />

research. Trends Biotechnol 7:215–219<br />

Li J, Glick BR (2001) Transcriptional regulation of the Enterobacter cloacae UW4<br />

1–aminocyclopropane-1-carboxylate (ACC) deaminase gene (acdS). Antonie van<br />

Leewenhoek 80:255–261<br />

Li J, Ovakim DH, Charles TC, Glick BR (2000) An ACC deaminase minus mutant of Enterobacter<br />

cloacae UW4 no longer promotes root elongation. Curr Microbiol 41:101–105<br />

Lund ST, Stall, RE, Klee HJ (1998) Ethylene regulates the susceptible response to<br />

pathogen infection in tomato. Plant Cell 10:371–382<br />

Mattoo AK, Suttle JC (1991) The <strong>plant</strong> hormone ethylene. CRC Press, Boca Raton<br />

Minami R, Uchiyama K, Murakami T, Kawai J, Mikami K, Yamada T, Yokoi D, Ito H, Matsui<br />

H, Honma M (1998) Properties, sequence, and synthesis in Escherichia coli of 1aminocyclopropane-1-carboxylate<br />

deaminase from Hansenula saturnus. J Biochem<br />

123:1112–1118<br />

Mizutani F, Golam Rabbany ABM, Akiyoshi H (1998) Inhibition of ethylene production<br />

by tropolone compounds in young excised peach pits. J Jpn Soc Hortic Sci 67:166–169<br />

Mol JNM, Holton TA, Koes RE (1995) Floriculture: genetic engineering of commercial<br />

traits. Trends Biotechnol 13:350–355<br />

Morgan PW, Drew CD (1997) Ethylene and <strong>plant</strong> responses to stress. Physiol Plant<br />

100:620–630<br />

Nayani S, Mayak S, Glick BR (1998) The effect of <strong>plant</strong> growth promoting rhizobacteria<br />

on the senescence of flower petals. Ind J Exp Biol 36:836–839<br />

Nie L, Shah S, Rashid A, Burd GI, Dixon GD, Glick BR (2002) Phytoremediation of arsenate<br />

contaminated soil by transgenic canola and the <strong>plant</strong> growth-promoting bacterium<br />

Enterobacter cloacae CAL2. Plant Physiol Biochem 40:355–361<br />

Patten CL, Glick BR (1996) Bacterial biosynthesis of indole-3-acetic acid. Can J Microbiol<br />

42:207–220<br />

Penrose DM, Glick BR (2001) Levels of 1–aminocyclopropane-1-carboxylic acid (ACC)<br />

in exudates and extracts of canola seeds treated with <strong>plant</strong> growth-promoting bacteria.<br />

Can J Microbiol 47:368–372


144<br />

Bernard R. Glick and Donna M. Penrose<br />

Penrose DM, Moffatt BM, Glick BR (2001) Determination of 1-aminocyclopropane-1carboxylic<br />

acid (ACC) to assess the effects of ACC deaminase-containing bacteria on<br />

roots of canola seedlings. Can J Microbiol 47:77–80<br />

Robinson MM, Shah S, Tamot B, Pauls PK, Moffatt BA, Glick BR (2001) Reduced symptoms<br />

of Verticillium wilt in tomato <strong>plant</strong>s transformed with ACC deaminase to control<br />

ethylene biosynthesis. Mol Plant Pathol 2:135–145<br />

Shah S, Li J, Moffatt BA, Glick BR (1997) ACC deaminase genes from <strong>plant</strong> growth promoting<br />

bacteria. In: Ogoshi A, Kobayashi K, Homma Y, Kodama F, Kondo N, Akino S<br />

(eds) Plant growth-promoting rhizobacteria: present status and future prospects.<br />

OECD, Paris, pp 320–324<br />

Shah S, Li J, Moffatt BA, Glick BR (1998) Isolation and characterization of ACC deaminase<br />

genes from two different <strong>plant</strong> growth promoting rhizobacteria. Can J Microbiol<br />

44:833–843<br />

Sheehy RE, Honma M, Yamada M, Sasaki T, Martineau B, Hiatt WR (1991) Isolation,<br />

sequence, and expression in Escherichia coli of the Pseudomonas sp. strain ACP gene<br />

encoding 1-aminocyclopropane-1-carboxylate deaminase. J Bacteriol 173:5260–5265<br />

Shiu OY, Oetiker JH, Yip WK, Yang SF (1998) The promoter of LE-ACS7, an early flooding-induced<br />

1-aminocyclopropane carboxylate synthase gene of the tomato, is tagged<br />

by a Sol3 transposon. Proc Natl Acad Sci USA 95:10334–10339<br />

Sisler EC, Serek M (1997) Inhibitors of ethylene responses in <strong>plant</strong>s at the receptor level:<br />

recent developments. Physiol Plant 100:577–582<br />

Tien TM, Gaskins MH, Hubell DH (1979) Plant growth substances produced by Azospirillum<br />

brasilense and their effect on the growth of pearl millet (Pennisetum americanum<br />

L). Appl Environ Microbiol 37:1016–1024<br />

Van Loon LC (1984) Regulation of pathogenesis and symptom expression in diseased<br />

<strong>plant</strong>s by ethylene. In: Fuchs Y, Chalutz E (eds) Ethylene: biochemical, physiological<br />

and applied aspects. Martinus Nijhoff/Dr. W. Junk, The Hague, pp 171–180<br />

Wang C, Knill E, Glick BR, Défago G (2000) Effect of transferring 1–aminocyclopropane-<br />

1-carboxylic acid (ACC) deaminase genes into Pseudomonas fluorescens strain CHA0<br />

and its gacA derivative CHA96 on their growth-promoting and disease-suppressive<br />

capacities. Can J Microbiol 46:898–907<br />

Whipps JM (1990) Carbon utilization. In: Lynch JM (ed) The rhizosphere. Wiley Interscience,<br />

Chichester, pp 59–97<br />

Woltering EJ, Van Doorn WG (1988) Role of ethylene in senescence of petals – morphological<br />

and taxonomical relationships. J Exp Bot 39:1605–1616<br />

Yang SF, Hoffman NE (1984) Ethylene biosynthesis and its regulation in higher <strong>plant</strong>s.<br />

Annu Rev Plant Physiol 35:155–189


9 Interactions Between Epiphyllic Microorganisms<br />

and Leaf Cuticles<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

1 Introduction<br />

Leaves of higher <strong>plant</strong>s are exposed to the atmosphere. Due to the pronounced<br />

two-dimensional structure of leaves, the <strong>surface</strong> area of <strong>plant</strong>s is significantly<br />

enlarged. This allows an efficient absorption of visible light used in photosynthesis<br />

and it supports the rapid gas exchange of carbon dioxide and oxygen,<br />

occurring across stomates. With most leaves, stomates representing small<br />

pores, cover only between 0.5 to 1 % of the total leaf <strong>surface</strong> area (Larcher<br />

1996), whereas the largest part of the leaf <strong>surface</strong> is covered by the <strong>plant</strong> cuticle<br />

forming the major interface between the leaves and the atmosphere (Kerstiens<br />

1996). The cuticle developed during evolution when <strong>plant</strong>s moved from<br />

their aqueous habitats to the dry land. It protects land living <strong>plant</strong>s from desiccation.<br />

The water potential in the atmosphere is nearly always lower than the<br />

water potential of <strong>plant</strong>s, which causes a constant driving force for the flow of<br />

water from the <strong>plant</strong> body to the atmosphere (Nobel 1991). Without the cuticle<br />

forming a very efficient transport barrier for the passive diffusion of water<br />

from the turgescent <strong>plant</strong> to the atmosphere, most of the land-living higher<br />

<strong>plant</strong>s would never be able to survive.<br />

Besides this major function as a watertight barrier, the <strong>plant</strong> cuticle also<br />

limits the leaching of ions and nutrients from the leaf interior (Tukey 1970),<br />

and it forms a mechanical barrier for most microorganisms trying to infect<br />

the living leaf tissues (Mendgen 1996; Schafer 1998). Looking at the <strong>surface</strong>s<br />

of healthy, green leaves collected in the environment in their natural habitats<br />

using different microscopical techniques (fluorescence microscopy, confocal<br />

laser scanning microscopy or scanning electron microscopy), it becomes<br />

obvious that leaf <strong>surface</strong>s are always covered by epiphyllic microorganisms to<br />

a certain degree (Fig. 1). This epiphyllic flora is composed of bacteria, yeasts<br />

and filamentous fungi belonging to different systematic categories (Morris et<br />

al. 1996). The degree of coverage strongly depends on a series of parameters<br />

like the <strong>plant</strong>s species, the structure of the leaf <strong>surface</strong>, the habitat of the <strong>plant</strong><br />

and the age of the leaf (Preece and Dickinson 1971; Dickinson and Preece<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


146<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

Fig. 1. Micrograph (SEM) of the lower stomatous leaf side of walnut (Juglans regia L.).A<br />

dense epiphyllic microflora of bacterial (smaller rod-like cells) and yeast cells (larger<br />

spherical cells) in a depression of the lower leaf <strong>surface</strong> can be seen<br />

1976). Population densities of leaf <strong>surface</strong> microorganisms are characterised<br />

by large fluctuations, since they are strongly dependent on environmental<br />

conditions (Fokkema and van den Heuvel 1986). Rapid changes from<br />

favourable environmental conditions (e.g., high humidity and low irradiance)<br />

to unfavourable conditions (e.g., low humidity, high irradiance and high temperatures)<br />

as they can naturally occur within hours or days are followed by<br />

rapid changes in the density and the number of epiphyllic microorganisms<br />

(Leben 1988).<br />

Thus, it can be concluded that the phyllosphere forms a characteristic<br />

habitat for microorganisms, a fact which has largely been neglected in the<br />

past (Beattie and Lindow 1995). Plant biologists normally investigate the<br />

structure and the function of the cuticle as a barrier for water and organic<br />

compounds (Schönherr and Riederer 1989), whereas <strong>plant</strong> pathologists are<br />

mostly interested in the interaction between pathogens and the living epidermal<br />

cells or the cell walls (Dixon and Lamb 1990). Environmental microbiologists<br />

are mostly interested in describing the epiphyllic population<br />

dynamics, the species composition and their potential as antagonists


9 Interactions Between Epiphyllic Microorganisms and Leaf Cuticles 147<br />

Fig. 2. A scheme of the phyllosphere<br />

as a habitat for<br />

microorganisms showing most<br />

of the relevant climatic and<br />

<strong>plant</strong> parameters determining<br />

the living conditions of the leaf<br />

<strong>surface</strong><br />

atmosphere<br />

waxes<br />

cutin<br />

epidermal<br />

wall<br />

wetting<br />

biofilm<br />

penetration<br />

leaching<br />

(Andrews 1992; Jacques and Morris 1995; Fiss et al. 2000). However, looking<br />

at the leaf <strong>surface</strong> as a microhabitat with very specific boundary conditions,<br />

investigations of the parameters, limitations and interactions between the<br />

lipophilic leaf <strong>surface</strong> and the microorganisms have rarely been carried out<br />

(Fig. 2). This habitat is characterised by an extreme microclimate due to<br />

large variations in climatic parameters like light intensity and temperature<br />

(Andrews and Harris 2000). Due to specific physical, chemical and biological<br />

properties of leaf <strong>surface</strong>s, the phyllosphere is also dominated by a low<br />

availability of water and nutrients (Schönherr and Baur 1996; Beattie and<br />

Lindow 1999). Investigating the microbial ecology of the phyllosphere will be<br />

a combined approach including <strong>plant</strong> ecophysiological and microbiological<br />

tools. In the following, several important aspects of the microbial ecology of<br />

the phyllosphere will be discussed and selected examples for the interactions<br />

occurring between epiphyllic microorganisms and the leaf <strong>surface</strong> will be<br />

given.<br />

2 Physical and Chemical Parameters of the Phyllosphere<br />

water<br />

vapor<br />

gradient<br />

The <strong>plant</strong> cuticle covering the leaf <strong>surface</strong> is a lipophilic, extracellular<br />

biopolymer. It is composed of the cutin polymer (Kolattukudy 2001), which is<br />

a polyester of esterified hydroxy fatty acids, and of cuticular waxes (Walton<br />

1990), deposited as monomeric compounds to the cutin polymer (intracuticular<br />

waxes) and to the cutin <strong>surface</strong> (epicuticular waxes). Cuticular waxes are<br />

basically linear long chain aliphatic compounds of different chain length and<br />

different substance classes. Typical wax constituents are alkanes, aldehydes,<br />

primary and secondary alcohols, acids and esters composed of the respective<br />

acids and alcohols (Bianchi 1995). Besides these linear long-chain aliphatics,


148<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

cuticular waxes of some <strong>plant</strong> species are dominated by a large degree by<br />

triterpenoic acids and triterpenols (Gülz 1994; Markstädter et al. 2000). The<br />

chemical environment which will be sensed by epiphyllic microorganisms living<br />

on leaf <strong>surface</strong>s will be the outermost layer of wax compounds forming<br />

the true interface between the leaf and the atmosphere. For this reason, analytical<br />

chemistry such as gas chromatography coupled to different detector<br />

systems (FID and MS) is an important tool for describing the chemical environment<br />

of the leaf <strong>surface</strong> (Riederer and Markstädter 1996).<br />

Epicuticular waxes often form characteristic three-dimensional structures<br />

like platelets (Fig. 3), rods, ribbons or filaments (Jeffree 1986). This significantly<br />

increases leaf <strong>surface</strong> roughness. As a consequence, water drops, small<br />

particles, as well as spores and bacterial cells located on the tips of these crystals<br />

strongly reduce the attachment of particles to the leaf <strong>surface</strong>. Thus, rain<br />

or water can simply wash off these loosely attached particles on rough leaf<br />

<strong>surface</strong>s (Barthlott and Neinhuis 1997). Both parameters, the very hydrophobic<br />

nature of cutin and wax and the often very pronounced roughness of the<br />

leaf <strong>surface</strong>, are responsible for the fact that leaf <strong>surface</strong>s are a very dry habitat<br />

since water is very efficiently rejected (Holloway 1970). Nevertheless, with<br />

increasing leaf age in most cases epicuticular wax crystals tend to disappear,<br />

probably due to erosion, and the factor roughness will become less significant<br />

Fig. 3. Micrograph (SEM) of the upper astomatous leaf side of oak (Quercus robur L.)<br />

showing the dense accumulation of epicuticular wax crystals. The crystals, having the<br />

shape of small platelets, are oriented in a rectangular angle to the leaf <strong>surface</strong> leading to<br />

a pronounced <strong>surface</strong> roughness


9 Interactions Between Epiphyllic Microorganisms and Leaf Cuticles 149<br />

for epiphyllic microorganisms trying to colonise older leaves (Neinhuis and<br />

Barthlott 1998).<br />

In addition, cuticular waxes are responsible for establishing the transport<br />

barrier of the <strong>plant</strong> cuticle (Riederer and Schreiber 1995). Extraction of cuticular<br />

waxes with appropriate solvents such as chloroform increased cuticular<br />

transport for water and dissolved compounds by two to three orders of magnitude<br />

(Schönherr and Riederer 1989). At room temperature, waxes form<br />

solid partially crystalline aggregates with a high degree of order (Reynhardt<br />

and Riederer 1994; Schreiber et al. 1997) and, thus, they efficiently seal the<br />

amorphous cutin polymer, which itself is fairly permeable for water and dissolved<br />

compounds (Schönherr and Riederer 1989). The structure of the cuticle<br />

can best be compared to the technical principle realised in wax-coated<br />

papers, where the wax establishes a transpiration barrier, whereas the cellulose<br />

polymer forms a stable matrix for deposition of the wax (Fox 1958). Thus,<br />

although epiphyllic microorganisms live on a substrate, below which the best<br />

conditions in terms of water supply and nutrient concentrations exist, the leaf<br />

<strong>surface</strong> is an environment with extremely unfavourable conditions, because<br />

this reservoir below the cuticle is rarely accessible to leaf <strong>surface</strong> microorganisms<br />

under normal conditions (Schönherr and Baur 1996).<br />

3 Leaf Surface Colonisation and Species Composition<br />

Freshly emerging leaves are basically clean, unwettable and they often have a<br />

pronounced roughness due to epicuticular waxes crystals (Neinhuis and<br />

Barthlott 1998). Pronounced succession in leaf <strong>surface</strong> colonisation has been<br />

described by several authors (Ercolani 1991; Kinkel 1991; Blakeman 1993).<br />

Normally, the first detectable microorganisms are bacteria starting to<br />

colonise the leaf <strong>surface</strong> (Blakeman 1991). Later in the season, yeasts become<br />

more and more abundant in the phyllosphere due to additional nutrients like<br />

pollen and high amounts of sugars becoming available by the activity of<br />

aphids (Stadler and Müller 1996). Towards the end of the season, especially<br />

with deciduous trees, leaf <strong>surface</strong>s are often densely covered with filamentous<br />

fungi. This might be related to decreasing barrier properties of the cuticle due<br />

to leaf ageing.<br />

Once epiphyllic microorganisms have succeeded in colonising the leaf<br />

<strong>surface</strong> they are strongly attached to the <strong>surface</strong> (Romantschuk 1992) and<br />

can rarely be removed even after excessive washing (Schreiber and Schönherr<br />

1993). They often tend to protect themselves in an extracellular matrix<br />

(Beattie and Lindow 1999), and it has also been shown that biofilms, containing<br />

different bacterial species, may develop in the phyllosphere (Morris<br />

et al. 1997, 1998). The species living in the leaf <strong>surface</strong> belong to diverse taxonomic<br />

groups. Most abundant bacterial species which have been described<br />

belonged to the genera Corynebacterium, Erwinia, Pseudomonas, Xan-


150<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

thomonas and Bacillus (Ercolani 1991; Morris et al. 1998). Cladosporium,<br />

Alternaria and Aureobasidium have been described as being abundant filamentous<br />

fungal species and Cryptococcus and Sporobolomyces were<br />

described as abundant yeast species in the phyllosphere (Andrews and Harris<br />

2000; Blakeman 1993).<br />

However, it must be mentioned that the description of the epiphyllic<br />

microflora up to now is exclusively based on an identification of the species<br />

after cultivation on standard media. However, it is well known today from<br />

environmental <strong>microbiology</strong> that many bacterial species cannot be cultivated<br />

with standard techniques (Amann et al. 1995). PCR-based approaches<br />

showed that the bacterial species composition of aquatic environments, but<br />

also of soil and rhizosphere communities, is much more complex and<br />

diverse as it was originally concluded from cultivation-based approaches<br />

(Marilley et al. 1998; Tiedje et al. 1999; Ogram 2000). A similar approach has<br />

rarely been carried out in the leaf <strong>surface</strong> and it is absolutely necessary in<br />

leaf <strong>surface</strong> <strong>microbiology</strong> in future in order to obtain a more realistic and<br />

complete picture of the species composition in the phyllosphere. Results of<br />

one of the first approaches, comparing the bacterial species identified using<br />

PCR versus cultivation-based techniques (Yang et al. 2001), in fact, yielded<br />

two quite different pictures of the species composition of the phyllosphere.<br />

This proves that our knowledge of the species composition on leaf <strong>surface</strong>s<br />

obtained from cultivation-based techniques is still rather limited and needs<br />

further research.<br />

4 Alteration of Leaf Surface Wetting<br />

Investigations of the seasonal development of leaf <strong>surface</strong> wetting have shown<br />

several times that leaf <strong>surface</strong>s become more and more wettable with increasing<br />

leaf age (Cape 1983; Turunen and Huttunen 1989; Cape and Percy 1993;<br />

Neinhuis and Barthlott 1998). This was normally attributed to chemical<br />

changes in the physico-chemical properties of the waxy leaf <strong>surface</strong> at the<br />

leaf/atmosphere interface caused by environmental pollution. In addition, it<br />

was shown that wax erosion due to the constant exposure of the leaf <strong>surface</strong> to<br />

wind, rain and the deposition of dust particles from the atmosphere to the leaf<br />

<strong>surface</strong> also occurs (van Gardingen et al. 1991), and may be further contributed<br />

to these observed increases in wetting. However, epiphyllic microorganisms<br />

as a further parameter contributing to an increased wetting of the<br />

leaf <strong>surface</strong> may not be neglected here.<br />

In simple model experiments, silanised glass <strong>surface</strong>s, which are rarely wetted<br />

by water due to their high hydrophobicity, were colonised by bacteria and<br />

wetting properties were quantified by measuring contact angles (Knoll and<br />

Schreiber 1998, 2000). From these experiments, it became obvious that already<br />

at a coverage of 10 % of the total <strong>surface</strong>, contact angles decreased by 25°


9 Interactions Between Epiphyllic Microorganisms and Leaf Cuticles 151<br />

(Fig. 4A). Maximum effects were a decrease of the contact angle from about<br />

95° to 30° at a coverage of 70 %. Similar results were obtained when clean ivy<br />

leaf <strong>surface</strong>s were colonised by bacteria. A bacterial coverage of 10 % of the<br />

leaf <strong>surface</strong> resulted in a decrease in the contact angle by 25° and only a 25 %<br />

coverage resulted in decrease from 90° to 40° (Fig. 4B). These experiments<br />

clearly proved that leaf <strong>surface</strong> wetting properties can be altered to a large<br />

degree by the presence of epiphyllic microorganisms.<br />

Using scanning electron microscopy, gas chromatography and contact<br />

angle measurements in parallel, investigation of needle (Abies grandis Lindl.)<br />

and leaf <strong>surface</strong>s (Juglans regia L.) during one season supported this observation<br />

(Schreiber 1996; Knoll and Schreiber 1998). The pronounced increase in<br />

Contact angle (degree)<br />

contact angle (degree)<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

80<br />

76<br />

72<br />

68<br />

64<br />

60<br />

56<br />

Silanized<br />

glass<br />

pH 3.0<br />

<strong>surface</strong><br />

pH 9.0<br />

20/6/1994<br />

20/7/1994<br />

15/8/1994<br />

Abies<br />

grandis<br />

current year needles<br />

pH 3.0<br />

pH 9.0<br />

15/10/1994<br />

180 200 220 240 260 280 300 320<br />

day of the year 1994<br />

A<br />

(a) t = 6 h<br />

0 10 20 30 40 50 60 70 80<br />

Area covered by P. fluorescens (%)<br />

15/11/1994<br />

C<br />

Contact angle (degree)<br />

Contact angle (degree)<br />

80<br />

75<br />

70<br />

65<br />

60<br />

55<br />

50<br />

45<br />

40<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

Hedera<br />

helix<br />

0 5 10 15 20 25 30<br />

Area covered by epiphytic micro-organisms (%)<br />

5<br />

6<br />

/<br />

9<br />

0<br />

/<br />

2<br />

5<br />

5<br />

7<br />

/<br />

9<br />

0<br />

/<br />

1<br />

9<br />

Juglans<br />

regia<br />

8<br />

/<br />

9<br />

0<br />

/<br />

2<br />

1<br />

160 180 200 220 240 260 280 300<br />

5<br />

Julian day (1995)<br />

Fig. 4. Degree of wetting of a silanised glass <strong>surface</strong> (A) and an ivy (Hedera helix L.) leaf<br />

<strong>surface</strong> (B) as a function of the coverage of the leaf <strong>surface</strong> with epiphyllic microorganisms.<br />

Seasonal increase of needle (Abies grandis Lindl.) <strong>surface</strong> (C) and leaf (Juglans<br />

regia L.) <strong>surface</strong> (D) wetting due to increasing amounts of microorganisms growing in<br />

the phyllosphere<br />

5<br />

9<br />

/<br />

9<br />

0<br />

/<br />

1<br />

1<br />

B<br />

D<br />

5<br />

0<br />

/<br />

9<br />

1<br />

/<br />

0<br />

5


152<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

the needle and the leaf <strong>surface</strong> wetting properties quantified by contact angle<br />

measurements (Fig. 4C, D) was always in parallel with a significant increase in<br />

the colonisation of the needle and leaf <strong>surface</strong>s with epiphyllic microorganisms<br />

as seen in scanning electron microscopy. However, changes in the qualitative<br />

and quantitative wax composition, measured by gas chromatography,<br />

were not at all correlated with the changes in the wetting properties of the leaf<br />

<strong>surface</strong>s (Schreiber 1996; Knoll and Schreiber 1998).<br />

From this, it is evident that leaf <strong>surface</strong> microorganisms have the ability to<br />

significantly change leaf wettability by altering the physico-chemical properties<br />

of leaf <strong>surface</strong>s. This is probably an important ecological strategy of epiphyllic<br />

microorganisms improving the living conditions in their environment.<br />

Increased wetting will increase the water availability in the leaf <strong>surface</strong>,<br />

which in turn is highly favourable for the microorganisms living there. Furthermore,<br />

increased wetting will also more easily lead to the formation of thin<br />

water films, which is necessary in order to dissolve substances leaching from<br />

the apoplast to the leaf <strong>surface</strong>. As a consequence, the availability and the<br />

amount of nutrients in the phyllosphere will increase as well, which again is<br />

favourable for epiphyllic microorganisms.<br />

5 Interaction of Bacteria with Isolated Plant Cuticles<br />

It is generally believed that <strong>plant</strong> cuticles form more or less impermeable<br />

mechanical barriers for bacteria (Agrios 1995). Whereas fungi may have the<br />

ability to penetrate the cuticle using extracellular enzymes (Schäfer 1998), for<br />

bacteria an infection of the leaf tissue only seems to be possible via stomates<br />

or hydathodes forming natural openings or via artificial openings like cracks<br />

caused by injuries. In order to test this hypothesis, isolated cuticular membranes<br />

from different <strong>plant</strong> species were mounted in transpiration chambers<br />

and cuticular water permeability was quantified as a measure of the effect of<br />

microorganisms on leaf <strong>surface</strong> barrier properties.<br />

Cuticular water permeability of selected species (Vinca major L., Hedera<br />

helix canariensis L. and Prunus laurocerasus L.) was measured before and<br />

after inoculation with Pseudomonas fluorescens, which was chosen as a characteristic<br />

and representative epiphyllic microorganism. With all three investigated<br />

species, cuticular water permeability significantly increased by factors<br />

between 40 to 60 % after inoculation with P. fluorescens for 10–12 days (Fig. 5).<br />

In parallel to the observed increase in cuticular water permeability, it was<br />

always observed that the bacteria had successfully penetrated the cuticle,<br />

since bacteria were growing on the inner side of the isolated cuticle, which<br />

was sterile at the beginning of the experiment. From this observation, it must<br />

be concluded that the bacteria had induced additional defects to the transport<br />

barrier of the cuticle, leading to increased rates of water permeability as well<br />

as paths for penetrating the cuticle (Knoll 1998).


However, at the moment, the mechanism as to how this was achieved by the<br />

bacteria is not clear. One possibility might be a dissolution of the cutin polymer<br />

by extracellular bacterial enzymes. Alternatively, one could also image a<br />

pure physical basis. It was shown in the past that cuticular permeability for<br />

water and many dissolved compounds can be increased by surfactants<br />

(Riederer and Schönherr 1990). A similar mechanism might be used by<br />

microorganisms, since for many of them it has been shown that they are able<br />

to synthesise biosurfactants (Persson et al. 1988; Bunster et al. 1989; Karanth<br />

et al. 1999). Moreover, it also may not be forgotten that in reality there are living<br />

epidermal cells below the cuticle. They probably will significantly contribute<br />

to inhibiting leaf <strong>surface</strong> microorganisms from penetrating the cuticle,<br />

which is not the case in the artificial system using isolated cuticular<br />

membranes. Future work will have to concentrate on this important question<br />

of the interaction between epiphyllic microorganisms and the <strong>plant</strong> cuticle.<br />

6 Conclusions<br />

9 Interactions Between Epiphyllic Microorganisms and Leaf Cuticles 153<br />

Fig. 5. Interaction<br />

between Pseudomonas<br />

fluorescens growing on<br />

isolated cuticles of different<br />

<strong>plant</strong> species and<br />

cuticular water permeability.<br />

The effect, which<br />

was calculated from the<br />

ratio of cuticular water<br />

permeability after inoculation<br />

divided by cuticular<br />

transpiration before<br />

inoculation, indicates the<br />

relative increase in cuticular<br />

water permeability<br />

after inoculation with<br />

bacteria<br />

effect (P2/P1)<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0.0<br />

Pseudomonas fluorescens<br />

control<br />

Vinca<br />

major<br />

Hedera<br />

helix can.<br />

Prunus<br />

laurocerasus<br />

In conclusion, it must be stated that lipophilic <strong>surface</strong>s of leaves form microhabitats<br />

for many microorganisms, although living conditions in terms of<br />

water and nutrient availability and climatic conditions in the phyllosphere are<br />

far from optimal. Specific interactions between epiphyllic microorganisms<br />

and the <strong>plant</strong> cuticle, leading to increased leaf <strong>surface</strong> wetting and elevated<br />

rates of cuticular permeability, have been shown to occur. Nevertheless, there<br />

is still a series of questions which deserves further attention in future<br />

research. Using molecular biological tools, a more realistic description of the


154<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

diversity of the species composition in the phyllosphere must be achieved.<br />

Physiological experiments will have to analyse in more detail the mechanisms<br />

forming the basis for the different interactions occurring between epiphyllic<br />

microorganisms and the <strong>plant</strong> cuticle. Furthermore, an important question is<br />

to what extent the aggregation of different epiphyllic species forming biofilms<br />

increases their ecological fitness in the phyllosphere. Answering these questions<br />

in the future will significantly help to improve our knowledge of the<br />

microbial ecology of the phyllosphere.<br />

Acknowledgements. The authors gratefully acknowledge financial support of this work<br />

by the Deutsche Forschungsgemeinschaft and the FCI.<br />

References and Selected Reading<br />

Agrios GN (1995) Plant pathology. San Diego, Academic Press<br />

Amann R, Ludwig W, Schleifer KH (1995) Phylogenetic identification and in situ detection<br />

of individual microbial cells without cultivation. Microbiol Rev 59:143–169<br />

Andrews JH (1992) Biological control in the phyllosphere. Annu Rev Phytopathol<br />

30:603–635<br />

Andrews JH, Harris RF (2000) The ecology and biogeography of microorganisms of<br />

<strong>plant</strong> <strong>surface</strong>s. Annu Rev Phytopathol 38:145–180<br />

Barthlott W, Neinhuis C (1997) Purity of the sacred lotus, or escape from contamination<br />

in biological <strong>surface</strong>s. Planta 202:1–8<br />

Beattie GA, Lindow SE (1995) The secret life of foliar bacterial pathogens on leaves.Annu<br />

Rev Phytopathol 33:145–172<br />

Beattie GA, Lindow SE (1999) Bacterial colonization of leaves: a spectrum of strategies.<br />

Phytopathology 89:353–359<br />

Bianchi G (1995) Plant waxes. In: Hamilton RJ (ed) Waxes: chemistry, molecular biology<br />

and functions. The Oily Press, Dundee, pp 175–222<br />

Blakeman JP (1991) Foliar bacterial pathogens: epiphytic growth and interactions on<br />

leaves. J Appl Bacteriol 70:49–59<br />

Blakeman JP (1993) Pathogens in the foliar environment. Plant Path 42:479–493<br />

Bunster L, Fokkema NJ, Schippers B (1989) Effect of <strong>surface</strong>-active Pseudomonas sp. on<br />

leaf wettability. Appl Environ Microbiol 55:1340–1345<br />

Cape JN (1983) Contact angles of water droplets on needles of Scots pine (Pinus sylvestris)<br />

growing in polluted atmospheres. New Phytol 93:293–299<br />

Cape JN, Percy KE (1993) Environmental influences on the development of spruce needle<br />

cuticles. New Phytol 125:787–799<br />

Dickinson CH, Preece TF (1976) Microbiology of aerial <strong>plant</strong> <strong>surface</strong>s. Academic Press,<br />

London<br />

Dixon RA, Lamb CJ (1990) Molecular communication in interactions between <strong>plant</strong>s<br />

and microbial pathogens. Annu Rev Plant Phys Plant Mol Biol 41:339–367<br />

Ercolani GL (1991) Distribution of epiphytic bacteria on olive leaves and the influence of<br />

leaf age and sampling time. Microb Ecol 21:35–48<br />

Fiss M, Kucheryava N, Schonherr J, Kollar A, Arnold G, Auling G. (2000) Isolation and<br />

characterization of epiphytic fungi from the phyllosphere of apple as potential biocontrol<br />

agents against apple scab (Venturia inaequalis). J Plant Dis Prot 107:1–11


9 Interactions Between Epiphyllic Microorganisms and Leaf Cuticles 155<br />

Fokkema NJ, van den Heuvel J (1986) Microbiology of the phyllosphere.Academic Press,<br />

New York<br />

Fox RC (1958) The relationship of wax crystal structure to the water vapor transmission<br />

rate of wax films. Tech Assoc Pulp Paper Ind 41:283–289<br />

Gülz PG (1994) Epicuticular leaf waxes in the evolution of the <strong>plant</strong> kingdom. J Plant<br />

Physiol 143:453–464<br />

Holloway PJ (1970) Surface factors affecting the wetting of leaves. Pest Sci 1:156–163<br />

Jacques MA, Morris CE (1995) A review of issues related to the quantification of bacteria<br />

from the phyllosphere. FEMS Microbiol Ecol 18:1–14<br />

Jeffree CE (1986) The cuticle, epicuticular waxes and trichomes of <strong>plant</strong>s, with reference<br />

to their structure, functions and evolution. In: Juniper BE, Southwood R (eds) Insects<br />

and <strong>plant</strong> <strong>surface</strong>s. Edward Arnold, London, pp 23–64<br />

Karanth NGK, Deo PG, Veenanadig NK (1999) Microbial production of biosurfactants<br />

and their importance. Curr Sci 77:116–126<br />

Kerstiens G (1996) Signalling across the divide: a wider perspective of cuticular structure–function<br />

relationships. Trends Plant Sci 1: 125–129<br />

Kinkel LL (1991) Microbial population dynamics on leaves. Annu Rev Phytopathol<br />

35:327–347<br />

Knoll D (1998) Die Bedeutung der Kutikula bei der Interaktion zwischen epiphyllen<br />

Mikroorganismen und Blattoberflächen. PhD Thesis, University of Würzburg, Germany<br />

Knoll D, Schreiber L (1998) Influence of epiphytic micro-organisms on leaf wettability:<br />

wetting of the upper leaf <strong>surface</strong> of Juglans regia and of model <strong>surface</strong>s in relation to<br />

colonization by microorganisms. New Phytol 140:271–282<br />

Knoll D, Schreiber L (2000) Plant-microbe interactions: wetting of ivy (Hedera helix L.)<br />

leaf <strong>surface</strong>s in relation to colonization by epiphytic microorganisms. Microb Ecol<br />

41:33–42<br />

Kolattukudy PE (2001) Polyesters in higher <strong>plant</strong>s. Adv Biochem Engin Biotech 71:1–49<br />

Larcher W (1996) Physiological <strong>plant</strong> ecology: ecophysiology and stress physiology of<br />

functional groups. Springer, Berlin Heidelberg New York<br />

Leben C (1988) Relative humidity and the survival of epiphytic bacteria with buds and<br />

leaves of cucumber <strong>plant</strong>s. Phytopathology 78:179–185<br />

Marilley L,Vogt G, Blanc M,Aragno M (1998) Bacterial diversity in the bulk soil and rhizosphere<br />

fractions of Lolium perenne and Trifolium repens as revealed by PCR<br />

restriction analysis of 16S rDNA. Plant Soil 198:219–224<br />

Markstädter C, Federle W, Jetter R, Riederer M, Hölldobler B (2000) Chemical composition<br />

of the slippery epicuticular wax blooms on Macaranga (Euphorbiaceae) ant<strong>plant</strong>s.<br />

Chemoecology 10:33–40<br />

Mendgen K (1996) Fungal attachment and penetration. In: Kerstiens G (ed) Plant cuticles:<br />

an integrated functional approach. BIOS Scientific Publishers, Oxford, pp<br />

175–188<br />

Morris CE, Monier JM, Jacques MA (1997) Methods for observing microbial biofilms<br />

directly on leaf <strong>surface</strong>s and recovering them for isolation of culturable microorganisms.<br />

Appl Environ Microbiol 63:1570–1576<br />

Morris CE, Monier JM, Jacques MA (1998) A technique to quantify the population size<br />

and composition of the biofilm component in communities of bacteria in the phyllosphere.<br />

Appl Environ Microbiol 64:4789–4795<br />

Morris CE, Nicot PC, Nguyen CN (1996) Aerial <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>. Plenum<br />

Press, New York<br />

Neinhuis C, Barthlott W (1998) Seasonal changes of leaf <strong>surface</strong> contamination in beech,<br />

oak, and gingko in relation to leaf micromorphology and wettability. New Phytol<br />

138:91–98


156<br />

Lukas Schreiber, Ursula Krimm and Daniel Knoll<br />

Nobel PS (1991) Physicochemical and environmental <strong>plant</strong> physiology. Academic Press,<br />

San Diego<br />

Ogram A (2000) Soil molecular microbial ecology at age 20: methodological challenges<br />

for the future. Soil Biol Biochem 32:1499–1504<br />

Persson A, Oesterberg E, Dostalek M (1988) Biosurfactant production by Pseudomonas<br />

fluorescens 378: growth and product characteristics. Appl Microbiol Biotech 29:1–4<br />

Preece TF, Dickinson CH (1971) Ecology of leaf <strong>surface</strong> microorganisms. Academic<br />

Press, London<br />

Reynhardt EC Riederer M (1994) Structures and molecular dynamics of <strong>plant</strong> waxes. II<br />

Cuticular waxes from leaves of Fagus sylvatica L. and Hordeum vulgare L. Eur Biophys<br />

J 23:59–70<br />

Riederer M, Markstädter C (1996) Cuticular waxes: a critical assessment of current<br />

knowledge. In: Kerstiens G (ed) Plant cuticles: an integrated functional approach.<br />

BIOS Scientific Publishers, Oxford, pp 189–200<br />

Riederer M, Schönherr J (1990) Effects of surfactants on water permeability of isolated<br />

<strong>plant</strong> cuticles and on the composition of their cuticular waxes. Pest Sci 29:85–94<br />

Riederer M, Schreiber L (1995) Waxes: the transport barriers of <strong>plant</strong> cuticles. Plant<br />

waxes. In: Hamilton RJ (ed) Waxes: chemistry, molecular biology and functions. The<br />

Oily Press, Dundee, pp 131–156<br />

Romantschuk M (1992) Attachment of <strong>plant</strong> pathogenic bacteria to <strong>plant</strong> <strong>surface</strong>s. Annu<br />

Rev Phytopathol 30:225–243<br />

Schäfer W (1998) The involvement of fungal cutinase in early processes of <strong>plant</strong> infection.<br />

Mol Genet Host-Specific Toxins Plant Dis 13:273–280<br />

Schönherr J, Baur P (1996) Cuticle permeability studies: a model for estimating leaching<br />

of <strong>plant</strong> metabolites to leaf <strong>surface</strong>s. In: Morris CE, Nicot PC, Nguyen CN (eds) Aerial<br />

<strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>. Plenum Press, New York, pp 1–23<br />

Schönherr J, Riederer M (1989) Foliar penetration and accumulation of organic chemicals<br />

in <strong>plant</strong> cuticles. Rev Environ Cont Toxicol 108:1–70<br />

Schreiber L (1996) Wetting of the upper needle <strong>surface</strong> of Abies grandis: influence of pH,<br />

wax chemistry and epiphyllic microflora on contact angles. Plant Cell Environ<br />

19:455–463<br />

Schreiber L, Schönherr J (1993) Determination of foliar uptake of chemicals: influence of<br />

leaf <strong>surface</strong> microflora. Plant Cell Environ 16:743–748<br />

Schreiber L, Schorn K, Heimburg T (1997) 2 H NMR study of cuticular wax isolated from<br />

Hordeum vulgare L. leaves: identification of amorphous and crystalline wax phases.<br />

Eur Biophys J 26:371–380<br />

Stadler B, Müller T (1996) Aphid honeydew and its effect on the phyllosphere microflora<br />

of Picea abies (L.) Karst. Oecologia 108:771–776<br />

Tiedje JM, Asuming Brempong S, Nusslein K, Marsh TL, Flynn SJ (1999) Opening the<br />

black box of soil microbial diversity. Appl Soil Ecol 13:109–122<br />

Tukey HB (1970) The leaching of substances from <strong>plant</strong>s. Annu Rev Plant Phys 21:<br />

305–324<br />

Turunen M, Huttunen S (1989) A review of the response of epicuticular wax of conifer<br />

needles to air pollution. J Environ Qual 19:35–45<br />

van Gardingen PR, Grace J, Jeffree CE (1991) Abrasive damage by wind to the needle <strong>surface</strong>s<br />

of Picea sitchensis (Bong) Carr and Pinus sylvestris L. Plant Cell Environ<br />

14:185–193<br />

Walton TJ (1990) Waxes, cutin and suberin. Meth Plant Biochem 4:105–158<br />

Yang CH, Crowley DE, Borneman J, Keen NT (2001) Microbial phyllosphere populations<br />

are more complex than previously realized. Proc Natl Acad Sci USA 98:3889–3894


10 Developmental Interactions Between<br />

Clavicipitaleans and Their Host Plants<br />

James F. White Jr., Faith Belanger, Raymond Sullivan,<br />

Elizabeth Lewis, Melinda Moy, William Meyer<br />

and Charles W. Bacon<br />

1 Introduction<br />

Clavicipitalean fungi have evolved to survive as saprophytes, degrading<br />

organic material, as well as biotrophs of <strong>plant</strong>s, fungi, nematodes, and insects.<br />

They have become particularly successful as epibionts and endophytes of<br />

grasses. We believe that the associations between clavicipitalean fungi and<br />

their hosts constitute unique biotrophic symbioses where the stages of physiological<br />

adaptation to the <strong>plant</strong> host may be examined to gain an understanding<br />

of how evolution among these fungi has progressed.<br />

2 Endophyte/Epibiont Niche<br />

In recent years, awareness has developed that many microbes colonize and<br />

inhabit interior and exterior <strong>surface</strong>s of <strong>plant</strong>s. Many microbes may colonize<br />

<strong>plant</strong>s without eliciting defense responses from host <strong>plant</strong>s or causing disease<br />

symptoms (Bacon and White 2000). The benefits to <strong>plant</strong>s of hosting beneficial<br />

microbes are numerous. Diazotrophic bacterial endophytes in sugarcane<br />

have been shown to fix atmospheric nitrogen that enables hosts to grow indefinitely<br />

in soils low in available nitrogen. Bacillus subtilis-infected seedlings of<br />

many <strong>plant</strong>s have been shown to have an enhanced growth rate and survival<br />

in pathogen-laden soils. Tall fescue seedlings infected by the endophyte Neotyphodium<br />

coenophialum show enhanced resistance to “damping off” disease<br />

caused by Rhizoctonia solani (Gwinn and Gavin 1992). Mature <strong>plant</strong>s of F.<br />

arundinacea show increased drought tolerance and resistance to above<br />

ground and below ground insect and nematode pests (Gwinn et al. 1991).<br />

Similarly, several grasses infected by the endophytes Epichloë typhina, E. festucae,<br />

and E. clarkii were found to deter the feeding of migratory locusts;<br />

while endophyte-free <strong>plant</strong>s were readily consumed by the locusts (Lewis et<br />

al. 1993). Arizona fescue (Festuca arizonica) infected by a Neotyphodium<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


158<br />

James F. White Jr. et al.<br />

endophyte has not been found to possess insect deterrent properties, instead<br />

growth enhancements have been proposed (Faeth et al. 2000). Finally several<br />

species of fine fescues infected by Epichloë festucae were found to have an<br />

increased resistance to the dollar spot disease caused by Sclerotinia homeocarpa.<br />

It seems evident that <strong>plant</strong>s benefit tremendously from the colonization of<br />

symbiotic microbes. The benefits to hosting mutualistic microbes likely outweigh<br />

losses in terms of nutrient use by the microbes. The widespread level of<br />

infection of grasses by Epichloë and asexual forms in Neotyphodium are evidence<br />

that these associations also have evolutionary value. In one study of<br />

endophytes in grasses, infection levels in some hosts (e.g., Achnatherum<br />

robustum and Festuca versuta) were estimated to be greater than 90 % in populations<br />

throughout the ranges of the grasses (White 1987).<br />

3 Coevolution of Clavicipitalean Fungi with Grass Hosts<br />

It is evident that Epichloë (Clavicipitaceae; Ascomycetes) and related asexual<br />

endophytes have co-evolved with the cool-season (C-3) grasses in which they<br />

perennate (White 1988). Species of these endophytes are unknown from<br />

warm-season (C-4) grasses (White 1987). On the other hand, endophytic<br />

species in genus Balansia, also in family Clavicipitaceae, appear to have coevolved<br />

with warm-season grasses and are rarely or never found on cool-season<br />

hosts (White and Owens 1992). In co-evolving with grasses, it is logical to<br />

expect that their interactions with hosts became more sophisticated.<br />

4 The Jump from Insects to Plants<br />

4.1 Trans-Kingdom Jump<br />

Analysis of rDNA 26S sequence data indicates that the predominantly insectinfecting<br />

subfamily Cordycipitoideae (Clavicipitaceae) is the most deeply<br />

rooted group and is, therefore likely ancestral to grass-infecting species (Sullivan<br />

et al. 2000).A trans-kingdom host jump is postulated to have occurred to<br />

<strong>plant</strong>s. Such a jump could have occurred gradually through intermediate<br />

forms that were parasitic on both insects and <strong>plant</strong>s.<br />

4.2 Intermediate Stages in the Transition to Plants<br />

Several Cordycipitoideae exhibit stages of such a transition. Most of the<br />

Cordycipitoideae (e.g., Cordyceps militaris and C. sinensis) infect insect hosts<br />

and mummify them, using their necrotrophied bodies as energy to fuel fun-


10 Development Interactions Between Clavicipitaleans and Their Host Plants 159<br />

gal development (Tzean et al. 1997). In these associations, there is no association<br />

with <strong>plant</strong>s and no opportunity for the fungi to adapt to <strong>plant</strong>s as hosts.<br />

However, some species of Cordycipitoideae infect scale insects that are sedentary<br />

and parasitic on <strong>plant</strong> hosts by use of stylets with which they penetrate<br />

and suck sugars from host vascular tissues. In these species some simple<br />

adaptations for parasitism of <strong>plant</strong>s are evident. In Hypocrella africana, H.<br />

gaertneriana,andH. schizostachyi, infection of the scale insect is biotrophic<br />

with the fungus obtaining nutrients from the <strong>plant</strong> through the living body of<br />

the insect (Hywel-Jones and Samuels 1998). Here, the parasitized scale insect<br />

is a bridge to obtain <strong>plant</strong> nutrients; however, the fungus does not interface<br />

directly with the <strong>plant</strong> in any way. This is an indirect adaptation to parasitism<br />

on <strong>plant</strong>s. The quantity of nutrients available to Hypocrella in this type of<br />

association far exceeds that available in the body of the insect. Hywel-Jones<br />

and Samuels (1998) estimated that the stroma attained some 1000 times or<br />

more the mass of the body of the insect. Hyperdermium bertonii exhibits<br />

another step toward direct parasitism of <strong>plant</strong>s. This species infects scale<br />

insects, necrophytizes them, then develops epibiotically on the <strong>surface</strong> of the<br />

<strong>plant</strong>, nourishing itself on sugars that continue to flow from the stylet wound<br />

left by the scale insect (Sullivan et al. 2000). Although H. bertonii relies on<br />

scale insects to prepare its parasitism site on <strong>plant</strong>s, it directly absorbs and<br />

utilizes <strong>plant</strong> sugars. It is also possible that H. bertonii produces compounds<br />

that interfere with scar tissue development to prevent the stylet wound from<br />

sealing. This possibility should be further evaluated. However, at present we<br />

have no evidence that wound retardant compounds or growth regulator compounds<br />

are being produced by H. bertonii. Regardless, it is evident that H.<br />

bertonii has taken physiological steps in adapting to growth on <strong>plant</strong> sugars.<br />

In experiments, conducted in vitro where H. bertonii is grown on a minimal<br />

medium containing minerals and combinations of simple sugars glucose and<br />

fructose, we have demonstrated that mycelium and conidial production are<br />

stimulated by equal ratios of glucose to fructose; while higher levels of fructose<br />

in media induce the fungus to differentiate pigmentation and its mature<br />

stromal morphology. Hyperdermium bertonii has adapted to utilize changes<br />

in host sugar content on which it nourishes itself to guide its development.<br />

Sucrose, leaking directly from the stylet wound, is cleaved to its component<br />

monomers glucose and fructose. The glucose is likely preferentially absorbed.<br />

As a result, fructose is left behind to accumulate in the liquid film of sugars on<br />

which the fungus grows. Increasing concentrations of fructose, or the fructan<br />

polymers of it, are the probable cues employed by the epiphyte to shift its<br />

growth from early stroma development to differentiation and maturation.<br />

The possession of invertases by H. bertonii may also be evidence of adaptation<br />

to <strong>plant</strong>s. Sucrose is only available in <strong>plant</strong> tissues. It is a short step from<br />

the condition of Hyperdermium to infection of <strong>plant</strong>s without the use of<br />

insects.


160<br />

James F. White Jr. et al.<br />

4.3 Parasitism of Grass Meristematic Tissues<br />

On the meristematic tissues of grasses, wounds created by insects are unnecessary<br />

since the tissues of meristems, such as the inflorescence primordia, are<br />

bathed in sucrose. Atkinsonella hypoxylon illustrates this point. Atkinsonella<br />

hypoxylon grows superficially on young leaves of grasses as an epiphyte, perhaps<br />

degrading wax in the cuticle to obtain nutrients for epiphytic growth<br />

(White et al. 1991). When the grass begins to produce an inflorescence primordium,<br />

sucrose is mobilized into the primordium to provide energy for its<br />

development. The primordium is surrounded by nutrients in liquid, which in<br />

turn is surrounded by layers of developing leaves. It is believed that the sudden<br />

increase in sucrose availability and concentration triggers rapid mycelial<br />

growth that eventually results in formation of the stroma (White and Chambless<br />

1991). It is interesting that leaves and inflorescence primordia within the<br />

stroma never develop a cuticular layer that would impede flow of nutrients<br />

and moisture to the fungus. Prevention of cuticular development and prevention<br />

of maturation of the inflorescence primordial tissues may be a growth<br />

regulator effect, although they have not yet been identified for this species.<br />

5 Developmental Differentiation of Endophytic and<br />

Epiphyllous Mycelium<br />

5.1 Plant Cell Wall Alteration<br />

Epichloë sp. illustrate many of the physiological capacities needed by<br />

Clavicipitaceae to colonize grasses (White et al. 1991). Epichloë inhabits leaf<br />

sheaths and growing tillers of grass <strong>plant</strong>s. Endophytic mycelium is largely<br />

nonbranched and exclusively intercellular (Fig. 1) and often seen to adhere<br />

closely to parenchyma cell walls as if attached by glue. This may be due to the<br />

partial degradation of cell wall components by the endophytic mycelium.<br />

While it is possible that cell walls are modified by endophytes, they remain<br />

largely intact as evidenced by electron microscopic studies. It is notable that<br />

during stroma development, profound changes have been observed in cell<br />

walls of the grass epidermis.Walls of epidermal cells appear to lose structural<br />

integrity with mycelium of the endophyte frequently penetrating the wall<br />

(Fig. 2).<br />

5.2 Endophytic Mycelial Growth<br />

Endophytic mycelium in young leaves or elongating tillers is frequently narrow<br />

(1 mm across), straight, oriented parallel to the axis of expansion of the<br />

cells and <strong>plant</strong> organ (Fig. 3). Sometimes in very young tissues, the hyphae


10 Development Interactions Between Clavicipitaleans and Their Host Plants 161<br />

Fig. 1. Electron micrograph (TEM) showing hyphae (arrows) of Epichloë amarillans in<br />

intercellular spaces of vascular tissues of the grass Agrostis hiemalis (¥10,000)


162<br />

James F. White Jr. et al.<br />

Fig. 2. Electron micrograph (TEM) showing hyphae (arrows) of Epichloë amarillans<br />

penetrating epidermal tissues of the grass Agrostis hiemalis (¥10,000)<br />

may be observed to taper to a fine point on the ends where it has been<br />

stretched and sheered during elongation of the <strong>plant</strong> tissues. Sheered hyphae<br />

are seen to recover rapidly with elongation of the sheared ends of the endophytic<br />

hyphae. In later stages of growth, the endophytic hyphae fully elongate,<br />

then become convoluted, apparently due to excessive elongation. This has the<br />

effect of increasing the <strong>surface</strong> area of the cell wall that an individual hypha<br />

may come into contact with. Whether an increased contact <strong>surface</strong> area<br />

results in increased nutrients leaking from the parenchyma cells to the endophytic<br />

mycelium is yet to be determined. Such endophytic mycelium is abundant<br />

in leaf sheaths where many nutrients are stored, but they are rare in the<br />

leaf blades where photosynthesis is occurring. It is the abundant presence of<br />

photosynthate within the cells of the leaf sheath that likely accounts for the<br />

abundance of mycelium in this tissue.


10 Development Interactions Between Clavicipitaleans and Their Host Plants 163<br />

Fig. 3. Section of developing<br />

Agrostis hiemalis culm<br />

showing endophytic hypha<br />

(arrow) oriented parallel<br />

to the direction of culm<br />

elongation (¥1000)<br />

5.3 Control of Endophytic Mycelial Development<br />

Endophytic mycelium is never observed to produce conidia within tissues of<br />

the <strong>plant</strong>. One experiment suggests that conidial development and branching<br />

are suppressed by unknown factors present within tissues of the leaf sheath<br />

parenchyma. Media containing basal salts (Murashige and Skoogs), agar<br />

(1 %), ground leaf sheath tissues of Agrostis hiemalis (0.5 % dry wt.), and low<br />

concentrations of glucose (0.5 %) produced mycelium of E. amarillans that<br />

sparsely branched and rarely produced conidiogenous cells, while controls<br />

that lacked only the ground leaf sheath tissues, branched and produced conidia<br />

abundantly.<br />

5.4 Epiphyllous Mycelial Development<br />

Some species of Epichloë and their asexual derivatives have been found to<br />

produce an epiphyllous stage where they grow superficially on the <strong>surface</strong> of


164<br />

James F. White Jr. et al.<br />

leaf blades. Epiphyllous mycelium tends to be present in the groves at cell<br />

junctions of the epidermis, may adhere closely to the cuticular <strong>surface</strong>, is frequently<br />

branched, and produces abundant wind-disseminated conidia (White<br />

et al. 1996; Moy et al. 2000). The epiphyllous network of mycelium and conidia<br />

are frequently connected to internal sources of nutrients by intercellular<br />

bridges, but are shielded from direct association with the interior leaf substances<br />

by the waxy cuticle layer on which it spreads. There is no evidence that<br />

species of Epichloë have the capacity to degrade cuticular waxes. Previous<br />

studies of the capacity of various Clavicipitaceae to colonize and degrade<br />

paraffin showed that Epichloë does not colonize paraffin beads in agar culture<br />

and apparently cannot degrade waxes to gain nutrients, although other<br />

Clavicipitaceae, such as the predominantly epiphytic Atkinsonella hypoxylon,<br />

do posses that capacity (White et al. 1991).<br />

5.5 Expression of Fungal Secreted Hydrolytic Enzymes<br />

in Infected Plants<br />

All fungi, whether saprophytic, pathogenic, or mutualistic, acquire their carbon<br />

and nitrogen by absorption of small molecules from their surroundings.<br />

Fungi typically secrete numerous enzymes that function in degradation of<br />

polymeric substances in the environment to their monomeric constituents<br />

that can then be absorbed by the fungal cells.<br />

Endophytic fungi are exclusively intercellular and do not invade the <strong>plant</strong><br />

cells. They must, therefore obtain all their carbon and nitrogen compounds<br />

from the nutrient-poor apoplastic space. Endophytic fungal-secreted proteins<br />

are likely to be important components of the mutualistic interaction as they<br />

are located at the interface of the two species. Fungal secreted proteins are<br />

expected to be synthesized for growth and nutrient acquisition and perhaps<br />

for defense.<br />

We have detected expression of several fungal-secreted enzymes in Poa<br />

ampla infected with a Neotyphodium sp. endophyte. A fungal subtilisin-like<br />

proteinase was purified from infected leaf sheaths and cDNA and genomic<br />

clones for the gene were characterized (Lindstrom and Belanger 1994; Reddy<br />

et al. 1996). The fungal proteinase was found to be expressed at surprisingly<br />

high levels in the infected <strong>plant</strong> tissues. It was estimated to be 1–2 % of the<br />

total leaf sheath protein, suggesting it was a major fungal protein. The amino<br />

acid sequence of the proteinase is homologous to proteinases believed to be<br />

important in pathogenicity of entomopathogenic, nematophagous, and mycoparasitic<br />

fungi (Geremia et al. 1993; Bonants et al. 1995; St. Leger 1995).<br />

A fungal secreted endochitinase and an endo-b-1,6-glucanase are also<br />

expressed in the infected P. ampla <strong>plant</strong>s. Sequencing of cDNA clones for the<br />

chitinase and glucanase revealed they are 38 and 74 % identical, respectively,<br />

to the homologous enzymes from Trichoderma harzianum. T. harzianum is a


10 Development Interactions Between Clavicipitaleans and Their Host Plants 165<br />

potent mycoparasite of many <strong>plant</strong> pathogenic fungi (Papavizas 1985; Chet<br />

1987). Because of this property, it is being investigated as a potential biocontrol<br />

agent in crop production.<br />

The physiological roles of the endophytic proteinase, chitinase, and endob-1,6-glucanase<br />

are not yet known. The endophytic chitinase transcript is<br />

very abundant as determined from a blot of total RNA isolated from the<br />

infected <strong>plant</strong>s. This is similar to the situation with the endophytic proteinase<br />

(Reddy et al. 1996). The endo-b-1,6-glucanase appears to be expressed at<br />

lower levels. Several roles have been proposed for chitinases and endoglucanases<br />

from filamentous fungi. Roles in hyphal growth, branching, and<br />

autolysis have been proposed (Bartnicki-Garcia 1973; Gooday and Gow 1990;<br />

Peberdy 1990) as well as roles in mycoparasitism. Functions in fungal growth<br />

and/or in mycoparasitism would be relevant to endophytic infection. Interestingly,<br />

the homologous proteinase, chitinase, and endo-b-1,6-glucanase from<br />

T. harzianum are believed to be synergistic components of its mycoparasitic<br />

activity (Geremia et al. 1993; Garcia et al. 1994; Lora et al. 1995). These<br />

hydrolytic enzymes function together to break down the cell walls of the fungal<br />

hosts allowing entry of the T. harzianum hyphae.<br />

Expression of these hydrolytic enzymes in endophyte-infected <strong>plant</strong>s<br />

raises the possibility that they may also function as a mycolytic system for the<br />

endophyte. Such a system could provide the endophyte with a source of nutrients<br />

in addition to <strong>plant</strong> derived nutrients found in the apoplast. With a<br />

mycolytic system, the endopytic hyphae located on the <strong>surface</strong> of the <strong>plant</strong><br />

(Moy et al. 2000) would have access to additional sources of nutrients from<br />

other <strong>surface</strong>-located fungi. By attacking invading fungi, an endophytic<br />

mycolytic system could also protect the <strong>plant</strong>s from pathogenic fungi, perhaps<br />

resulting in enhanced disease resistance. Current research is aimed at<br />

determining the roles of these enzymes in endophyte infection.<br />

6 Modifications of Plant Tissues for Nutrient Acquisition<br />

6.1 Development of the Stroma in Epichloë<br />

The development of sexual reproductive structures in <strong>plant</strong>s poses some special<br />

problems for Clavicipitaceae. Larger quantities of nutrients are needed to<br />

provide the fuel for construction of the external mycelial stroma on which are<br />

produced first spermatia, then perithecia and ascospores (White and Bultman<br />

1987; Bultman et al. 1995). To obtain large quantities of nutrients from hosts,<br />

many other groups of biotrophic fungi,e.g.,powdery mildews,downy mildews,<br />

and rusts may produce haustoria to suck nutrients from individual host cells<br />

(Alexopoulos et al. 1996). However, clavicipitalean <strong>plant</strong> biotrophs have<br />

another strategy.They grow on meristematic tissues before the cuticle has been<br />

formed and by some unknown mechanism prevent development of the waxy


166<br />

James F. White Jr. et al.<br />

cuticle and alter the epidermis itself, effectively removing a key barrier to the<br />

flow of nutrients to the stromal mycelium. The following two examples will<br />

illustrate this method of nutrient acquisition by these clavicipitaleans. In<br />

Epichloë the abundance of sucrose in the developing inflorescence primordium<br />

triggers the fungus to proliferate rapidly and permeate the young<br />

inflorescence and the leaf sheath of a leaf that surrounds it.This process is comparable<br />

to that already suggested for stroma development in Atkinsonella<br />

hypoxylon, except that in Epichloë the mycelium is endophytic and frequently<br />

permeates vascular tissues as well as nonvascular tissues (White et al. 1991).<br />

The stroma is composed of a mix of <strong>plant</strong> tissues and fungal mycelium. These<br />

stromata are much like those of the scale insect parasites Hypocrella africana,<br />

H. gaertneriana,andH. schizostachyi,in that the host tissues embedded within<br />

the stromata remain alive, but are modified so that nutrients will flow freely<br />

into the developing stromata. Plant tissues embedded within the stroma are<br />

not only permeated by mycelium, but also possess epidermal cells that are<br />

hypertrophied, often collapsed, and lack waxy cuticles (White et al. 1997).<br />

Through these modifications of the host tissues, the endophyte removes all<br />

barriers to nutrient flow into the stromal mycelium. The development of<br />

mycelium within the vascular bundle enhances the transfer of nutrients to the<br />

fungal stroma. By mummifying the living inflorescence primordium and the<br />

sheath of the leaf that surrounds it, the fungus can intercept all nutrients that<br />

are transported into the flowering tiller. Mature stromata of Epichloë always<br />

possess the stromal leaf blade emergent from the top of the stroma (Fig.4).The<br />

reason for this emergent leaf blade is unknown, but may be a source of <strong>plant</strong><br />

hormones that are needed as a signal to the <strong>plant</strong> to continue to send nutrients<br />

into the culm. Experimental work is needed to evaluate this hypothesis.<br />

6.2 Stroma Development in Myriogenospora<br />

A second clavicipitalean biotroph that modifies host tissues for nutrient<br />

acquisition during stroma development is the epiphytic fungus Myriogenospora<br />

atramentosa. Myriogenospora atramentosa grows superficially on<br />

the epidermis of young leaves at the crown of many warm-season grasses and<br />

sedges. As the leaves develop, conidia of M. atramentosa proliferate on the<br />

folded leaves of the grass. The leaves continue to expand and the conidial<br />

stroma develops into a linear black perithecial stroma, composed of a single<br />

line of perithecia (Figs. 5, 6). The <strong>plant</strong> leaf tissues beneath the stroma are<br />

modified with hypertrophied epidermal cells that lack a cuticular layer<br />

(Rykard et al. 1985; White and Glenn 1994). The absence of a cuticle layer on<br />

the leaf epidermis and modification of the epidermal cells by the fungus permits<br />

M. atramentosa to absorb nutrients directly through the epidermis of the<br />

leaf blades to provide energy for stroma development.


10 Development Interactions Between Clavicipitaleans and Their Host Plants 167<br />

Fig. 4. Stroma (arrow) of Epichloë<br />

amarillans showing white stromal<br />

mycelium and apical stromal leaf<br />

(¥3)<br />

Fig. 5. Black, linear, stroma (arrow) of Myriogenospora<br />

atramentosa on upper <strong>surface</strong> of<br />

leaf of Andropogon sp. (¥2)


168<br />

James F. White Jr. et al.<br />

6.3 Mechanisms for Modifying Plant Tissues<br />

Fig. 6. Cross-section of<br />

stroma of Myriogenospora<br />

atramentosa showing a single<br />

perithecium (arrow) bordered<br />

by the leaf blades on<br />

either side (¥500)<br />

The mechanisms whereby the Clavicipitaceae alter development of <strong>plant</strong> tissues<br />

is unknown. One hypothesis is that at least some of their secondary<br />

products may have growth regulator effects. In this respect, it is notable that<br />

several Clavicipitaceae, including Epichloë festucae and Balansia epichloë<br />

have been shown to produce the <strong>plant</strong> auxin indole acetic acid (IAA; Porter et<br />

al. 1985; Yue et al. 2000). Indeed, other indole derivatives such as the ergot<br />

alkaloids may also possess auxin-like effects. One effect that auxin has is to<br />

loosen cell wall fibers, allowing cells to expand.<br />

Moubarak et al. (1993) demonstrated that ergovaline, an ergot alkaloid<br />

commonly produced by Epichloë/Neotyphodium endophytes, interferes with<br />

cell membrane polarization and ATPase activities in animal tissues. These<br />

data suggest a potential mechanism by which ergot alkaloids may alter physiology<br />

and structure of <strong>plant</strong> tissues and acquire nutrients from those tissues.<br />

If ergot alkaloids, such as ergovaline, inhibit ATPases in grass cells, they may<br />

enhance leakage of nutrients from cells adjacent to mycelium. Without use of<br />

ATPases, <strong>plant</strong> cells would be incapable of utilizing active transport proteins<br />

to reacquire leaking nutrients.Almost no research has been pursued to evalu-


10 Development Interactions Between Clavicipitaleans and Their Host Plants 169<br />

ate impacts of clavicipitalean-produced secondary metabolites on <strong>plant</strong> tissues<br />

themselves. The current scientific wisdom holds that clavicipitalean secondary<br />

metabolites have impacts on animal tissues as feeding deterrents and<br />

other defensive compounds. Whether ergot alkaloids, auxin-like compounds<br />

or other secondary metabolites of these fungi are involved in effecting<br />

changes in <strong>plant</strong> tissues embedded within or adjacent to stromal mycelium<br />

must be further evaluated. It seems likely that this will be a fruitful area for<br />

future investigation.<br />

6.4 Evaporative-Flow Mechanism for Nutrient Acquisition<br />

The stroma of Epichloë maintains a constant flow of water and nutrients into its<br />

mycelium through an evaporation-driven process (White and Camp 1996;<br />

White et al. 1997). Water evaporates rapidly from the <strong>surface</strong> of stromata. As<br />

water evaporates from the stroma it is replaced by water from the <strong>plant</strong>. This<br />

process establishes a flow of water and dissolved nutrients into the stroma from<br />

mycelium interfacing with the vascular bundles and other tissues embedded<br />

within the stroma. Evaporative flow mimics the enhanced transpiration that<br />

occurs in developing inflorescences during elongation of the flowering tillers<br />

of uninfected grasses, but here the stroma is the recipient of the nutrients.<br />

6.5 The Cytokinin Induction Hypothesis<br />

In Atkinsonella hypoxylon stromata form on the inflorescence primordium<br />

and include parts of several leaves as well (Fig. 7). The stromata are gray<br />

(sometimes with areas of a yellow pigment) and produce several different<br />

spore states, including cup-shaped sporodochia that produce moist masses of<br />

ephelidial conidia, and a layer of neotyphodial conidia borne on tips of elongate<br />

conidiogenous cells. It is reasonable to expect that the fungus would<br />

coordinate its development with that of its host grass. For example, the fungus<br />

mycelium must be able to detect when it is growing on an inflorescence primordium<br />

rather than on the tiller meristems. On the tiller meristems it will<br />

produce a low biomass of nonpigmented mycelium and ephelidial conidia,<br />

but no neotyphodial conidia or other structures; while on the inflorescence<br />

primordium the entire suite of morphological structures is produced. One<br />

way for the fungus to coordinate its development to that of the host <strong>plant</strong><br />

would be to use compounds present in the host during different stages of<br />

development as ‘cues’ to initiate developmental stages in the fungus. Our<br />

approach to the search for host compounds that may serve as cues for fungal<br />

development has been a trial and error approach. Over several years, we have<br />

screened hundreds of compounds that might be present in grass tissues to<br />

determine how they affect differentiation of A. hypoxylon.


170<br />

James F. White Jr. et al.<br />

Fig. 7. Two stromata<br />

(arrows) of Atkinsonella<br />

hypoxylon on culms of<br />

Danthonia spicata (¥4)<br />

Studies on Atkinsonella hypoxylon and A. texensis in vitro have demonstrated<br />

that certain media additives will induce the fungus to develop in a way<br />

comparable to that seen on the host grass inflorescence primordia (Bacon and<br />

White 1994). When these claviciptaleans are grown on media containing agar<br />

(1 %), basal salts (Murashige and Skoog; Sigma Chemical Company, Inc.), and<br />

glucose (3 %), colonies are white, with no aerial mycelium or conidia of any<br />

type. This is an undifferentiated mycelium, the fungus equivalent of ‘callus tissue’.<br />

Stroma-like colonies with gray pigmentation, sporodochia producing<br />

ephelidial conidia, and a layer of neotyphodial conidiogenous cells and conidia<br />

can be induced by inclusion of 100 ppm of the cytokinin zeatin or kinetin<br />

(Research Organics, Inc. Cleveland, Ohio) in the medium. The stroma-like<br />

states in culture are most striking when the grass cytokinin zeatin is<br />

employed. Partial induction of stroma-like states may be induced through use<br />

of 1 % citrate (sodium or potassium salt) and 0.1–0.5 % acetate (sodium or<br />

potassium salt). With acetate in the medium the gray pigmentation is seen to<br />

develop, but differentiated reproductive cells do not form. With citrate in the<br />

medium, pigmentation, sporodochia and ephelidial conidia form, but the<br />

neotyphodial conidia do not form. Because induction of differentiation is<br />

incomplete with the use of acetate and citrate, we believe that these compounds<br />

are not the primary cues for stroma differentiation, but instead may<br />

be indirectly causing differentiation by turning on secondary metabolism<br />

pathways. On the other hand, cytokinins are <strong>plant</strong> hormones and are expected<br />

to be present in the developing ovary tissues embedded within the fungal<br />

stroma since ovaries produce cytokinins for regulation of their own development<br />

(Miller 1961; Mauseth 2003). Thus the presence of cytokinins may be a


10 Development Interactions Between Clavicipitaleans and Their Host Plants 171<br />

key signal for the fungus to begin a sequence of developmental events that<br />

end in production of the mature stroma. Some preliminary differential display<br />

studies were also conducted to examine genes that may be upregulated<br />

and downregulated when A. hypoxylon was exposed to cytokinin. The results<br />

of these differential display studies showed that several genes were turned on<br />

while several others were turned off, however, none of the genes were identified.<br />

One preliminary study on another clavicipitalean-producing stromata<br />

on inflorescence primordia, Epichloë festucae, employed a monoclonal antibody-based<br />

cytokinin detection kit (Phytodetek-t-ZR, Sigma, St. Louis, Missouri)<br />

to compare levels of cytokinin in stromata and other tissues of the<br />

grass. The result of this test suggested that cytokinin was present in high concentrations<br />

within the stromata. However, this test must be considered preliminary<br />

because of the possibility for cross-reactivity of the antibody with<br />

other compounds. More precise tests for the presence of cytokinins must be<br />

employed to evaluate levels in the stromata. Presently, the hypothesis that<br />

cytokinins are a key cue for development of stromata on the grass inflorescence<br />

primordium for the grass inflorescence-colonizing clavicipitaleans is<br />

an interesting hypothesis. However, additional work must be done to evaluate<br />

this hypothesis.<br />

7 Evolution of Asexual Derivatives of Epichloë<br />

7.1 Reproduction and Loss of Sexual Reproduction<br />

One notable feature of genus Epichloë is the abundance of asexual species,<br />

often classified in form genus Neotyphodium. Formation of asexual derivatives<br />

is apparently a relatively frequent phenomenon based on how common<br />

these asexual forms are in grasses (White 1987). In the sexual cycle of<br />

Epichloë stromata are produced on grasses, and on stromata spermatia<br />

develop. In a heterothallic mating process symbiotic flies in genus<br />

Botanophila (Anthomyidae) vector spermatia between stromata of the opposite<br />

mating type (Bultman et al. 1995). Following deposition of spermatia on<br />

a compatible stroma, an ascogenous (dikaryotic) mycelium develops in<br />

which perithecia and ascospores form. Meiosis takes place within the asci to<br />

result in the haploid ascospores that are ejected from asci onto surrounding<br />

vegetation, where they may germinate to form wind-disseminated conidia<br />

(White and Bultman 1987). Precisely how primary infections of grasses<br />

occur is still unknown, but may involve a period of epiphyllous growth prior<br />

to penetration of <strong>plant</strong> tissues (Moy et al. 2000). Other investigators (Diehl<br />

1950; Chung and Schardl 1997) have suggested that ovules may be the site of<br />

entry into <strong>plant</strong>s. However, definitive data that will answer this question are<br />

still lacking. The asexual forms of Epichloë are seed-transmitted and stromata<br />

do not form on grass inflorescences. Since these asexual forms do not


172<br />

James F. White Jr. et al.<br />

form stromata, sexual recombination does not occur. Seed transmission is<br />

the result of growth of the endophyte in inflorescence primordia. When<br />

ovules differentiate in the primordia, the fungus is incorporated into tissues<br />

of the nucellus. When the embryo differentiates within the nucellus, it is<br />

invaded by endophytic mycelium and the next generation of host has been<br />

effectively colonized (White and Cole 1986).<br />

7.2 The Hypotheses<br />

Two hypotheses have been proposed to explain loss of the sexual cycles by<br />

species of Epichloë. In the ‘hybridization hypothesis’ it is suggested that<br />

hybridization between two different species of Epichloë results in ‘hybrids’<br />

that cannot undergo sexual reproduction due to meiotic incompatibility of<br />

the two sets of chromosomes (Schardl and Wilkerson 2000). The frequent<br />

occurrence of multiple sets of genes in some asexual endophytes supports<br />

this hypothesis (Leuchtmann and Clay 1990; Tsai et al. 1994; Cabral et al.<br />

1999). The occurrence of asexual forms such as the endophyte of Lolium<br />

rigidifolium that do not show multiple copies of genes is problematic for the<br />

hybridization hypothesis (Moon et al. 2000). The second problem with the<br />

hybridization hypothesis is that it suggests a very unlikely scenario. It suggests<br />

that haploid spermatia of one species fuse with haploid mycelium on a<br />

stroma of an opposite species to produce a dikaryotic mycelium. The next<br />

steps would involve formation of perithecia, asci, and ascospores. Within the<br />

asci the two nuclei from different species of Epichloë would fuse to become a<br />

diploid which would be immediately followed by meiosis to result in production<br />

of the haploid ascospores. Without formation of ascospores, the hybrid<br />

would be unable to spread. If the two genomes were meiotically incompatible<br />

as the ‘hybridization hypothesis’ suggests that first meiosis would not occur<br />

and ascospores could not be produced. This hypothesis invokes meiotic<br />

incompatibility, yet demands that meiosis occurred at least once following<br />

hybridization. It seems unlikely that hybridization and meiotic incompatibility<br />

account for the origins of asexual Epichloë endophytes. It should be noted<br />

that speciation by hybridization does work in <strong>plant</strong>s. However, in <strong>plant</strong>s meiosis<br />

does not occur immediately after hybridization, instead, a diploid forms.<br />

The diploid may reproduce clonally for a time (Grant 1977).<br />

The plurality of gene copies present within many asexual endophytes may<br />

be an indication of a parasexual process that is acting in asexual endophytes<br />

to produce variation. To evaluate whether multiple gene copies reflect parasexual<br />

recombinations within populations of asexual endophytes, it is necessary<br />

to conduct populational studies on gene variation. To this point studies<br />

examining gene variation in asexual endophytes have involved only a few<br />

isolates. It will be important to determine whether this parasexual recombination<br />

(hybridization) is a populational phenomenon and occurring rela-


10 Development Interactions Between Clavicipitaleans and Their Host Plants 173<br />

tively frequently within populations of the fungi or is a rare event, resulting<br />

in the origins of new species. Until we understand how frequent the asexual<br />

recombinatorial events are, their importance and significance will be speculation.<br />

The alternative hypothesis to explain loss of stromata and the sexual cycle<br />

invokes ecological factors and is termed the ‘environmental selection hypothesis’.<br />

This hypothesis suggests that stroma development reduces fitness of the<br />

symbiotic unit (grass and endophyte) and is selected against under certain<br />

environmental conditions. It is supported by work demonstrating that stromata<br />

increase the losses of water from <strong>plant</strong> tissues (White and Camp 1996),<br />

and decrease the fecundity of hosts by replacing inflorescences with stromata<br />

(White and Chambless 1991). It has been observed that stromata tend to form<br />

on <strong>plant</strong>s in soils that contain high levels of moisture, whereas asexual forms<br />

occur in <strong>plant</strong>s that live in soils that range from very dry to moist. Additional<br />

ecological studies are needed to confirm the association between soil moisture<br />

and stromata occurrence.<br />

7.3 The Process of Stroma Development and its Loss<br />

To understand the mechanism of loss of stroma-forming ability in Epichloë,<br />

it is necessary to understand the mechanism of stroma development and the<br />

interactions between endophyte and grass during stroma development.<br />

Kirby (1961) proposed that the capacity to form stromata on grasses was a<br />

function of the growth rate of fungal mycelium in the inflorescence primordium<br />

versus the growth rate of the inflorescence primordium. That is,<br />

endophytes that grow rapidly in the inflorescence primordium tissues can<br />

outgrow the inflorescence primordium, surround it, and trap it in a stromal<br />

mycelium, thus successfully forming a stroma. If an endophyte cannot grow<br />

rapidly enough to trap the inflorescence primordium in a stromal mycelium,<br />

the inflorescence emerges, and develops flowers and seeds that may contain<br />

the endophyte.<br />

Central to the issue of stroma development is the question of which nutrients<br />

provide the energy for stroma formation. Lam et al. (1995) demonstrated<br />

that Epichloë festucae possesses the sucrose degrading enzyme invertase and<br />

suggested that this enzyme may play a role in stroma development. Earlier<br />

work by White et al. (1991) suggests another mechanism. White et al. (1991)<br />

examined the growth rate of a range of endophytes producing stromata of different<br />

sizes on several different sugars likely to be found in grass inflorescence<br />

primordia. These sugars included glucose, fructose, xylose, and arabinose.<br />

Glucose and fructose result from the cleavage of sucrose that is abundant in<br />

and around the inflorescence primordium tissues. Xylose and arabinose are<br />

sugars present in the cell wall polysaccharides of grasses and may be available<br />

in meristems of the primordium. In this study it was found that there is a pos-


174<br />

James F. White Jr. et al.<br />

itive correlation between the size of stromata and the growth rate on a selection<br />

of sugars. Apparently, the larger the stroma formed on a particular host,<br />

the faster an endophyte must grow to develop that stroma. It was further<br />

found that endophytes that failed to reach the critical growth rate on any of<br />

the sugars, tended to produce fewer stromata per <strong>plant</strong> than endophytes that<br />

grew rapidly on all of the sugars. From an evolutionary perspective, selection<br />

against stroma development may be selection for endophytes that grow more<br />

slowly on nutrients available in host tissues. This hypothesis is consistent with<br />

at least one important observation. Many asexual endophytes (e.g., Neotyphodium<br />

coenophialum and N. lolii) are slow growing in culture while stromaforming<br />

endophytes grow comparably faster.<br />

7.4 The Shift from Pathogen to Mutualist<br />

Much is known of the biochemistry and genetics of the interactions between<br />

<strong>plant</strong>s and pathogenic organisms and how these interactions result in disease<br />

or in <strong>plant</strong> resistance (Oliver and Osbourne 1995; Hammond-Kosack and<br />

Jones 1996). Mutualistic associations, such as those between the fungal endophytes<br />

and their grass hosts, are believed to have evolved from pathogenic<br />

associations (Clay 1988). Little is known regarding the genetic changes that<br />

result in a change from a pathogenic to a mutualistic lifestyle.<br />

Plant fungal pathogens typically secrete a number of <strong>plant</strong> cell wall degrading<br />

enzymes such as cellulases, glucanases, xylanases, and polygalacturonases.<br />

It is likely that expression of these cell wall degrading enzymes plays<br />

some role in pathogenicity (Oliver and Osbourne 1995; Mendgen et al. 1996),<br />

although disruption of individual genes has not resulted is reduced virulence<br />

(Scott-Craig et al. 1990; Apel et al. 1993; Schaeffer et al. 1994; Bowen et al. 1995;<br />

Sposato et al. 1995). The presence of other genes encoding the same enzyme<br />

activity and synergistic activity of different cell wall degrading enzymes in<br />

pathogenicity may explain these results.<br />

Claviceps purpurea, a <strong>plant</strong> pathogen closely related to the Epichloë and<br />

Neotyphodium endophytes, secretes a polygalacturonase during infection of<br />

rye ovaries (Tenberge et al. 1996). Polygalacturonase activity is believed to be<br />

important in splitting the host middle lamellae allowing intercellular growth<br />

of the fungus (Tenberge et al. 1996). Since the fungal endophytes also have an<br />

intercellular mode of growth, we have investigated the possibility of endophytic<br />

polygalacturonase expression in the Neotyphodium sp. endophyte that<br />

infects the grass Poa ampla. No hybridization was detected in a DNA blot<br />

using the cloned C. purpurea gene as a probe. Also, nothing was detected in<br />

PCR reactions using degenerate primers based on conserved amino acid<br />

regions of polygalacturonase genes from diverse organisms. It appears that<br />

this endophytic fungus may have lost the gene(s) for polygalacturonase. Perhaps<br />

loss of this cell wall degrading activity is a factor in the evolution of


10 Development Interactions Between Clavicipitaleans and Their Host Plants 175<br />

pathogen to mutualist. Since the fungal endophytes are exclusively intercellular<br />

and do not invade the <strong>plant</strong> cells, it is likely that genes for other cell wall<br />

degrading enzymes have also been lost. Ultimately, genome sequencing of an<br />

endophytic fungus will reveal the differences in enzyme coding capacity<br />

between fungal pathogens and fungal endophytes.<br />

8 Conclusions<br />

Our understanding of the range of physiological interactions between<br />

clavicipitalean mycosymbionts and grasses is virtually nonexistent. The<br />

majority of the research to date has focused on the agronomic aspects of the<br />

toxicity problem or on ecology of the hosts as modified by these fungi. As a<br />

consequence, physiology of clavicipitalean – <strong>plant</strong> interactions is a fertile and<br />

potentially important area of research.<br />

References and Selected Reading<br />

Alexopoulos CJ, Mims CW, Blackwell M (1996) Introductory mycology. Wiley, New York<br />

Apel PC, Panaccione DG, Holden FR, Walton JD (1993) Cloning and targeted gene disruption<br />

of XYL1, a 1,4-xylanase gene from the maize pathogen Cochliobolus carbonum.<br />

Mol Plant-Microbe Interact 6:457–473<br />

Bacon CW, White JF Jr (1994) Stains, media, and procedures for analyzing endophytes.<br />

In: Bacon CW, White JF Jr (eds) Biotechnology of endophytic fungi of grasses. CRC<br />

Press, Boca Raton, Florida, pp 47–58<br />

Bacon CW, White JF Jr (2000) Microbial endophytes. Marcel-Dekker, New York, pp 341–<br />

388<br />

Bartnicki-Garcia S (1973) Fundamental aspects of hyphal morphogenesis. In: Ashworth<br />

JM, Smith JE (eds) Microbial differentiation. Cambridge University Press, Cambridge<br />

Bonants PJM, Fitters PFL, Thijs H, den Belder E, Waalwijk C, Henfling JWDM (1995) A<br />

basic serine protease from Paecilomyces lilacinus with biological activity against<br />

Meloidogyne hapla eggs. Microbiology 141:775–784<br />

Bowen JK, Templeton MD, Sharrock KR, Crowhurst RN, Rikkerink EHA (1995) Gene<br />

inactivation in the <strong>plant</strong> pathogen Glomerella cingulata: three strategies for the disruption<br />

of the pectin lyase gene pnlA. Mol Gen Genet 246:196–205<br />

Bultman TL,White JF Jr, Bowdish TI,Welch AM, Johnston J (1995) Mutualistic transfer of<br />

Epichloë spermatia by Phorbia flies. Mycologia 87:182–189<br />

Cabral D, Cafaro M, Saidman B, Lugo M, Reddy PV, White JF Jr (1999) Evidence supporting<br />

the occurrence of a new species of endophyte in some South American grasses.<br />

Mycologia 91:315–325<br />

Chet I (1987) Trichoderma – application, mode of action, and potential as a biocontrol<br />

agent of soil borne <strong>plant</strong> pathogenic fungi. In: Chet I (ed) Innovative approaches to<br />

<strong>plant</strong> disease control. Wiley, New York<br />

Chung KR, Schardl CL (1997) Sexual cycle and horizontal transmission of the grass symbiont,<br />

Epichloë typhina. Mycological Res 101:295–301<br />

Clarke BB, White, JF Jr, Funk CR Jr, Sun S, Huff DR, Hurley RH (2003) Enhanced resistance<br />

to dollar spot in endophyte-infected fine fescues. Plant Dis (in press)


176<br />

James F. White Jr. et al.<br />

Clay K (1988) Clavicipitaceous fungal endophytes of grasses: coevolution and the<br />

change from parasitism to mutualism. In: Pirozynski KA, Hawksworth DL (eds)<br />

Coevolution of fungi with <strong>plant</strong>s and animals. Academic Press, London, pp 79–105<br />

Diehl (1950) Balansia and Balansiae in America, USDA Monograph, US Govt Printing<br />

Office, Washington, DC, 99 pp<br />

Faeth S, Sullivan H, Hamilton CE (2000) What maintains high levels of Neotyphodium<br />

endophytes in native grasses? A dissenting view and alternative hypotheses, In: Paul<br />

VH, Krohn K, Dapprich PD, Gutter B (eds) Proceedings Fourth International Neotyphodium/Grass<br />

Interactions Symposium. The University of Paderborn, Soest, p 14<br />

Garcia I, Lora JM, de la Cruz J, Benitez T, Llobell A, Pintor-Toro JA (1994) Cloning and<br />

characterization of a chitinase (CHIT42) cDNA from the mycoparasitic fungus Trichoderma<br />

harzianum. Curr Genet 27:83–89<br />

Geremia RA, Goldman GH, Jacobs D, Ardiles W, Vila SB, Van Montagu M, Herrera-<br />

Estrella (1993) A. Molecular characterization of the proteinase-encoding gene, prb1,<br />

related to mycoparasitism by Trichoderma harzianum. Mol Microbiol 8:603–613<br />

Gooday GW, Gow NAR (1990) Enzymology of tip growth in fungi. In: Heath IB (ed) Tip<br />

growth in <strong>plant</strong> and fungal cells. Academic Press, New York, pp 31–58<br />

Grant V (1977) Organismic Evolution. WH Freeman, San Francisco<br />

Gwinn KD, Gavin AM (1992) Relationship between endophyte infestation level of tall<br />

fescue seed lots and Rhizoctonia zeae seedling disease. Plant Dis 76:911–914<br />

Gwinn KD, Blank CA, Cole AM, Pless CD (1991) Resistance of endophyte-infected tall<br />

fescue seedlings to pathogens and pests. Tenn Farm Home Sci 160:72<br />

Hammond-Kosack KE, Jones JDG (1996) Resistance gene-dependent <strong>plant</strong> defense<br />

responses. Plant Cell 8:1773–1791<br />

Hywel-Jones NL, Samuels GJ (1998) Three species of Hypocrella with large stromata<br />

pathogenic on scale insects. Mycologia 90:36–46<br />

Kirby EJM (1961) Host-parasite relations in the choke disease of grasses. Trans Br Mycol<br />

Soc 44:493–503<br />

Lam CK, Belanger FC, White JF Jr, Daie J (1995) Invertase activity in Epichloë/Acremonium<br />

fungal endophytes and its possible role in choke disease. Mycol Res 99:867–873<br />

Lane GA, Christensen MJ, Miles CO (2000) Coevolution of fungal endophytes with<br />

grasses: the significance of secondary metabolites. In: Bacon CW, White JF Jr (eds)<br />

Microbial Endophytes. Marcel-Dekker, New York, pp 341–388<br />

Leuchtmann A, Clay K (1990)Isozyme variation in the Acremonium/Epichloë fungal<br />

endophyte complex. Phytopathology 80:1133–1139<br />

Lewis GC, White JF Jr, Bonnefont J (1993) Evaluation of grasses infected with fungal<br />

endophytes against locusts. Ann Appl Biol; Tests Agrochem Cultivars 14:142–143<br />

Lindstrom JT, Belanger FC (1994) A novel fungal protease expressed in endophytic infection<br />

of Poa species. Plant Physiol 102:645–650<br />

Lora JM, de la Cruz J, Llobell A, Benitez T, Pintor-Toro JA (1995) Molecular characterization<br />

and heterologous expression of an endo-b-1,6-glucanase gene from the mycoparasitic<br />

fungus Trichoderma harzianum. Mol Gen Genet 247:639–645<br />

Mauseth JD (2003) Botany: An introduction to <strong>plant</strong> biology. Jones and Bartlett, Boston<br />

Mendgen K, Hahn M, Deising H (1996) Morphogenesis and mechanisms of penetration<br />

by <strong>plant</strong> pathogenic fungi. Ann Rev Phytopathol 34:367–386<br />

Miller CO (1961) A kinetin-like compound in maize. Proc Natl Acad Sci USA 47:170–174<br />

Moon CD, Scott B, Schardl CL, Christensen MJ (2000) The evolutionary origins of<br />

Epichloë endophytes from annual ryegrasses. Mycologia 92:1103–1118<br />

Moubarak AS, Piper EL, West CP, Johnson ZB (1993) Interaction of purified ergovaline<br />

from endophyte-infected tall fescue with synaptosomal ATPase enzyme system. J<br />

Agric Food Chem 41:407–409


10 Development Interactions Between Clavicipitaleans and Their Host Plants 177<br />

Moy M, Belanger F, Duncan R, Freehof A, Leary C, Meyer W, Sullivan R,White JF Jr (2000)<br />

Identification of epiphyllous mycelial nets on leaves of grasses infected by clavicipitaceous<br />

endophytes. Symbiosis 28:291–302<br />

Oliver R, Osbourn A (1995) Molecular dissection of fungal phytopathogenicity. Microbiology<br />

141:1–9<br />

Papavizas GC (1985) Trichoderma and Gliocladium: biology, ecology, and potential for<br />

biocontrol. Annu Rev Phytopathol 23:23–54<br />

Peberdy JF (1990) Fungal cell walls – a review. In: Kuhn PJ, Trinci APJ, Jung MJ, Goosey<br />

MW, Copping LG (eds) Biochemistry of cell walls and membranes in fungi. Springer,<br />

Berlin Heidelberg New York<br />

Porter JK, Bacon CW, Cutler HG,Arrendale RF, Robbins JD (1985) In vitro auxin production<br />

by Balansia epichloë. Phytochemistry 24:1429–1431<br />

Reddy PV, Lam CK, Belanger FC (1996) Mutualistic fungal endophytes express a proteinase<br />

which is homologous to proteases suspected to be important in fungal pathogenicity.<br />

Plant Physiol 111:1209–1218<br />

Rykard DM, Bacon CW, Luttrell ES (1985) Host relations of Myriogenospora atramentosa<br />

and Balansia epichloë (Clavicipitaceae). Phytopathology 75:950–956<br />

Schaeffer JH, Leykam J,Walton JD (1994) Cloning and targeted gene disruption of EXG1,<br />

encoding exo-1,3-glucanase, in the phytopathogenic fungus Cochliobolus carbonum.<br />

Appl Environ Microbiol 60:594–598<br />

Schardl CL, Wilkinson HH (2000) Hybridization and cospeciation hypotheses for the<br />

evolution of grass endophytes. In: Bacon CW,White JF Jr (eds) Microbial endophytes.<br />

Marcel-Dekker, New York, pp 63–83<br />

Scott-Craig JS, Panaccione DG, Cervone F, Walton JD (1990) Endopolygalacturonase is<br />

not required for pathogenicity of Cochliobolus carbonum on maize. Plant Cell<br />

2:1191–1200<br />

Sposato P, Ahn J-H, Walton JD (1995) Characterization and disruption of a gene in the<br />

maize pathogen Cochliobolus carbonum encoding a cellulose binding domain and<br />

hinge region. Mol Plant-Microbe Interact 8:602–609<br />

St. Leger RJ (1995) The role of cuticle-degrading proteases in fungal pathogenesis of<br />

insects. Can J Bot 73:1119–1125<br />

Sullivan RF, Bills GF, Hywel-Jones NL, White JF Jr (2000) Hyperdermium: a new clavicipitalean<br />

genus for some tropical epibionts of dicotyledonous <strong>plant</strong>s. Mycologia 92:908–<br />

919<br />

Tenberge KB, Homann V, Oeser B, Tudzynski P (1996) Structure and expression of two<br />

polygalacturonase genes of Claviceps purpurea oriented in tandem and cytological<br />

evidence for pectinolytic enzyme activity during infection of rye. Phytopathology<br />

86:1084–1097<br />

Tsai H-F, Liu J-S, Staben C, Christensen MJ, Latch GCM, Siegel MR, Schardl CL (1994)<br />

Evolutionary diversification of fungal endophytes of tall fescue grass by hybridization<br />

with Epichloë species. Proc Natl Acad Sci USA 91:2542–2546<br />

Tzean SS, Hsieh LS, Wu WJ (1997) Atlas of entomopathogenic fungi from taiwan. Council<br />

of Agriculture, Yuan, Taiwan, Republic of China<br />

White JF Jr (1987) Widespread distribution of endophytes in the Poaceae. Plant Dis<br />

71:340–342<br />

White JF Jr (1988) Endophyte-host associations in forage grasses. XI. A proposal concerning<br />

origin and evolution. Mycologia 80:442–446<br />

White JF Jr, Cole GT (1986) Endophyte-host associations in forage grasses. IV. The endophyte<br />

of Festuca versuta. Mycologia 78:102–107<br />

White JF Jr, Bultman TL (1987) Endophyte-host associations in forage grasses.VIII. Heterothallism<br />

in Epichloë typhina. Am J Bot 74:1716–1721


178<br />

James F. White Jr. et al.<br />

White JF Jr, Chambless DA (1991) Endophyte-host associations in forage grasses. XV.<br />

Clustering of stromata-bearing individuals of Agrostis hiemalis infected by Epichloë<br />

typhina. Am J Bot 78:527–533<br />

White JF Jr, Owens JR (1992) Stromal development and mating system of Balansia<br />

epichloë, a leaf-colonizing endophyte of warm-season grasses. Appl Environ Microbiol<br />

58:513–519<br />

White JF Jr, Glenn AE (1994) A study of two fungal epibionts of grasses: structural features,<br />

host relationships, and classification in genus Myriogenospora Atk. (Clavicipitales).<br />

Am J Bot 81:216–223<br />

White JF Jr, Camp CR (1996) A study of water relations of Epichloë amarillans White, an<br />

endophyte of the grass Agrostis hiemalis (Walt.) B.S.P. Symbiosis 18:15–25<br />

White JF Jr, Breen JP, Morgan-Jones G (1991) Substrate utilization in selected Acremonium,<br />

Atkinsonella, and Balansia species. Mycologia 83:601–610<br />

White JF Jr, Morrow AC, Morgan-Jones G, Chambless DA (1991) Endophyte-host associations<br />

in forage grasses. XIV. Primary stromata formation and seed transmission in<br />

Epichloë typhina: developmental and regulatory aspects. Mycologia 83:72–81<br />

White JF Jr, Martin TI, Cabral D (1996) Endophyte-host associations in grasses. XXIII.<br />

Conidia formation by Acremonium endophytes in the phylloplanes of Agrostis<br />

hiemalis and Poa rigidifolia. Mycologia 88:174–178<br />

White JF Jr, Bacon CW, Hinton DM (1997) Modifications of host cells and tissues by the<br />

biotrophic endophyte Epichloë amarillans (Clavicipitaceae; Ascomycotina). Canadian<br />

J Bot 75:1061–1069<br />

Yue C, Miller CJ, White JF, Richardson M (2000) Isolation and characterization of fungal<br />

inhibitors from Epichloë festucae. J Agric Food Chem 48:4687–4692


11 Interactions of Microbes with Genetically<br />

Modified Plants<br />

Michael Kaldorf, Chi Zhang, Uwe Nehls, Rüdiger Hampp<br />

and François Buscot<br />

1 Introduction<br />

The introduction of molecular biological methods into <strong>plant</strong> breeding has<br />

offered the possibility to construct genetically modified <strong>plant</strong>s (GMPs) with<br />

new qualities. Major goals of genetic engineering are the improvement of<br />

product quality as well as the enhancement of resistance or tolerance to<br />

pathogen infections, herbicides and abiotic stress factors.<br />

Attempts to improve the quality of agricultural products include the<br />

manipulation of the softening of fruits like strawberry (Jimenez-Bermudez et<br />

al. 2002) and tomato (Quiroga and Fraschina 1997) in order to allow longer<br />

storage after harvesting, the modification of oil composition of oilseed crops<br />

(Thelen and Ohlrogge 2002), or the elevation of the provitamin A content of<br />

rice (Ye et al. 2000) and tomato (Romer et al. 2000). Even in forestry, increased<br />

wood production and quality are of great commercial interest (Mullin and<br />

Bertrand 1998). For example, the lignin content of transgenic aspen, in which<br />

the lignin biosynthesis pathway was downregulated by antisense inhibition,<br />

was greatly reduced (Hu et al. 1999), indicating that some technical limitations<br />

for the use of these fast growing trees for cellulose fiber production (e.g., in<br />

paper industry) might be reduced by genetic engineering.<br />

In contrast to the examples given above, the basic target of constructing<br />

GMPs with enhanced resistance to biotic or abiotic stress factors is not a modified<br />

product quality, but an enhanced productivity and reduction of the production<br />

costs in agriculture and forestry. The possibility to overcome different<br />

types of abiotic stress in GMPs has been demonstrated in several<br />

experiments [e.g., drought-resistant sugar beet (Pilon-Smits et al. 1999), salttolerant<br />

tomato <strong>plant</strong>s (Zhang and Blumwald 2001), or aluminium-resistant<br />

Brassica napus <strong>plant</strong>s (Basu et al. 2001)], but until now, none of these GMPs is<br />

being used for commercial production.<br />

All GMPs introduced on a large scale into agriculture in the 1990s possess<br />

resistance genes either against herbicides or against <strong>plant</strong> pathogens. Many<br />

different herbicide-resistant transgenic <strong>plant</strong>s like corn, cotton, lettuce,<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


180<br />

Michael Kaldorf et al.<br />

poplar, potato, rapeseed, soybean, sugar beet, tobacco, tomato, and wheat have<br />

been developed and field-tested (Saroha et al. 1998). Especially soybean, corn<br />

and oilseed resistant to the herbicide glyphosate were <strong>plant</strong>ed on at least<br />

15 million ha in the USA, Argentina, Canada and other countries since 1998,<br />

occupying about 70 % of the total release area for GMPs in 1998 (Warwick et<br />

al. 1999; Owen 2000). Among the pathogen-resistant GMPs, transgenic corn<br />

(Owen 2000), cotton (Perlak et al. 2001) and other <strong>plant</strong>s producing insecticidal<br />

proteins from Bacillus thuringiensis (Bt) are grown on a similar scale<br />

(11.4 million ha worldwide in 2000, Shelton et al. 2002) as herbicide-resistant<br />

GMPs. Transgenic <strong>plant</strong>s with different resistances to many other viral, bacterial<br />

and fungal pathogens have been described (e.g., Düring et al. 1993; Punja<br />

2001; Solomon-Blackburn and Barker 2001, and references cited therein),<br />

which are not yet used commercially.<br />

Since the first field experiment in 1986, more than 25,000 field trials with<br />

GMPs have been performed worldwide (Warwick et al. 1999), and for all<br />

GMPs, some common features have to be demonstrated in field experiments<br />

prior to commercial production. First, the new quality of the transgenic <strong>plant</strong><br />

must be stable under field conditions. Second, all other characteristics important<br />

for the agricultural use of a <strong>plant</strong> should remain unaffected in the GMPs<br />

compared to their parental breeds. Third, negative effects on the environment<br />

and particularly on nontarget organisms have to be low or missing. Such nontarget<br />

effects include cases like the negative and – in the worst case – lethal<br />

impact of Bt transgenic corn pollen on larvae of the monarch butterfly (Losey<br />

et al. 1999; Hansen Jesse and Obrycki 2000), which correspond to environmental<br />

risks without direct influence on the performance of the GMPs in the<br />

field.A further category of nontarget effects includes reduced compatibility of<br />

GMPs in symbiotic interactions or damage of <strong>plant</strong> growth promoting bacteria,<br />

which are not only environmental risks, but might also reduce the productivity<br />

of GMPs in the field. Especially in the case of pathogen-resistant<br />

GMPs, a negative impact on nontarget organisms is likely and has to be investigated<br />

thoroughly prior to the decision to use a GMP commercially.<br />

The aim of this review is to summarize the effects of transgenic <strong>plant</strong>s on<br />

nontarget microorganisms. Depending on the specific characters of a GMP,<br />

these effects might be positive (e.g., enrichment of <strong>plant</strong> growth-promoting<br />

bacteria), neutral, or even negative (e.g., increase in <strong>plant</strong> pathogenic bacteria<br />

or fungi). Experimental work in this field can be grouped into three categories:<br />

(1) analysis of effects of GMPs on changes in microorganism communities<br />

at the root <strong>surface</strong> and in the rhizosphere; (2) investigations that focus<br />

on positive interactions between <strong>plant</strong>s and microorganisms like mycorrhizal<br />

or Rhizobium symbioses that are important factors for <strong>plant</strong> nutrition and<br />

health; (3) analysis of horizontal gene transfer (HGT) events as a result of<br />

tight interactions between GMPs and microorganisms.


11 Interactions of Microbes with Genetically Modified Plants 181<br />

2 Changes in Microbial Communities Induced by<br />

Genetically Modified Plants<br />

Many studies dealing with the impact of GMPs on microbial communities<br />

have been published since the urgent call of Morra (1994) for this kind of<br />

research (Table 1). At present, transgenic potatoes producing T4 lysozyme to<br />

obtain resistance to Erwinia carotovora and other bacterial pathogens<br />

(Düring et al. 1993) are the best characterized GMP system with respect to<br />

microbial interactions. In addition, some of the commercially most important<br />

GMPs like corn and cotton producing the insecticidal Bacillus thuringiensi<br />

(Bt) toxin, or glyphosate-tolerant Brassica sp., as well as different other GMPs<br />

have been tested.<br />

Selective advantages of specific bacteria in the rhizosphere of GMPs were<br />

demonstrated in two studies. A T4 lysozyme-tolerant Pseudomonas putida<br />

strain with antibacterial activity to the bacterial pathogen Erwinia carotovora<br />

was introduced into the rhizosphere of T4 lysozyme-producing potatoes.<br />

During flowering of the <strong>plant</strong>s, when the highest lysozyme level was detected<br />

in <strong>plant</strong>a, significantly more colonies of the introduced P. putida strain could<br />

be reisolated from transgenic potatoes compared to controls (Lottmann et al.<br />

2000). In the second experiment, the culture of transgenic Lotus corniculatus<br />

<strong>plant</strong>s producing different opines led to a significant increase in opine-utilizing<br />

bacteria in the rhizosphere. When the opine-producing <strong>plant</strong>s were<br />

replaced by nontransgenic <strong>plant</strong>s, the concentration of different fractions of<br />

opine utilizers in the soil slowly decreased over 22 weeks. However, even then,<br />

the density of the bacterial fraction specifically using mannopine was still five<br />

times higher than in soil which had not been <strong>plant</strong>ed with the transgenic<br />

Lotus <strong>plant</strong>s (Oger et al. 2000). This demonstrates that alterations in the soil<br />

microflora induced by the cultivation of GMPs may be persistent.<br />

Nontarget effects of GMPs on bacterial communities seem to be common.<br />

A broad spectrum of both physiological and molecular biological methods<br />

like community-level physiological profile (CLPP; Griffiths et al. 2000; Dunfield<br />

and Germida 2001), BIOLOG substrate utilization test (Siciliano et al.<br />

1998; Di Giovanni et al. 1999), fatty acid methyl ester (FAME) patterns (Siciliano<br />

et al. 1998; Dunfield and Germida 2001), plate-counting of bacterial<br />

groups (e.g., spore-forming or cellulose-utilizing bacteria, Donegan et al.<br />

1999) and T-RFLP (Lukow et al. 2000) have been used to characterize bacterial<br />

communities associated with GMPs. In addition, population analyses of<br />

protozoa, nematodes and microarthropods have been included in some studies<br />

(e.g., Donegan et al. 1997; Saxena and Stotzky 2001). Significant changes in<br />

the rhizosphere of different GMPs have been shown (see Table 1), which are<br />

not necessarily linked directly to the presence of new gene product(s). For<br />

example, changes in the rhizospheric bacterial community of transgenic cotton<br />

<strong>plant</strong>s producing the Bt toxin were significant, but the purified Bt toxin<br />

itself displayed no detectable effect on soil microorganisms (Donegan et al.


182<br />

Table 1: Studies assessing the impact of GMPs on <strong>plant</strong>-associated and soil microorganisms<br />

Michael Kaldorf et al.<br />

GMP species, Group(s) of organisms investigated Observations Reference<br />

new characteristics<br />

Brassica sp., tolerance to the Soil bacterial communities Indications for changes in the soil bacterial Siciliano et al. (1998)<br />

herbicide glyphosate communities<br />

Brassica sp., tolerance to the Soil bacterial communities Significant changes in the soil bacterial Dunfield and Germida<br />

herbicide glyphosate communities (2001)<br />

Gossypium hirsutum, Bt toxin Leaf material decomposing bacterial, Two of three transgenic cotton lines caused Donegan et al. (1995)<br />

production resistance to insects fungal, and protozoan populations significant stimulation and qualitative changes<br />

of bacterial and fungal populations<br />

Lotus corniculatus, Opine-utilizing bacteria, soil Increase of opine-utilizing bacteria in the rhizo- Oger et al. (2000)<br />

opine production bacterial community sphere of the GMPs; effect persistent for 22 weeks<br />

Medicago sativa, a-amylase or Soil bacteria, fungi, protozoa, Significant changes in bacterial populations of Donegan et al. (1999)<br />

lignin peroxidase production nematodes and micro-arthropods lignin peroxidase <strong>plant</strong>s; population levels of<br />

fungi, protozoa, nematodes and microarthropods<br />

not affected<br />

Medicago sativa, a-amylase or Soil bacterial communities Significant changes in bacterial populations Di Giovanni et al.<br />

lignin peroxidase production associated with lignin peroxidase <strong>plant</strong>s (1999)<br />

Nicotiana tabaccum, proteinase Protozoa, nematodes, and Changes in nematode populations, reduced Donegan et al. (1997)<br />

inhibitor I, resistance to insects microarthropods Collembola populations on litter from GMPs<br />

Solanum tuberosum, T4 lysozyme Plant-associated bacteria Minor effects on community structure Lottmann et al. (1999)<br />

production, resistance to bacteria<br />

Solanum tuberosum, Introduced, pathogen-antagonistic Significant increase of introduced lysozyme- Lottmann et al. (2000)<br />

T4 lysozyme production bacteria with high lysozyme tolerance tolerant bacteria on GMPs<br />

Solanum tuberosum, Pseudomonads and enterics from No correlation between phenotypic or genotypic Lottmann and Berg<br />

T4 lysozyme production the rhizosphere profile and transgenic character (2001)


GMP species, Group(s) of organisms investigated Observations Reference<br />

new characteristics<br />

11 Interactions of Microbes with Genetically Modified Plants 183<br />

Solanum tuberosum, Rhizosphere bacterial community Only minor effects of lysozyme Heuer and Smalla<br />

T4 lysozyme production (1999)<br />

Solanum tuberosum, Rhizosphere bacterial community No detectable effects of lysozyme production Heuer et al. (2002)<br />

T4 lysozyme production<br />

Solanum tuberosum, Pathogenic Erwinia carotovara Similar lysozyme sensitivity of Erwinia and most de Vries et al. (1999)<br />

T4 lysozyme production strains; soil bacteria other soil bacteria<br />

Solanum tuberosum, Bacillus subtilis Increased killing of B. subtilis on the hairy roots Ahrenholtz et al. (2000)<br />

T4 lysozyme production of lysozyme-producing <strong>plant</strong>s<br />

Solanum tuberosum, Total bacterial and fungal populations Only minor effects of the GMPs Donegan et al. (1996)<br />

Bt toxin production colonizing leaves<br />

Solanum tuberosum, production Soil bacterial communities; protozoa No effect on protozoa, but significant changes Griffiths et al. (2000)<br />

of anti-feedant lectines in the physiological profiles of bacterial<br />

communities<br />

Solanum tuberosum, Barstar/ Soil bacterial community structure Some significant differences between GMPs Lukow et al. (2000)<br />

Barnase genes, fungal pathogen and control <strong>plant</strong>s<br />

resistance<br />

Zea mays, Bt toxin production Soil bacteria, fungi, protozoa, No significant differences detected Saxena and Stotzky<br />

and nematodes (2001)


184<br />

Michael Kaldorf et al.<br />

1995). Similarly, the effects of transgenic potatoes producing the lectin GNA<br />

on nontarget soil organisms could not be attributed to the formation of the<br />

lectin GNA itself (Griffiths et al. 2000). So far, only one GMP producing an<br />

antibacterial substance, namely T4 lysozyme-producing potatoes with<br />

enhanced resistance to Erwinia carotovora, has been investigated in detail for<br />

nontarget effects on soil bacteria. Most soil bacteria were lysozyme-sensitive<br />

when tested in laboratory experiments with pure cultures (de Vries et al.<br />

1999). In addition, increased killing of Bacillus subtilis was observed on the<br />

root <strong>surface</strong> of T4 lysozyme-producing potatoes from the field, and this effect<br />

was ascribed directly to the lysozyme release by the roots (Ahrenholtz et al.<br />

2000). Nevertheless, the production of lysozyme had only a minor influence<br />

on the bacterial phyllo- and rhizosphere communities (Heuer and Smalla<br />

1999; Heuer et al. 2002), which was considered negligible relative to natural<br />

factors by the authors. Further studies with the same system which focused on<br />

potentially beneficial <strong>plant</strong>-associated microbes like auxin-producing bacteria<br />

or bacteria antagonistic to the pathogenic E. carotovora did not reveal correlations<br />

between the transgenic character of <strong>plant</strong>s and the pheno- or genotypic<br />

features of bacterial isolates (Lottmann and Berg 2001). Thus, up to now,<br />

there is no direct evidence from field experiments that the primary product of<br />

transgene expression is responsible for significant changes in the soil microbial<br />

community in any GMP. Instead, secondary effects of GMP generation,<br />

like somaclonal variation and changes in general <strong>plant</strong> metabolism induced<br />

by the transgene insertion or expression, may contribute to a major part of<br />

the effects described above.<br />

3 Impact of Genetically Modified Plants on Symbiotic<br />

Interactions<br />

The question whether the genetical transformation of <strong>plant</strong>s might reduce<br />

their ability to form mutualistic symbioses with microorganisms has been<br />

addressed in a surprisingly small number of studies.<br />

Biological nitrogen fixation accounts for about 65 % of the nitrogen utilized<br />

in agriculture worldwide (Vance and Graham 1995). The ability to<br />

reduce atmospheric nitrogen to ammonia (nitrogen fixation) is restricted to<br />

prokaryotes. Beside free-living and <strong>plant</strong>-associated bacteria, members of the<br />

Rhizobiaceae living symbiotically in the typical root nodules of legumes such<br />

as alfalfa, clover, pea, and soybean are the agriculturally most important<br />

group of nitrogen fixing organisms. The symbiotic interaction between rhizobia<br />

and legumes requires a sequential signal exchange between both partners,<br />

and therefore, exhibits a high degree of host specificity (Bothe 1993). Transgenic<br />

<strong>plant</strong>s have been used as a tool to investigate the host recognition of rhizobia<br />

(Diaz et al. 1989, 2000). For example, the transfer of lectin genes between<br />

different legumes has been shown as a possible way to modify host specificity


11 Interactions of Microbes with Genetically Modified Plants 185<br />

(Diaz et al. 2000; van Rhijn et al. 2001). While the GMPs used in these studies<br />

have been modified specifically to change the <strong>plant</strong>–rhizobia interactions,<br />

possible alterations in the rhizobia symbiosis would be untargeted in<br />

pathogen- or herbicide-resistant legumes. The constitutive expression of a<br />

rice basic chitinase gene with putative antifungal effects in alfalfa had no negative<br />

influence on the interaction with Rhizobium (Masoud et al. 1996). No<br />

further information is available about interference between genetical modifications<br />

of <strong>plant</strong>s and nitrogen fixing bacteria, particularly about the herbicide-resistant<br />

soybean cultivars <strong>plant</strong>ed on a large scale since 1996.<br />

The second important group of <strong>plant</strong>-microbe symbioses is the mycorrhiza.<br />

Under natural conditions, the roots of most <strong>plant</strong>s are colonized by<br />

mycorrhizal fungi, which increase their uptake of water and nutrients, as well<br />

as their resistance against pathogens and abiotic stress (Smith and Read<br />

1997). Ectomycorrhiza (EM) is the dominating type of mycorrhiza in gymnosperms<br />

and many woody angiosperms. EM formation is accompanied by<br />

morphological changes of both the fungal hyphae and the <strong>plant</strong> fine roots.<br />

Typically, hyphae form a mantle of varying thickness around the fine roots.<br />

From there they extend into the apoplast of the root cortex, forming a highly<br />

branched network and thus establishing a large <strong>surface</strong> area for solute<br />

exchange, the Hartig net (Kottke and Oberwinkler 1986). Arbuscular mycorrhiza<br />

(AM) can be found in mosses, ferns and many angiosperms, including<br />

agronomically important <strong>plant</strong>s like barley, corn, potato, rice, soybean, and<br />

wheat (Smith and Read 1997). The successful use of transgenic host <strong>plant</strong>s in<br />

basic research on mycorrhiza has demonstrated that the use of common molecular<br />

biological methods, like the introduction of antibiotic resistance genes<br />

[e.g., transgenic aspen carrying a hygromycin resistance gene in addition to<br />

indoleacetic acid-biosynthetic genes, (Tuominen et al. 1995; Hampp et al.<br />

1996)] or reporter gene constructs (e.g., the gus reporter gene system, Gianinazzi-Pearson<br />

et al. 2000) into <strong>plant</strong>s, has normally no impact on mycorrhiza<br />

formation. Only in the case of the symbiosis-related gene enod40 from Medicago<br />

truncatula, overexpression of the gene accelerated AM colonization,<br />

while transgenic lines with suppressed enod40 transcript levels exhibited<br />

reduced mycorrhization (Staehelin et al. 2001).<br />

Negative nontarget effects of GMPs on mycorrhizal fungi seem to be most<br />

likely in GMPs constitutively expressing antifungal proteins in order to obtain<br />

resistance against fungal pathogens (Glandorf et al. 1997). Transgenic Nicotiana<br />

sylvestris <strong>plant</strong>s with more than tenfold enhanced chitinase activity<br />

were significantly less colonized by the fungal pathogen Rhizoctonia solani<br />

compared to control <strong>plant</strong>s. However, neither the quantity of AM colonization<br />

nor the anatomy of AM hyphae, arbuscules or vesicles were significantly<br />

affected in the chitinase overproducing <strong>plant</strong>s (Vierheilig et al. 1993). In a further<br />

study, several pathogenesis-related (PR) proteins were constitutively<br />

expressed in transgenic tobacco <strong>plant</strong>s to investigate their influence on the<br />

AM fungus Glomus mosseae (Vierheilig et al. 1995). Two acidic and two basic


186<br />

Michael Kaldorf et al.<br />

chitinases, one acidic and two basic glucanases, as well as three PR proteins of<br />

unknown function had no detectable influence on AM colonization. Only one<br />

of the PR proteins tested, an acidic, extracellular b-1,3-glucanase of class II,<br />

reduced the mycorrhization of transgenic tobacco roots by G. mosseae significantly.<br />

This observation demonstrates that mycorrhiza formation could be<br />

affected in GMPs expressing antifungal PR proteins. Therefore, a case-to-case<br />

investigation of GMPs with increased fungal pathogen resistance seems to be<br />

necessary to exclude negative effects on AM formation.<br />

In addition to the quantity of mycorrhizal colonization, the structural and<br />

functional diversity of mycorrhizal fungi colonizing the root system might be<br />

influenced in GMPs. Probably due to methodical difficulties, this question has<br />

not been addressed for AM fungi. Compared to the rather uniform morphology<br />

of AM fungi, which makes their morphological identification quite difficult,<br />

EM fungi exhibit many morphological and anatomical characters that<br />

could be used for their characterization (Agerer 1991). In combination with<br />

PCR-RFLP and sequence analysis of the ITS region within the fungal rDNA<br />

(Buscot et al. 2000), EM communities can be described with a sufficient resolution<br />

to compare the mycorrhization of transgenic and nontransgenic trees,<br />

even in the field.A release experiment with transgenic aspen carrying the rolC<br />

gene from Agrobacterium rhizogenes (Fladung and Muhs 2000) was accompanied<br />

by a detailed analysis of the EM status of the trees. Although rolC modified<br />

the hormonal balance in the trees, and therefore, might have affected<br />

their mycorrhization ability, no significant difference in the degree of mycorrhization<br />

was observed in the transformed aspen. The structure of the EM<br />

community of the different aspen lines was similar in the first two years of the<br />

experiment (Kaldorf et al. 2002), but in the third and fourth years, a significantly<br />

reduced EM diversity was observed on the rolC transgenic aspen compared<br />

to controls (Kaldorf et al. 2001). In addition, one EM morphotype<br />

formed by Phialocephala fortinii appeared to be significantly less represented<br />

on the transgenic line “E2/5” compared to all other transgenic and control<br />

lines (Kaldorf et al. 2002). This reduced compatibility for one mycobiont represents<br />

the first example of a clone-specific effect concerning mycorrhization<br />

of transgenic <strong>plant</strong>s.<br />

4 Horizontal Gene Transfer<br />

Three potential pathways have been proposed for the spread of GMPs or the<br />

transgenes introduced into these <strong>plant</strong>s. Two of these pathways, the establishment<br />

of self-sustaining GMP populations and the introgression of genes into<br />

wild populations, regarded as the major risks of GMPs for natural <strong>plant</strong> communities<br />

(Wolfenbarger and Phifer 2000), do not involve <strong>plant</strong>/microorganism<br />

interactions. The third possibility is the horizontal transfer of genes from<br />

GMPs to microorganisms, which might lead to bacterial or fungal strains car-


11 Interactions of Microbes with Genetically Modified Plants 187<br />

rying genes from GMPs. The exchange of genetic information between different<br />

bacterial species by transformation, transduction or conjugation seems to<br />

be common in nature (Krishnapillai 1996; Wöstemeyer et al. 1997 and references<br />

therein). The detailed analysis of DNA and amino acid sequence data<br />

has indicated that horizontal transmission of genes, even between bacteria<br />

and eukaryotes or between eukaryotes from different systematic kingdoms,<br />

probably occurred in rare cases during evolution (Dröge et al. 1998 and references<br />

therein), but there is no experimental access to further investigation or<br />

verification of such horizontal gene transfer (HGT) events.<br />

The focus of the experimental work on HGT has been the question whether<br />

antibiotic resistance genes, used as selectable markers in GMPs, can be transferred<br />

to bacteria, enhancing the frequency of antibiotic-resistant bacteria of<br />

medical importance (Nielsen et al. 1998). Beside the relevance of this question<br />

for the risk assessment of GMPs, the transfer of antibiotic-resistance genes is<br />

easy to detect compared to a possible horizontal transfer of genes which cannot<br />

be used as a selectable marker for the isolation of transformed bacteria.<br />

Therefore, experimental data about the possible HGT of other genes are<br />

scarce.<br />

The transformation of different bacterial species has been demonstrated<br />

under optimized laboratory conditions using isolated plasmid DNA, total<br />

DNA from GMPs and even homogenized <strong>plant</strong> material from GMPs as the<br />

source for antibiotic-resistance genes (Schlüter et al. 1995; Gebhard and<br />

Smalla 1998). The efficiency of the integration of the nptII gene, causing resistance<br />

to kanamycin, into the genome of Acinetobacter sp. strongly depended<br />

on the presence of homologous sequences in the bacterial DNA (Nielsen et al.<br />

1997). This observation was confirmed by de Vries et al. (2001) using Acinetobacter<br />

sp. and Pseudomonas stutzeri as well as by Bertolla et al. (2000) using<br />

the <strong>plant</strong> pathogenic bacterium Ralstonia solanacearum as recipient for<br />

antibiotic-resistance genes.<br />

While transformation of bacteria is common under optimized laboratory<br />

conditions, all experiments under natural conditions indicated that the frequency<br />

of HGT is drastically reduced compared to optimized conditions.<br />

Although DNA from transgenic <strong>plant</strong>s can persist in soil for up to 2 years<br />

(Gebhard and Smalla 1999), the availability of DNA from GMPs could be a<br />

limiting factor for HGT. Even under otherwise optimized conditions (e.g., use<br />

of purified DNA from transgenic sugar beet as source for the nptII gen, construction<br />

of an Acinetobacter strain carrying a deleted nptII gene to allow<br />

homologous recombination in the recipient bacteria), the frequency of HGT<br />

was low in sterilized soil microcosms and below the detection limit in nonsterilized<br />

soil (Nielsen et al. 2000). In a field release experiment with nptIItransgenic<br />

sugar beet, a total of 4000 kanamycin-resistant colonies of soil bacteria<br />

isolated from the field release site was checked for the presence of the<br />

nptII gene from the transgenic <strong>plant</strong>s by dot blot hybridization and PCR.<br />

None of the isolates carried the nptII gene, indicating a natural kanamycin


188<br />

Michael Kaldorf et al.<br />

resistance of all strains tested, which was not acquired by HGT (Gebhard and<br />

Smalla 1999). Thus, the conclusion of Bertolla and Simonet (1999) that we are<br />

a long way from demonstrating that <strong>plant</strong>–bacterium gene transfer does<br />

occur under natural conditions, is still valid.<br />

Approaches to detect HGT from transgenic <strong>plant</strong>s to eukaryotic microorganism<br />

are sparse. Particular fungi often grow in intimate contact with <strong>plant</strong>s<br />

or – in the case of endoparasitic and mycorrhizal symbioses – even within<br />

<strong>plant</strong>s. In these cases, uptake of <strong>plant</strong> DNA by fungi might be more likely compared<br />

to soil bacteria, as the <strong>plant</strong> DNA does not come in contact with soil.<br />

Indeed, evidence has been presented that the phytopathogenic fungus Plasmodiophora<br />

brassicae takes up host <strong>plant</strong> DNA during each infection cycle<br />

(Bryngelsson et al. 1988), but interactions between Plasmodiophora and<br />

transgenic Brassica sp. have not been investigated. HGT from <strong>plant</strong>s to saprophytic<br />

fungi has also been reported (Hoffmann et al. 1994). Transgenic <strong>plant</strong>s<br />

expressing the hygromycin gene (hph) as selection marker under control of a<br />

fungal promoter were generated. After cocultivation of dead <strong>plant</strong> material<br />

together with Aspergillus, fungal progenies were isolated that revealed resistance<br />

to hygromycin B on selective agar plates (Hoffmann et al. 1994). The hph<br />

gene and other foreign DNA sequences could be detected in some of these<br />

hygromycin B-resistant fungal strains. Nevertheless, in most cases the foreign<br />

DNA was not stably integrated into the Aspergillus genome.<br />

In the following, we present some unpublished data evaluating the possibility<br />

of HGT in mycorrhizal symbioses. Ectomycorrhizas are of special interest<br />

in this aspect, due to the long life time of the host trees. Therefore, there is<br />

a need to investigate the possibility of HGT from transformed forest trees to<br />

EM fungi. Plant cells frequently die during EM interaction and thus, fungal<br />

hyphae of the Hartig net come in close contact with <strong>plant</strong> DNA. Filamentous<br />

fungi are naturally not very competent in the uptake of large DNA fragments.<br />

In EM however, hyphae of the Hartig net are coenocytic and have a highly<br />

enlarged plasma membrane <strong>surface</strong> area due to extensive invaginations (Kottke<br />

and Oberwinkler 1987). Therefore, they might be more competent for<br />

DNA uptake than normal hyphae.<br />

Two different approaches were used to study HGT between <strong>plant</strong> and fungal<br />

cells in ectomycorrhizas. In the first approach, transgenic aspen carrying<br />

the rolC gene from Agrobacterium rhizogenes under control of the lightinducible<br />

rbcS promoter from potato (Fladung et al. 1997) were grown in vitro<br />

together with the ectomycorrhizal ascomycete Phialocephala fortinii strain<br />

5B, isolated from mycorrhizal aspen roots collected in the field (Fladung et al.<br />

2000).After 12 weeks of cocultivation, P. fortinii was reisolated from colonized<br />

aspen roots. Fungal hyphae growing out from mycorrhizas were transferred<br />

to fresh medium for further growth to avoid contamination with <strong>plant</strong> material.<br />

Genomic DNA was isolated from the fungal mycelium and analyzed for<br />

the presence of the rolC gene. To enhance the sensitivity of the PCR assay, a<br />

“nested” PCR strategy was followed. The first rolC specific primer pair should


11 Interactions of Microbes with Genetically Modified Plants 189<br />

amplify a 950-bp DNA fragment. In a second PCR step, a 500-bp fragment of<br />

rolC should be amplified from the products of the first PCR using a second<br />

inner primer pair, again specific for rolC. Isolated DNA from transgenic aspen<br />

leaves was used as positive control for the nested PCR, while the quality of the<br />

fungal DNA was checked with the primer pair ITS1/ITS4 (White et al. 1990),<br />

specific for a part of fungal rDNA clusters. The rolC gene was not detected in<br />

any of the 24 Phialocephala colonies analyzed. The number of 24 samples was<br />

sufficient to demonstrate that the uptake of <strong>plant</strong> DNA in Phialocephala EM<br />

does not occur on a regular basis, as suggested for the Plasmodiophora–Brassica<br />

interaction (Bryngelsson et al. 1988).<br />

The second approach was with transgenic <strong>plant</strong>s that contained a small<br />

marker gene, which could confer herbicide resistance into the target organism<br />

after HGT. The advantage of this strategy is that a large number of samples<br />

can be simultaneously screened, but only a small number of the samples able<br />

to grow on the selection medium have to be investigated in detail. In order to<br />

monitor HGT in ectomycorrhizas formed between poplar and Amanita muscaria,<br />

a 1250-bp EcoRI/XbaI fragment of pBG (Straubinger et al. 1992) containing<br />

the Streptomyces hygroscopicus bar gene under the control of the<br />

Cochlibolus heterostrophus GPD1 promoter was inserted into the Agrobacterium<br />

vector pBI121 (Clontech, Palo Alto, CA, USA; Fig. 1). The function of<br />

amp<br />

Fig. 1. Cloning strategy for the<br />

construction of the binary vector<br />

pBI121/3<br />

pBG<br />

4.21 Kb<br />

A<br />

ori<br />

XbaI<br />

BamH1<br />

bar<br />

GPD1<br />

Nos-P<br />

RB<br />

Nos-ter<br />

BamH1<br />

NPTII (Kan)<br />

Pst1<br />

EcoR1<br />

EcoRV<br />

HindIII<br />

Cla1<br />

Xho1<br />

HindIII<br />

CaMV 35S P<br />

pBI121/3<br />

12.27 Kb<br />

C<br />

Bar-Gene<br />

Nos-ter<br />

Nos-P<br />

RB<br />

XbaI<br />

GPD1 P<br />

NPTII (Kan)<br />

LB<br />

EcoRI<br />

HindIII<br />

CaMV 35S P<br />

pBI121<br />

13.00 Kb<br />

B<br />

GUS<br />

XbaI<br />

Nos-ter<br />

LB<br />

SstI<br />

EcoRI


190<br />

Michael Kaldorf et al.<br />

the GPD/bar construct in the ectomycorrhizal fungus A. muscaria was previously<br />

verified by PEG-mediated protoplast transformation. Transgenic<br />

poplars containing the GPD/bar construct were generated by Agrobacteriummediated<br />

transformation. Twenty <strong>plant</strong>s were isolated that originate from different<br />

calli. PCR amplification was carried out with genomic DNA from transgenic<br />

poplars using bar-specific primers. PCR products of the expected size<br />

were obtained from 19 out of a total of 20 putative transgenic poplar <strong>plant</strong>s<br />

(Fig. 2). Isolated PCR-fragments of three clones were sequenced and revealed<br />

the introduced bar gene.<br />

For the investigation of a HGT event, 35,000 ectomycorrhizas formed<br />

between transgenic poplars and A. muscaria were isolated and transferred to<br />

selective agar plates.After the first round of selection, 102 putative Basta resistant<br />

fungal colonies were obtained. However, none of these colonies was able<br />

to grow after transfer to a fresh selection medium. Genomic DNA isolated<br />

from fungal hyphae initially growing on the selection medium was investigated<br />

for the presence of the bar-gene using PCR. No bar-fragment could be<br />

obtained from any of these investigated clones. The utilization of primers for<br />

Fig. 2. Analysis of genomic DNA isolated from putative kanamycin-resistant poplar<br />

transformants. PCR amplification was performed on genomic DNA using primers specific<br />

for the bar gene that amplifies an internal fragment of 550-bp length. Lanes 1 to 20<br />

Isolated DNA from putative transformants. P Positive control with diluted DNA of<br />

pBI121/3, K DNA isolated from a nontransformed poplar <strong>plant</strong>, M molecular size marker<br />

(l/HindIII DNA marker)


a single copy gene of A. muscaria (SCIV038, Nehls et al. 2001) revealed PCR<br />

fragments in any case, indicating that no inhibitors of the PCR reaction were<br />

present in the genomic DNA preparation. The reason for false positives was<br />

most probably the low herbicide concentration in the growth medium.A concentration<br />

of 200 mg/ml Basta (as used in this study) results in fungal background<br />

growth. This relatively low Basta-concentration was chosen to recognize<br />

also lateral transfer of the resistance gene lacking its heterologous<br />

promoter. In this case, the bar-gene might integrate behind a weak A. muscaria<br />

promoter, resulting in only a weak herbicide resistance.<br />

The 35,000 mycorrhizas investigated in this study represent, of course, only<br />

a limited sample number. Nevertheless, since each mycorrhiza does contain a<br />

large number of competent fungal hyphae in direct contact with <strong>plant</strong> epidermal<br />

cells that die under the selection conditions, this sample number is large<br />

enough to reveal that HGT from the tree to the fungal partner is a quite rare<br />

or maybe completely missing event in EM symbiosis, at least under axenic<br />

conditions.<br />

5 Conclusions<br />

11 Interactions of Microbes with Genetically Modified Plants 191<br />

Taken together, many of the studies cited above demonstrate that transgenic<br />

<strong>plant</strong>s can induce changes in soil microorganism communities. Nevertheless,<br />

the importance of these findings is unclear as in most studies, the modifications<br />

in the rhizosphere of GMPs were not compared to the natural variance<br />

in the rhizosphere of different <strong>plant</strong> breeds generated by conventional methods.<br />

For example, the mycorrhization capacity of modern wheat varieties with<br />

high pathogen resistance has been shown to be reduced (Hetrick et al. 1992).<br />

Such potentially negative effects would be considered unacceptable in the<br />

case of any GMP introduced into agriculture.<br />

Concerning the investigations on HGT, there is some evidence for the possibility<br />

of HGT not only between bacteria, but also between <strong>plant</strong>s and<br />

microorganisms. In soil, HGT must be a rare event, as several attempts to<br />

detect HGT in field experiments failed. Despite the missing evidence for HGT<br />

in the field, the possibility of HGT should be kept in mind for risk assessment.<br />

The question to be answered in a case-to-case consideration is whether a possible<br />

rare HGT of the introduced genes from GMPs to microorganisms might<br />

cause specific problems. This is unlikely if the transgene itself is common in<br />

nature. For example, a natural transfer of the rolC gene from Agrobacterium to<br />

other bacteria seems much more likely than a HGT from the rolC transgenic<br />

aspen mentioned above to microorganisms. On the other hand, artificial<br />

genes generated by genetic engineering might have a high risk potential when<br />

released into nature.


192<br />

Michael Kaldorf et al.<br />

References and Selected Reading<br />

Agerer R (1991) Characterization of ectomycorrhiza. In: Norris JR, Read DJ, Varma AK<br />

(eds) Methods in <strong>microbiology</strong>, vol 23. Academic Press, London, pp 25–73<br />

Ahrenholtz I, Harms K, de Vries J, Wackernagel W (2000) Increased killing of Bacillus<br />

subtilis on the hair roots of transgenic T4 lysozyme-producing potatoes. Appl Environ<br />

Microbiol 66:1862–1865<br />

Basu U, Good AG, Taylor GJ (2001) Transgenic Brassica napus <strong>plant</strong>s overexpressing aluminium-induced<br />

mitochondrial manganese superoxide dismutase cDNA are resistant<br />

to aluminium. Plant Cell Environ 24:1269–1278<br />

Bertolla F, Simonet P (1999) Horizontal gene transfers in the environment: natural transformation<br />

as a putative process for gene transfers between transgenic <strong>plant</strong>s and<br />

microorganisms. Res Microbiol 150:375–384<br />

Bertolla F, Pepin R, Passelegue-Robe E, Paget E, Simkin A, Nesme X, Simonet P (2000)<br />

Plant genome complexity may be a factor limiting in situ the transfer of transgenic<br />

<strong>plant</strong> genes to the phytopathogen Ralstonia solanacearum. Appl Environ Microbiol<br />

66:4161–4167<br />

Bothe H (1993) Metabolism of inorganic nitrogen compounds. Progress in Botany 54.<br />

Springer, Berlin Heidelberg New York, pp 201–217<br />

Bryngelsson T, Gustafsson M, Green B, Lind C (1988) Uptake of host DNA by the parasitic<br />

fungus Plasmodiophora brassicae. Physiol Mol Plant Pathol 33:163–171<br />

Buscot F, Munch JC, Charcosset JY, Gardes M, Nehls U, Hampp R (2000) Recent advances<br />

in exploring physiology and biodiversity of ectomycorrhizas highlight the functioning<br />

of these symbioses in ecosystems. FEMS Microbiol Rev 699:1–14<br />

de Vries J, Harms K, Broer I, Kriete G, Mahn A, Düring K,Wackernagel W (1999) The bacteriolytic<br />

activity of transgenic potatoes expressing a chimeric T4 lysozyme gene and<br />

the effect of T4 lysozyme on soil- and phytopathogenic bacteria. Syst Appl Microbiol<br />

22:280–286<br />

de Vries J, Meier P, Wackernagel W (2001) The natural transformation of the soil bacteria<br />

Pseudomonas stutzeri and Acinetobacter sp. by transgenic <strong>plant</strong> DNA strictly<br />

depends on homologous sequences in the recipient cells. FEMS Microbiol Lett 195:<br />

211–215<br />

Diaz CL, Melchers LS, Hooykaas PJJ, Lugtenberg EJJ, Kijne JW (1989) Root lectin as a<br />

determinant of host <strong>plant</strong> specificity in the Rhizobium-legume symbiosis. Nature<br />

338:579–581<br />

Diaz CL, Spaink HP, Kijne JW (2000) Heterologous rhizobial lipochitin oligosaccharides<br />

and chitin oligomers induce cortical cell divisions in red clover roots, transformed<br />

with the pea lectin gene. Mol Plant-Microbe Int 13:268–276<br />

Di Giovanni GD, Wartrud LS, Seidler RJ, Widmer F (1999) Comparison of parental and<br />

transgenic alfalfa rhizosphere bacterial communities using Biolog GN metabolic fingerprinting<br />

and enterobacterial repetitive intergenic consensus sequence-PCR<br />

(ERIC-PCR). Microb Ecol 37:129–139<br />

Donegan KK, Palm CJ, Fieland VJ, Porteous LA, Ganio LM, Schaller DL, Bucao LQ, Seidler<br />

RJ (1995) Changes in levels, species and DNA fingerprints of soil microorganisms<br />

associated with cotton expressing the Bacillus thuringiensis var. kurstaki endotoxin.<br />

Appl Soil Ecol 2:111–124<br />

Donegan KK, Schaller DL, Stone JK, Ganio LM, Reed G, Hamm PB, Seidler RJ (1996)<br />

Microbial populations, fungal species diversity and <strong>plant</strong> pathogen levels in field<br />

plots of potato <strong>plant</strong>s expressing the Bacillus thuringiensis var. tenebrionis endotoxin.<br />

Transgen Res 5:25–35<br />

Donegan KK, Seidler RJ, Fieland VJ, Schaller DL, Palm CJ, Ganio LM, Cardwell DM, Steinberger<br />

Y (1997) Decomposition of genetically engineered tobacco under field condi-


11 Interactions of Microbes with Genetically Modified Plants 193<br />

tions: persistence of the proteinase inhibitor I product and effects on soil microbial<br />

respiration and protozoa, nematode and microarthropod populations. J Appl Ecol<br />

34:767–777<br />

Donegan KK, Seidler RJ, Doyle JD, Porteus LA, di Giovanni G, Widmers F, Wartrud LS<br />

(1999) A field study with genetically engineered alfalfa with recombinant Sinorhizonium<br />

meliloti: effects on the soil ecosystem. J Appl Ecol 36:920–936<br />

Dröge M, Pühler A, Selbitschka W (1998) Horizontal gene transfer as a biosafety issue: a<br />

natural phenomenon of public concern. J Biotechnol 64:75–90<br />

Dunfield KE, Germida JJ (2001) Diversity of bacterial communities in the rhizosphere<br />

and root interior of field-grown genetically modified Brassica napus. FEMS Microbiol<br />

Ecol 38:1–9<br />

Düring K, Porsch P, Fladung M, Lörz H (1993) Transgenic potato <strong>plant</strong>s resistant to the<br />

phytopathogenic bacterium Erwinia carotovora. Plant J 3:587–598<br />

Fladung M, Muhs H-J (2000) Field release with Populus tremula (rolC-gene) in Großhansdorf.<br />

In: Umweltbundesamt (ed) Release of transgenic trees – present achievements,<br />

problems, future prospects. Humboldt-Universität, Berlin, pp 40–45<br />

Fladung M, Kumar S, Ahuja MR (1997) Genetic transformation of Populus genotypes<br />

with different chimaeric gene constructs: transformation efficiency and molecular<br />

analysis. Transgen Res 6:111–121<br />

Fladung M, Kaldorf M, Buscot F, Muhs H-J (2000) Untersuchungen zur Stabilität und<br />

Expressivität fremder Gene in Aspenklonen (Populus tremula und P. tremula x P.<br />

tremuloides) unter Freilandbedingungen. In: Schiemann J (ed) Biologische Sicherheitsforschung<br />

bei Freilandversuchen mit transgenen Organismen und anbaubegleitendes<br />

Monitoring. BEO (Projektträger Biologie, Energie, Umwelt des BMBF), Braunschweig–Jülich,<br />

Braunschweig, pp 77–83<br />

Gebhard F, Smalla K (1998) Transformation of Acinetobacter sp. strain BD413 by transgenic<br />

sugar beet DNA. Appl Environ Microbiol 64:1550–1554<br />

Gebhard F, Smalla K (1999) Monitoring field release of genetically modified sugar beets<br />

for persistence of transgenic <strong>plant</strong> DNA and horizontal gene transfer. FEMS Microbiol<br />

Ecol 28:261–272<br />

Gianinazzi-Pearson V, Arnould C, Oufattole M, Arango M, Gianinazzi S (2000) Differential<br />

activation of H + -ATPase genes by an arbuscular mycorrhizal fungus in root cells<br />

of transgenic tobacco. Planta 211:609–613<br />

Glandorf DCM, Bakker PAHM, van Loon LC (1997) Influence of the production of<br />

antibacterial and antifungal proteins by transgenic <strong>plant</strong>s on the saprophytic soil<br />

microflora. Acta Bot Neerl 46:85–104<br />

Griffiths BS, Geoghegan IE, Robertson WM (2000) Testing genetically engineered potato,<br />

producing the lectins GNA and Con A, on non-target soil organisms and processes. J<br />

Appl Ecol 37:159–170<br />

Hampp R, Ecke M, Schaeffer C, Wallenda T, Wingler A, Kottke I, Sundberg B (1996)<br />

Axenic mycorrhization of wild type and transgenic hybrid aspen expressing T-DNA<br />

indoleacetic acid-biosynthetic genes. Tree 11:59–64<br />

Hansen Jesse LC, Obrycki JJ (2000) Field deposition of Bt transgenic corn pollen: lethal<br />

effects on the monarch butterfly. Oecologia 125:241–248<br />

Hetrick BAD, Wilson GWT, Cox TS (1992) Mycorrhizal dependence of modern wheat<br />

varieties, landraces, and ancestors. Can J Bot 70:2032–2040<br />

Heuer H, Smalla K (1999) Bacterial phyllosphere communities of Solanum tuberosum L.<br />

and T4-lysozyme-producing transgenic variants. FEMS Microbiol Ecol 28:357–371<br />

Heuer H, Kroppenstedt RM, Lottmann J, Berg G, Smalla K (2002) Effects of T4 lysozyme<br />

release from transgenic potato roots on bacterial rhizosphere communities are negligible<br />

relative to natural factors. Appl Environ Microbiol 68: 1325–1335


194<br />

Michael Kaldorf et al.<br />

Hoffmann T, Golz C, Schieder O (1994) Foreign DNA sequences are received by a wildtype<br />

strain of Aspergillus niger after co-culture with transgenic higher <strong>plant</strong>s. Curr<br />

Genet 27:70–76<br />

Hu WJ, Harding SA, Lung J, Popko JL, Ralph J, Stokke DD, Tsai CJ, Chiang VL (1999)<br />

Repression of lignin biosynthesis promotes cellulose accumulation and growth in<br />

transgenic trees. Nat Biotechnol 17:808–812<br />

Jimenez-Bermudez S, Redondo-Nevado J, Munoz-Blanco J, Caballero JL, Lopez-Aranda<br />

JM, Valpuesta V, Pliego-Alfaro F, Quesada MA, Mercado JA (2002) Manipulation of<br />

strawberry fruit softening by antisense expression of a pectate lyase gene. Plant Physiol<br />

128:751–759<br />

Kaldorf M, Buscot F, Fladung M, Muhs H-J (2001) Establishment of mycorrhizas on rolCtransgenic<br />

aspen in a field trial. Abstract of the conference “Molekularbiologie der<br />

Pilze”, Jena, 30.9.-3.10.2002, pp 50<br />

Kaldorf M, Fladung M, Muhs H-J, Buscot F (2002) Mycorrhizal colonization of transgenic<br />

aspen in a field trial. Planta 214: 653–660<br />

Kottke I, Oberwinkler F (1986) Mycorrhiza of forest trees – structure and function. Tree<br />

1:1–24<br />

Kottke I, Oberwinkler F (1987) The cellular structure of the Hartig net: coenocytic and<br />

transfer cell-like organization. Nord J Bot 7:85–95<br />

Krishnapillai V (1996) Horizontal gene transfer. J Genet 75:219–232<br />

Losey JE, Rayor LS, Carter ME (1999) Transgenic pollen harms monarch larvae. Nature<br />

399:214<br />

Lottmann J, Berg G (2001) Phenotypic and genotypic characterization of antagonistic<br />

bacteria associated with roots of transgenic and non-transgenic potato <strong>plant</strong>s. Microbiol<br />

Res 156:75–82<br />

Lottmann J, Heuer H, Smalla K, Berg G (1999) Influence of transgenic T4-lysozyme-producing<br />

potato <strong>plant</strong>s on potentially beneficial <strong>plant</strong>-associated bacteria. FEMS Microbiol<br />

Ecol 29:365–377<br />

Lottmann J, Heuer H, de Vries J, Mahn A, Düring K, Wackernagel W, Smalla K, Berg G<br />

(2000) Establishment of introduced antagonistic bacteria in the rhizosphere of transgenic<br />

potatoes and their effect on bacterial community. FEMS Microbiol Ecol<br />

33:41–49<br />

Lukow T, Dunfield PF, Liesack W (2000) Use of the T-RFLP technique to assess spatial<br />

and temporal changes in the bacterial community structure within an agricultural<br />

soil <strong>plant</strong>ed with transgenic and non-transgenic potato <strong>plant</strong>s. FEMS Microbiol Ecol<br />

32:241–247<br />

Masoud SA, Zhu Q, Lamb C, Dixon RA (1996) Constitutive expression of an inducible b-<br />

1,3-glucanase in alfalfa reduces disease severity caused by the oomycete pathogen<br />

Phytophthora megasperma f. sp. medicagini, but does not reduce disease severity of<br />

chitin-containing fungi. Transgen Res 5:313–323<br />

Morra MJ (1994) Assessing the impact of transgenic <strong>plant</strong> products on soil organisms.<br />

Mol Ecol 3:53–55<br />

Mullin TJ, Bertrand S (1998) Environmental release of transgenic trees in Canada –<br />

potential benefits and assesment of biosafety. Forestry Chron 74:203–219<br />

Nehls U, Bock A, Ecke M, Hampp R (2001) Differential expression of the hexose-regulated<br />

fungal genes AmPAL and AmMst1 within Amanita/Populus ectomycorrhizas.<br />

New Phytol 150:583–589<br />

Nielsen KM, Gebhard F, Smalla K, Bones AM, van Elsas JD (1997) Evaluation of possible<br />

horizontal gene transfer from transgenic <strong>plant</strong>s to the soil bacterium Acinetobacter<br />

calcoaceticus BD413. Theor Appl Genet 95:815–821<br />

Nielsen KM, Bones AM, Smalla K, van Elsas JD (1998) Horizontal gene transfer from<br />

transgenic <strong>plant</strong>s to terrestrial bacteria – a rare event? FEMS Microbiol Rev 22:79–103


11 Interactions of Microbes with Genetically Modified Plants 195<br />

Nielsen KM, van Elsas JD, Smalla K (2000) Transformation of Acinetobacter sp. strain<br />

BD413 (pFG4DnptII) with transgenic <strong>plant</strong> DNA in soil microcosms and effects of<br />

kanamycin on selection of transformants. Appl Environ Microbiol 66:1237–1242<br />

Oger P, Mansouri H, Dessaux Y (2000) Effect of crop rotation and soil cover on alteration<br />

of the soil microflora generated by the culture of transgenic <strong>plant</strong>s producing opines.<br />

Mol Ecol 9:881–890<br />

Owen MDK (2000) Current use of transgenic herbicide-resistant soybean and corn in<br />

the USA. Crop Prot 19:765–771<br />

Perlak FJ, Oppenhuizen M, Gustafson K, Voth R, Sivasupraniam S, Heering D, Carey B,<br />

Ihrig RA, Roberts JK (2001) Development and commercial use of Bollgard® cotton in<br />

the USA – early promises versus today’s reality. Plant J 27:489–501<br />

Pilon-Smits EAH, Terry N, Sears T, van Dun K (1999) Enhanced drought resistance in<br />

fructan-producing sugar beet. Plant Physiol Bioch 37:313–317<br />

Punja ZK (2001) Genetic engineering of <strong>plant</strong>s to enhance resistance to fungal<br />

pathogens – a review of progress and future prospects. Can J Plant Pathol 23:216–235<br />

Quiroga GOS, Fraschina AA (1997) Evaluation of sensory attributes and biochemical<br />

parameters in transgenic tomato fruit with reduced polygalacturonase activity. Food<br />

Sci Technol Int 3:93–102<br />

Romer S, Fraser PD, Kiano JW, Shipton CA, Misawa N, Schuch W, Bramley PM (2000) Elevation<br />

of the provitamin A content of transgenic tomato <strong>plant</strong>s. Nat Biotechnol<br />

18:666–669<br />

Saroha MK, Sridhar P, Malik VS (1998) Glyphosate-tolerant crops: genes and enzymes. J<br />

Plant Biochem Biotechnol 7:65–72<br />

Saxena D, Stotzky G (2001) Bacillus thuringiensis (Bt) toxin released from root exudates<br />

and biomass of Bt corn has no apparent effect on earthworms, nematodes, protozoa,<br />

bacteria, and fungi in soil. Soil Biol Biochem 33:1225–1230<br />

Schlüter K, Fütterer J, Potrykus I (1995) “Horizontal” gene transfer from a transgenic<br />

potato line to a bacterial pathogen (Erwinia chrysanthemi) occurs – if at all – at an<br />

extremely low frequency. Biotech 13:1094–1098<br />

Shelton AM, Zhao JZ, Roush RT (2002) Economic, ecological, food safety, and social consequences<br />

of the development of Bt transgenic <strong>plant</strong>s. Annu Rev Entomol 47:845–881<br />

Siciliano SD, Theoret CM, de Freitas JR, Hucl PJ, Germida JJ (1998) Differences in the<br />

microbial communities associated with the roots of different cultivar of canola and<br />

wheat. Can J Microbiol 44:844–851<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis, 2nd edn. Academic Press, San Diego<br />

Solomon-Blackburn RM, Barker H (2001) Breeding virus resistant potatoes (Solanum<br />

tuberosum): a review of traditional and molecular approaches. Heredity 86:17–35<br />

Staehelin C, Charon C, Boller T, Crespi M, Kondorosi A (2001) Medicago truncatula<br />

<strong>plant</strong>s overexpressing the early nodulin gene enod40 exhibit accelerated mycorrhizal<br />

colonization and enhanced formation of arbuscules. Proc Natl Acad Sci USA<br />

98:15366–15371<br />

Straubinger B, Straubinger E, Wirsel S, Turgeon G,Yoder O (1992) Versatile fungal transformation<br />

vectors carrying the selectable bar gene of Streptomyces hygroscopicus.<br />

Fungal Genet Newslet 39:82–83<br />

Thelen JJ, Ohlrogge JB (2002) Metabolic engineering of fatty acid biosynthesis in <strong>plant</strong>s.<br />

Metab Eng 4:12–21<br />

Tuominen H, Sitbon F, Jacobsson C, Sandberg G, Olsson O, Sundberg B (1995) Altered<br />

growth and wood characteristics in transgenic hybrid aspen expressing Agrobacterium<br />

tumefaciens T-DNA indoleacetic acid-biosynthetic genes. Plant Physiol<br />

109:1179–1189<br />

Vance CP, Graham PH (1995) Nitrogen fixation in agriculture: application and perspectives.<br />

In: Tikhonovich IA, Provorov NA, Romanov VI, Newton WE (eds) Nitrogen fixation:<br />

fundamentals and applications. Kluwer, St. Petersburg, pp 77–86


196<br />

Michael Kaldorf et al.<br />

van Rhijn P, Fujishige NA, Lim PO, Hirsch AM (2001) Sugar-binding activity of pea lectin<br />

enhances heterologous infection of transgenic alfalfa <strong>plant</strong>s by Rhizobium leguminosarum<br />

biovar viciae. Plant Physiol 126:133–144<br />

Vierheilig H,Alt M, Neuhaus J-M, Boller T,Wiemken A (1993) Colonization of transgenic<br />

Nicotiana sylvestris <strong>plant</strong>s, expressing different forms of Nicotiana tabacum chitinase,<br />

by the root pathogen Rhizoctonia solani and by the mycorrhizal symbiont Glomus<br />

mosseae. Mol Plant-Microbe Int 6:261–264<br />

Vierheilig H, Alt M, Lange J, Gut-Rella M, Wiemken A, Boller T (1995) Colonization of<br />

transgenic tobacco constitutively expressing pathogenesis-related proteins by the<br />

vesicular arbuscular mycorrhizal fungus Glomus mosseae. Appl Environ Microbiol<br />

61:3031–3034<br />

Warwick SI, Beckie HJ, Small E (1999) Transgenic crops: new weed problems for Canada?<br />

Phytoprotection 80:71–84<br />

White TJ, Bruns T, Lee S, Taylor J (1990) Amplification and direct sequencing of fungal<br />

ribosomal RNA genes for phylogenetics. In: Innis MA, Gelfand DH, Sninsky JJ, White<br />

TJ (eds) PCR protocols: a guide to methods and applications. Academic Press, San<br />

Diego, pp 315–322<br />

Wolfenbarger LL, Phifer PR (2000) The ecological risks and benefits of genetically engineered<br />

<strong>plant</strong>s. Science 290:2088–2093<br />

Wöstemeyer J, Wöstemeyer A, Voigt K (1997) Horizontal gene transfer in the rhizosphere:<br />

a curiosity or a driving force in evolution? Adv Bot Res 24:399–429<br />

Ye X, Al-Babili S, Klöti A, Zhang J, Lucca P, Beyer P, Potrykus I (2000) Engineering the<br />

provitamin A (b-carotene) biosynthetic pathway into (carotenoid-free) rice endosperm.<br />

Science 287:303–305<br />

Zhang HX, Blumwald E (2001) Transgenic salt-tolerant tomato <strong>plant</strong>s accumulate salt in<br />

foliage but not in fruit. Nat Biotechnol 19:765–768


12 Interaction Between Soil Bacteria and<br />

Ectomycorrhiza-Forming Fungi<br />

Rüdiger Hampp and Andreas Maier<br />

1 Introduction<br />

Roots constitute important <strong>plant</strong> organs for water and nutrient uptake. However,<br />

they also release a wide range of carbon compounds of low molecular<br />

weight which are called exudates. These compounds form the basis for an<br />

environment rich in diversified microbiological populations, the rhizosphere<br />

(Hiltner 1904; the rhizosphere has been defined as a narrow zone of soil which<br />

is influenced by living roots). Bacteria are an important part of these populations.<br />

In addition, roots of most terrestrial <strong>plant</strong>s develop symbiotic structures<br />

(mycorrhiza) with soil-borne fungi. In these interactions, the fungal<br />

partner provides the <strong>plant</strong> with improved access to water and nutrients in the<br />

soil due to more or less complex hyphal structures that emanate from the root<br />

<strong>surface</strong> and extend far into the soil. The <strong>plant</strong>, in return, supplies carbohydrates<br />

for fungal growth and maintenance (Smith and Read 1997; Hampp and<br />

Schaeffer 1998). Due to leakage and the turnover of mycorrhizal structures,<br />

these solutes are also released into the rhizosphere where they can be accessed<br />

by other microorganisms. The term “rhizosphere” has, therefore been<br />

extended to “mycorrhizosphere” (Oswald and Ferchau 1968). In the latter, two<br />

different zones can be distinguished: the <strong>surface</strong> of the mycorrhizal structure,<br />

affected by both root and fungus, and that occupied by fungal hyphae only.<br />

The latter has been termed “hyphosphere” (Marschner 1995). Soil free of <strong>plant</strong><br />

and fungal components has been referred to as “bulk soil” (Andrade et al.<br />

1997). It is reasonable to believe that these different spheres may differ in their<br />

microbial activities, and it has been shown that microbial communities within<br />

the rhizosphere are distinct from those of nonrhizosphere soil (Curl and Truelove<br />

1986; Whipps and Lynch 1986).<br />

Interactions between soil bacteria and symbiotic fungi can be both negative<br />

and positive. Mycorrhiza-forming fungi have been shown to reduce bacterial<br />

viability (Meyer and Linderman 1986). Due to the transfer and exudation<br />

of <strong>plant</strong>-derived organic compounds to soil microsites not accessible to<br />

roots, fungi can promote bacterial growth and survival (Hobbie 1992; Söder-<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


198<br />

Rüdiger Hampp and Andreas Maier<br />

ström 1992; Frey-Klett et al. 1997). Furthermore, there is evidence that soil<br />

bacteria can also enhance the formation of mycorrhizal structures, either by<br />

promoting growth (helper bacteria; Garbaye 1994) or by protecting them<br />

from pathogenic micro-organisms.<br />

2 Bacteria<br />

Free-living soil bacteria which are beneficial for <strong>plant</strong> growth are named<br />

<strong>plant</strong>-growth-promoting rhizobacteria (PGPR; Kloepper et al. 1989). These<br />

include species and strains which belong to the genera Azotobacter, Pseudomonas,<br />

Burkholderia, Acetobacter, Herbaspirillum and Bacillus (Glick 1995;<br />

Probanza et al. 1996; see also Barea, Chap. 20, this Vol.).<br />

In contrast to agricultural soils where bacteria dominate, fungi constitute<br />

the major fraction of the microbial flora of forest soils especially in acidic,<br />

organic soils under cold climates (Söderström et al.1983; Nohrstedt et al.1989).<br />

3 Bacterial Community Structure<br />

Abundance and micro-stratification of bacteria and fungi inhabiting the<br />

organic layers of a Scots pine forest were analyzed by Berg et al. (1998). They<br />

counted approx. 5x10 9 bacteria/g dry wt. of organic matter. The mean bacterial<br />

biomass was between 0.34 and 0.25 mg C/g dry wt. of organic matter. This<br />

compared to a fungal biomass of between 0.05 and 0.009 mg C /g dry wt..<br />

Abundance of bacteria and fungi is influenced by the soil water content,<br />

and clear seasonal patterns with a peak of microbial biomass in winter were<br />

reported. The ratio of carbon due to bacteria biomass/fungal biomass was 2:1<br />

in fresh litter and 28:1 in humus. This is in contrast to reports which give evidence<br />

that in acid forest soils the fungal biomass exceeds that of bacteria<br />

(Söderström et al. 1983; Nohrstedt et al. 1989; see also citations in Berg et al.<br />

1998). The ratio may, however, be altered by increasing N input (Verhoef and<br />

Brussaard 1990), which may interfere with existing soil food webs (Moore and<br />

Hunt 1988).<br />

Generally, bacterial densities in forest soils are an order of magnitude<br />

lower than those determined from nursery peat (Timonen et al. 1998). In forest<br />

humus, the common soil species, Pseudomonas fluorescens, a potential<br />

mycorrhiza helper bacterium in pot cultures, could not be identified in the<br />

acidic environment. In contrast, spore-forming bacteria such as Bacillus ssp.,<br />

which are also classified as helper bacteria (Garbaye and Duponnois 1992),<br />

could be identified in mycorrhizospheres of pine.<br />

Following colonization with ectomycorrhiza (ECM)-forming fungi,changes<br />

in root exudates result in greater numbers of microbes in the rhizosphere and<br />

a change in the species types found (Oswald and Ferchau 1968; Malajczuk 1979;


12 Interaction Between Soil Bacteria and Ectomycorrhiza-Forming Fungi 199<br />

Linderman 1988).Different ectomycorrhizospheres indicate an overall bacterial-enrichment<br />

gradient from bulk soil to rhizosphere to mycorrhizosphere<br />

(Frey et al. 1997).<br />

Active exudation of readily usable carbon-rich substrates into the mycorrhizosphere<br />

results in enhanced catabolic community development in natural<br />

lignin-rich forest humus (Heinonsalo et al. 2000). The driving force for community<br />

development/diversity is obviously the continuous supply of carbohydrates<br />

from the host <strong>plant</strong>.A substrate-utilization analysis showed that simple<br />

carbohydrates are readily used by all inner and outer mycorrhizosphere bacteria<br />

(Timonen et al. 1998). Mannitol, an important intermediate storage form<br />

of carbohydrate in many fungi, was preferred by bacterial populations from<br />

all types of mycorrhizospheres. Bacteria from bulk soil, in contrast, show a<br />

preference for organic and amino acids (Timonen et al. 1998).<br />

Bacterial communities from mycorrhizospheres of Pinus sylvestris are<br />

characterized by a preferential utilization of carbohydrates and organic and<br />

amino acids (Frey et al. 1997; Timonen et al. 1998; Frey-Klett et al. 2000). Bacteria<br />

associated with Suillus bovinus ectomycorrhiza favored mannitol, while<br />

those co-occurring with Paxillus involutus preferred fructose as carbon<br />

source. Additional carbon sources used by the bacteria (trehalose, glycogen,<br />

mannitol, N-acetyl-D-glucosamine; Heinonsalo et al. 2000) suggest a limited<br />

saprophytic turnover in acidic forest soils.<br />

Rhizosphere bacteria can also make use of contaminating hydrocarbons as<br />

shown by a decrease in nonpolar hydrocarbons in the mycorrhizosphere<br />

(Heinonsalo et al. 2000).<br />

4 Association of Bacteria with Fungal/Ectomycorrhizal<br />

Structures<br />

Symbiotic interactions between roots and soil fungi comprise different types,<br />

the most important ones being endo- and ectomycorrhizas.Endomycorrhiza is<br />

the most abundant form in soils of most ecosystems. Typical for this mycorrhiza<br />

is the presence in roots of a series of structures which facilitate solute<br />

exchange between the partners of symbiosis. These comprise arbuscules, vesicles,<br />

coiled hyphae etc. (Smith and Read 1997). Ectomycorrhizas, the focus of<br />

this chapter, are mainly formed with roots of forest trees belonging to temperate<br />

and boreal regions.They are characterized by defined morphological structures.<br />

Extraradical mycelia which exploit the soil, form a mantle structure<br />

around fine roots of their host <strong>plant</strong>. From there hyphae emanate into the cell<br />

wall of cortex cells, forming a large <strong>surface</strong> area (Hartig net) which facilitates<br />

solute exchange (Smith and Read 1997; see also Kottke Chap. 13, this Vol.).<br />

In contrast to endomycorrhiza-forming fungi (compare Barea, Chap. 20,<br />

this Vol.), information about the interaction between bacteria and ectomycorrhiza-forming<br />

fungi is still rather limited.


200<br />

Rüdiger Hampp and Andreas Maier<br />

Pioneering work in this field has been carried out by Garbaye (for a review<br />

see Garbaye 1994). Experiments carried out with Picea abies, Pinus nigra,<br />

Pinus sylvestris, Pseudotsuga menziesii, and Quercus robur (Garbaye et al.<br />

1992) indicated that soil bacteria can stimulate the inoculation of roots with<br />

ectomycorrhiza-forming fungi, thereby also reducing the adverse effect of<br />

pathogens. Both effects resulted in a better seedling growth, and thus the term<br />

“helper bacteria” was coined. For more recent work, see Dunstan et al. (1998)<br />

and Poole et al. (2001).<br />

5 Bacteria Associated with Sporocarps and Ectomycorrhiza<br />

Twenty seven bacterial species were isolated from both the sporocarps of Suillus<br />

grevillei and the ECMs of S. grevillei/Larix decidua (Varese et al. 1996). The<br />

genera Pseudomonas, Bacillus, and Streptomyces were predominant. From<br />

sporocarps of white truffles (Tuber sp.), bacterial strains such as Micrococcus,<br />

Moraxella, Staphylococcus and Pseudomonas could be isolated (Citterio et al.<br />

1995). Gram-positive bacteria seldom stimulated in vitro fungal growth.<br />

Among gram-negative bacteria, Pseudomonas strains enhanced growth.<br />

Streptomyces significantly inhibited the fungus. Bacterial supernatants were<br />

not effective.Volatiles enhanced fungal growth to some extent, but not significantly.<br />

Most of the bacteria isolated produced siderophores.<br />

A distinction between the different structures of ECM showed that bacteria<br />

primarily occurred on the <strong>surface</strong> of the mantle and in the interhyphal spaces<br />

(Schelkle et al.1996),but also deep within the mantle (Foster and Marks 1967).<br />

Bacteria of subclasses of proteobacteria (containing <strong>plant</strong>-growth-promoting<br />

rhizobacteria such as Burkholderia, Azospirillum, Acetobacter and Herbaspirillum)<br />

were detected in high numbers on mantle <strong>surface</strong>s (Mogge et al.<br />

2000). The two most common fungi on beech, Lactarius vellereus and Lactarius<br />

subdulcis, were associated with members of the a- and b-subclasses of the<br />

proteobacteria. These bacteria have been shown to be abundant in winter and<br />

early spring (Timonen et al. 1998).<br />

Electron microscopy of ECM with Pinus sylvestris and S. bovinus and Paxillus<br />

involutus (Nurmiaho-Lassila et al. 1997) also revealed bacteria on the<br />

mantle <strong>surface</strong> and at inter- and intracellular locations in the mantle and the<br />

Hartig net (S. bovinus). Fungal strands were colonized only by a few bacteria,<br />

while the outermost external fine hyphae had extensive monolayers of bacteria<br />

attached.<br />

ECM with P. involutus were mostly devoid of bacteria, while the external<br />

mycelium supported bacteria (Nurmiaho-Lassila et al.1997).From their observations,<br />

the authors conclude that single ECM fungi create defined mycorrhizosphere<br />

habitats with distinct populations of bacteria. Knowing that several<br />

different types of ECM can be formed on the same root in close vicinity,a large<br />

local biodiversity of ECM-specific bacterial populations could be postulated.


12 Interaction Between Soil Bacteria and Ectomycorrhiza-Forming Fungi 201<br />

Intracellular bacteria as detected in certain endomycorrhiza-forming fungi<br />

(Bianciotto et al. 1996) are also described for P. sylvestris/Paxillus involutus<br />

mycorrhizospheres; Nurmiaho-Lassila et al. (1997) identified Burkholderia<br />

cepacia in extracts from the respective mycorrhizas. Intracellular bacteria<br />

were also detected in the mycelium of the ectomycorrhizal fungus Laccaria<br />

bicolor S238 N (Bertaux, Frey-Klett, Hartmann, Schmidt, Garbaye, pers.<br />

comm.).<br />

6 Benefits from Bacteria/Ectomycorrhiza Interactions<br />

Bacteria are producers of antibiotics. Newer studies show that a variety of<br />

genera, species and strains of these bacteria (e.g., Bacillus subtilis, Pseudomonas<br />

fluorescens) can inhibit the growth of pathogenic fungi (Fusarium<br />

oxysporum; Cylindrocarpon sp.) in co-culture with ECM fungi such as Laccaria<br />

bicolor, L. proxima and Suillus granulatus (Schelkle and Peterson 1996).<br />

They can, however, also affect ECM fungi. Burkholderia cepacia significantly<br />

reduced the in vitro growth of mycelia of Paxillus involutus. B. cepacia,<br />

Pseudomonas chlororaphis, Ps. fluorescens, and P. involutus reduced the<br />

mycelial growth of the root pathogens Fusarium moniliforme, F. oxysporum,<br />

and Rhizoctonia solani (Pedersen et al. 1999). Burkholderia cepacia also<br />

reduced the formation of ECM short roots by P. involutus on lodgepole pine<br />

and white spruce seedlings in the short term (2 months), but not upon longer<br />

incubation (4 months). Pseudomonas chlororaphis and Ps. fluorescens did not<br />

reduce mycelial growth and mycorrhiza formation. Treatment of the seedlings<br />

with either B. cepacia or P. involutus increased their survival in the presence of<br />

some of the root pathogens investigated. From the data given by Pedersen et<br />

al. (1999) it can thus be concluded that the simplest protective system exists<br />

when bacteria do not inhibit fungal growth/mycorrhiza formation, but affect<br />

potential root pathogens (see also Frey-Klett et al. 2000). There are obviously<br />

also synergistic effects between these bacteria and ECM fungi such as L. proxima<br />

in inhibiting pathogens (Schelkle and Peterson 1996).<br />

In addition to preventing pathogen attacks, bacteria can also support ECM<br />

development directly. This has been shown for different host/fungus combinations<br />

(Garbaye 1994; Frey-Klett et al. 1997). In general, the effect ascribed to<br />

the presence of bacteria consists of a significantly increased number of<br />

infected root tips (Dunstan et al. 1998; Poole et al. 2001).<br />

This should also have an impact on the respective host <strong>plant</strong>. Probanza et<br />

al. (2001) investigated the effect of a co-cultivation with P. tinctorius and<br />

PGPR belonging to the genus Bacillus in enhancing growth of Pinus pinea.<br />

Although the bacterial strains promoted seedling growth, this effect could not<br />

be related to a synergistic interaction with the fungus. A stimulation of shoot<br />

and root biomass production was also observed for Acacia holoserica<br />

seedlings, mycorrhizal with Pisolithus alba and after co-cultivation with two


202<br />

Rüdiger Hampp and Andreas Maier<br />

fluorescent pseudomonad strains (Founoune et al. 2002). After 3 months of<br />

co-culture, the bacterial inoculants disappeared, showing how difficult such<br />

experiments are to interpret. Obviously, the amount of inoculum supplied can<br />

also play an important role (Frey-Klett et al. 1999).<br />

7 Possible Mechanisms of Interaction<br />

As pointed out by Schelkle and Peterson (1996), and in addition to the production<br />

of antibiotics, protective or “helper” effects could be due to competition<br />

for nutrients in the rhizosphere. The formation of siderophores, for<br />

example could be such a mechanism. Siderophores are iron chelators which<br />

make iron available for uptake by the bacteria. As these compounds are<br />

species-specific, Fe-chelates can only be taken up by those bacteria that are<br />

able to produce them. Protective bacteria synthesizing siderophores could<br />

thus out-compete pathogens with regard to Fe (Neidhardt et al. 1990). Similar<br />

mechanisms are known for ECM fungi (Watteau and Berthelin 1990).<br />

Siderophore release from bacteria into the mycorrhizosphere could also<br />

improve absorption of Fe by the mycorrhizal fungus.<br />

Protection from pathogens could, however, also be a mass effect, simply<br />

due to the large number of nonpathogenic bacteria that accumulate in the rhizosphere<br />

due to the high nutrient supply. However, as outlined by Garbaye<br />

(1994), there can be many more mechanisms, such as an improved receptivity<br />

of the root for mycorrhizal infection, a modification of the rhizospheric soil,<br />

improvement of the root-fungus recognition, stimulation of germination of<br />

fungal propagules, as well as an enhancement of fungal growth in the rhizosphere<br />

(see also Brule et al. 2001) which would increase the probability of contact<br />

between fungus and root (compare Dunstan et al. 1998).<br />

In nutrient-poor acidic forest soils modification by micro-organisms<br />

should be an important factor; the C-rich environment provided by the <strong>plant</strong><br />

is attractive for soil micro-organisms, leading to the formation of functionally<br />

compatible microbial communities. These are jointly able to co-mobilize soil<br />

nutrients such as P and N in and around the vegetative mycelium. In addition,<br />

N-fixing bacterial species including Bacillus spp. are possibly present in the<br />

mycorrhizosphere of forest trees (Li et al. 1992) as the vegetative mycelium<br />

represents a niche that is ideally suited for the selection and enrichment of<br />

associative N-fixing bacteria (Sen 2000).<br />

In many of the possible mechanisms, phytohormones such as IAA could<br />

play an important role. A study on the rooting of derooted shoot hypocotyls<br />

of spruce showed that Laccaria bicolor and Pseudomonas fluorescens BBc6<br />

(MHB) both increased the number of roots formed per rooted hypocotyl<br />

(Karabaghli et al. 1998). The same effect was caused by the addition of IAA<br />

alone (control). Both organisms produced IAA in pure culture.


12 Interaction Between Soil Bacteria and Ectomycorrhiza-Forming Fungi 203<br />

8 Biochemical Evidence for Interaction<br />

Streptomycetes are widely distributed saprobic soil bacteria which produce a<br />

wide range of compounds affecting other organisms. Becker et al. (1999) studied<br />

mycorrhiza-associated Streptomyces strains with regard to their effect on<br />

the protein pattern of ECM-forming Laccaria bicolor and Cenococcum<br />

geophilum, and on two pathogenic fungi (Armillaria ostoyae and A. gallica).<br />

One of the strains improved the growth of ECM fungi while inhibiting that of<br />

the pathogens. The effects could be related to differences in fungal gene<br />

expression (mRNA) and the protein profile obtained after in vitro translation;<br />

new proteins were induced by the strain supporting the growth of ECM-fungi,<br />

while the Streptomyces strain leading to adverse effects caused the disappearance<br />

of bands.<br />

New techniques allow for the annotation of such protein spots. Only this<br />

way is it possible to obtain information about the function of the respective<br />

protein. In the following, we give an example for such an approach for<br />

Amanita muscaria.<br />

A. muscaria is a fungus which develops ECM with a wide range of forest<br />

trees. Grown in dual culture with bacterial isolates obtained from soil samples<br />

in close vicinity to spruce roots, this fungus exhibited distinct changes in protein<br />

pattern. Most effective were isolates which were members of the Actinomycetes.<br />

After 10 weeks of dual inoculation of A. muscaria with a respective soil bacterium<br />

in Petri dishes, the hyphae of the fungal mycelium changed their phenotype<br />

in comparison to controls. The hyphal diameter decreased, while cell<br />

length and the extent of hyphal branching increased. To investigate the molecular<br />

mechanisms behind these morphological changes, the proteome of A.<br />

muscaria was screened for differentially expressed polypeptides (two-dimensional<br />

SDS-PAGE electrophoresis). In Fig. 1, the protein patterns for mycelium<br />

from A. muscaria in pure culture (A) and after dual culture with the bacterium<br />

(B) are compared. The pattern reveals about 100 well-separated protein<br />

spots of which about 20 polypeptides were recognized as differentially<br />

expressed. Twelve spots were excised from the gels for sequence analysis by<br />

MALDI-TOF (matrix-assisted laser desorption/ionization time of flight) mass<br />

spectrometry.<br />

Reliable matches to known protein sequences with the peptide mass fingerprints<br />

were obtained for 7 of 12 selected spots.As an example, Table 1 gives<br />

the peptide masses obtained from protein spot no. 78. They show identity with<br />

several predicted peptide masses of actin 1 from the saprophytic fungus<br />

Schizophyllum commune and for actin 2 from the ectomycorrhizal fungus<br />

Suillus bovinus (Tarkka et al. 2000).<br />

Actins are highly conserved cytoskeletal proteins that are present in all<br />

eucaryotic cells. They are probably involved in various processes such as cytoplasmic<br />

streaming, cell shape determination, tip growth, cell wall deposition,


204<br />

Rüdiger Hampp and Andreas Maier<br />

Fig. 1. Two-dimensional maps of mycelial proteins from Amanita muscaria. Protein<br />

(300 mg) was loaded onto IEF gels. A A. muscaria pure culture; B dual culture of A. muscaria<br />

with a soil bacterium. Differentially expressed proteins are indicated in A (open<br />

circles). Downregulation (closed inverted triangles) or upregulation (closed triangles) of<br />

the analogous spots is indicated in B. (Gels were stained with SYPRO Ruby fluorescent<br />

dye; molecular probes, Eugene, OR, USA.). Results obtained from the computer-aided<br />

evaluation were rigorously compared by visual analysis of the original gels. Stained protein<br />

spots were excised and digested in-gel with modified trypsin (Promega) according<br />

to Williams and Stone (1997)<br />

etc. (Sheterline et al. 1992). At first sight, it looks surprising that a protein<br />

which is important in cell shape determination is downregulated when<br />

hyphal morphology changes. However, the decrease in the amount of actin<br />

coincides with the decrease of the fungal diameter. Interestingly, the amount<br />

of actin protein increases during fruiting body formation of A. muscaria.The<br />

hyphae of the fruiting body are swollen and branched (Manachére et al. 1983)<br />

and the results thus indicate differential regulation of actin during changes in<br />

A. muscaria hyphal growth pattern.<br />

These results emphasize the usefulness of proteome analysis in identifying<br />

molecular events occurring in fungus bacteria interactions.


Table 1. Protein features and data from the peptide mass fingerprint of the protein in spot no. 78. Comparison with the<br />

computer-generated peptides from Schizophyllum commune and Suillus bovinus indicates identity with fungal actins. Mass<br />

spectra of peptide mixtures were obtained by MALDI-TOF (matrix-assisted laser desorption/ionization time of flight) mass<br />

spectrometer (Dr. C. Niehaus, University of Bielefeld, Germany). The database search using the proteolytic peptide masses<br />

was performed with the Mascot program developed by Perkins et al. (1999)<br />

12 Interaction Between Soil Bacteria and Ectomycorrhiza-Forming Fungi 205<br />

Protein identity p78 Actin 1 Actin 2<br />

Organism Amanita muscaria Schizophyllum commune Suillus bovinus<br />

MW (Da) Approx. 42,000 41,876 41,979<br />

pI 5.35 5.30 5.31<br />

Observed peptides Calculated peptides Calculated peptides<br />

MW (Da) MW (Da)/sequence MW (Da)/sequence<br />

780.59 780.45/IVAPPER 780.45/IVAPPER<br />

908.69 908.54/IVAPPERK 908.54/IVAPPERK<br />

1141.69 1141.54/GYPFTTTAER -<br />

1485.89 1485.68/QEYDESGPGIVHR 1485.68/QEYDESGPGIVHR<br />

1589.19 1588.88/LDLAGRDLTDFLIK 1589.81/DLTDCLIKNLTER<br />

1790.19 1789.88/SYELPDGQVITIGNER 1789.88/SYELPDGQVITIGNER


206<br />

Rüdiger Hampp and Andreas Maier<br />

9 Impacts of Environmental Pollution<br />

Microcosms with S. bovinus, P. involutus/Pinus sylvestris in forest humus<br />

amended with petroleum hydrocarbon were investigated with regard to fungus/bacteria<br />

responses (Sarand et al. 1998). Hyphae emanating from mycorrhizas<br />

formed patches around contaminations with a microbial biofilm at the<br />

hydrocarbon/fungus interface. Bacteria consisted of isolates of Ps. fluorescens.<br />

This opens possibilities for the bioremediation of environmental pollutants<br />

by the use of degradative micro-organisms. Sarand et al. (1999) tested m-toluate<br />

as a model compound for petrol-contaminated sites. Fungal survival (Suillus<br />

bovinus) on medium containing this compound was increased in co-culture<br />

on agar plates with degradative bacterial strains of Ps. fluorescens. The<br />

activity of the bacterium was not affected in a tripartite system containing S.<br />

bovinus/Pinus sylvestris mycorrhizas. The fungus was not able to degrade mtoluate,<br />

although mycorrhizal fungi are able to produce enzymes capable of<br />

degrading complex organic compounds (see Sarand et al. 1999).<br />

10 Conclusions<br />

Many of the experiments carried out in order to investigate a possible interaction<br />

between bacteria and ECM-forming fungi have been carried out under<br />

sterile conditions or in pot cultures. Experience shows that, when transferred<br />

to field conditions the respective bacteria will not thrive, but will soon be substituted<br />

by other genera, species or strains. Thus, a more promising approach<br />

is to collect bacteria from mycorrhizas obtained from natural sites and introduce<br />

these into laboratory experiments, with dual cultures being the easiest<br />

way to investigate molecular interaction. In our experience, Gram-positive<br />

bacteria such as Actinomycetes, although largely neglected, are abundant at<br />

least in mycorrhizospheres of spruce stands, and are thus important candidates<br />

for future approaches.<br />

Acknowledgements. We gratefully acknowledge critical reading and helpful suggestions<br />

by Dr. Garbaye (INRA, Nancy, France). As far as our own work is concerned, we are<br />

indebted to the Deutsche Forschungsgemeinschaft for financial support (Graduate<br />

School “Infection Biology”)


12 Interaction Between Soil Bacteria and Ectomycorrhiza-Forming Fungi 207<br />

References and Selected Reading<br />

Andrade G, Mihara KL, Linderman RG, Bethlenfalvay GJ (1997) Bacteria from rhizosphere<br />

and hyphosphere soils of different arbuscular-mycorrhizal fungi. Plant Soil<br />

192:71–79<br />

Becker DM, Bagley ST, Podila GK (1999) Effects of mycorrhizal-associated streptomycetes<br />

on growth of Laccaria bicolor, Cenococcum geophilum, und Armillaria<br />

species and on gene expression in Laccaria bicolor. Mycologia 91:33–40<br />

Berg MP, Kniese JP,Verhoef HA (1998) Dynamics and stratification of bacteria and fungi<br />

in the organic layers of a Scots pine forest soil. Biol Fertil Soils 26:313–322<br />

Bianciotto V, Bandi C, Minerdi D, Sironi M, Tichy HV, Bonfante P (1996) An obligately<br />

endosymbiotic mycorrhizal fungus itself harbors obligately intracellular bacteria.<br />

Appl Environ Microbiol 62:3005–3010<br />

Brule C, Frey-Klett P, Pierrat JC, Courrier S, Gerard F, Lemoine MC, Rousselet JL, Sommer<br />

J, Garbaye J (2001) Survival in the soil of the ectomycorrhizal fungus Laccaria bicolor<br />

and the effects of a mycorrhiza helper Pseudomonas fluorescens. Soil Biol Biochem<br />

33:1683–1694<br />

Citterio B, Cardoni P, Potenza L, Amicucci A, Atocchi V, Gola G, Nuti M (1995) Isolation<br />

of bacteria from sporocarps of Tuber magnatum pico, tuber borchii vitt. and Tuber<br />

maculatum vitt.: identification and biochemical characterization. In: Stocchi V, Bonfante<br />

P, Nuti M (eds) Biotechnology of ectomycorrhizae. Plenum Press, New-York, pp<br />

241–248<br />

Curl EA, Truelove B (1986) The rhizosphere. Springer, Berlin Heidelberg New York, pp<br />

1–8<br />

Dunstan WA, Malajczuk N, Dell, B (1998) Effects of bacteria on mycorrhizal development<br />

and growth of container-grown Eucalyptus diversicolor F. Muell. seedlings. Plant Soil<br />

201:241–249<br />

Foster RC, Marks GC (1967) The fine structure of the mycorrhizas of Pinus radiata.Aust<br />

J Biol Sci 19:1027–1038<br />

Founoune H, Duponnois R, Ba AM, Sall S, Branget I, Lorquin J, Neyra M, Chote JL (2002)<br />

Mycorrhiza helper bacteria stimulate ectomycorrhizal symbiosis of Acacia holosericea<br />

with Pisolithus alba. New Phytol 153:81–89<br />

Frey-Klett P, Brule C, Garbaye J (1997) A proposed model for the mycorrhiza-helper<br />

effect of Pseudomonas fluorescens BBc6 on the ectomycorrhizal system Laccaria<br />

bicolorS238N-Douglas fir. 4th International Workshop on Plant Growth Promoting<br />

Rhizobacteria, Sapporo, Japan<br />

Frey-Klett P, Churin JL, Pierrat JC, Garbaye J (1999) Dose effect in the dual inoculation of<br />

an ectomycorrhizal fungus and a mycorrhiza helper bacterium in two forest nurseries.<br />

Soil Biol Biochem 31:1555–1562<br />

Frey-Klett P, Chavatte M, Courriers S, Martinotii G, Pierrat JC, Garbaye J (2000) Ectomycorrhizosphere<br />

effect of the Douglas fir-Laccaria bicolor symbiosis on the functional<br />

diversity of fluorescent pseudomonads in a forest nursery. 5th International Workshop<br />

on PGPR, Cordoba, Argentine. www.ag.auburn.edu/argentina/<strong>pdf</strong>manuscripts/<br />

freyklett.<strong>pdf</strong><br />

Frey P, Frey-Klett P, Garbaye J, Berge O, Heulin (1997) Metabolic and genotyping fingerprinting<br />

of fluorescent pseudomonads associated with the Douglas fir-Laccaria<br />

bicolor mycorrhizosphere. Appl Environ Microbiol 63:1852–1860<br />

Frey-Klett P, Pierrat JC, Garbaye J (1997) Location and survival of Mycorrhiza helper<br />

Pseudomonas fluorescens during establishment of ectomycorrhizal symbiosis<br />

between Laccaria bicolor and Douglas Fir. Appl Environ Microbiol 63:139–144<br />

Garbaye J (1994) Helper bacteria: a new dimension to the mycorrhizal symbiosis. New<br />

Phytol 128:197–210


208<br />

Rüdiger Hampp and Andreas Maier<br />

Garbaye J, Churin JL, Duponnois R (1992) Effects of substrate disinfection, fungicide<br />

treatment and mycorrhiza helper bacteria (MHB) on ectomycorrhiza formation of<br />

pedunculate oak inoculated with Laccaria laccata in two bare-root nurseries. Biol<br />

Fertil Soils 13:55–47<br />

Garbaye J, Duponnois R (1992) Specificity and function of mycorrhiza helper bacteria<br />

(MHB) associated with the Pseudotsuga menziesii–Laccaria laccata symbiosis.Symbiosis<br />

14:335–344<br />

Glick BR (1995) The enhancement of <strong>plant</strong> growth by free living bacteria. Can J Microbiol<br />

41:109–117<br />

Hampp R, Schaeffer C (1998) Mycorrhiza – Carbohydrate and energy metabolism. In:<br />

Varma A, Hock B (eds) Mycorrhiza – structure, function, molecular biology and<br />

biotechnology. Springer, Berlin Heidelberg New York, pp 273–303<br />

Heinonsalo J, Jorgensen KS, Haahtela K, Sen R (2000) Effects of Pinus sylvestris root<br />

growth and mycorrhizosphere development on bacterial carbon source utilization<br />

and hydrocarbon oxidation in forest and petroleum-contaminated soils. Can J Microbiol<br />

46:451–464<br />

Hiltner L (1904) Über neuere Erfahrungen und Probleme auf dem gebiet der Bodenbakteriologie<br />

und unter besonderer Berücksichtigung der Gründüngung und Brache.<br />

Arb Dtsch Landwirt Ges 98:59–78<br />

Hobbie SE (1992) Effects of <strong>plant</strong> species on nutrient cycling. Trends Ecol Evol 7:336–339<br />

Karabaghli C, Frey-Klett P, Sotta B, Bonnet M, Le Tacon F (1998) In vitro effects of Laccaria<br />

bicolor S238 N and Pseudomonas fluorescens strain BBc6 on rooting of derooted<br />

shoot hypocotyls of Norway spruce. Tree Physiol 18:103–111<br />

Kloepper JW, Lifshitz R, Zablotowicz RM (1989) Free-living bacterial inocula for<br />

enhancing crop productivity. Trends Biotechnol 7:39–43<br />

Li CY, Massicotte HB, Moore LVH (1992) Nitrogen-fixing Bacillus sp. associated with<br />

Douglas-fir tuberculate ectomycorrhizae. Plant Soil 140:35–40<br />

Linderman RG (1988) Mycorrhizal interactions with the rhizosphere microflora: the<br />

mycorrhizosphere effect. Phytopathology 78:366–371<br />

Malajczuk N (1979) The microflora of unsuberized roots of Eucalyptus calophylla R. Br.<br />

and Ecalyptus marginata Donn ex Sm seedlings grown in soils suppressive and conductive<br />

to Phytophthora cinnamomi Rands. II. Mycorrhizal roots and associated<br />

microflora. Aust J Bot 27:235–254<br />

Manachére G, Rober JC, Durand R, Bret JP, Févre M (1983) Differentiation in the basidiomycetes.<br />

In: Smith JE (ed) Mycology series; vol 4. Fungal differentiation: A contemporary<br />

synthesis. Marcel Dekker, New York, pp 481–514<br />

Marschner H (1995) Mineral nutrition of higher <strong>plant</strong>s. Academic Press, London, 571 pp<br />

Meyer JR Linderman RG (1986) Response of subterranean clover to dual inoculation<br />

with vesicular-arbuscular mycorrhizal fungi and a <strong>plant</strong> growth-promoting bacterium,<br />

Pseudomonas putida. Soil Biol Biochem 18:185–190<br />

Mogge B, Loferer C, Agerer R, Hutzler P, Hartmann A (2000) Bacterial community structure<br />

and colonialization patterns of Fagus sylvatica L. ectomycorrhizospheres as<br />

determined by fluorescence in situ hybridization and confocal laser scanning<br />

microscopy. Mycorrhiza 9:271–278<br />

Moore JC, Hunt HW (1988) Resource compartmentation and the stability of real ecosystems.<br />

Nature 333:261–263<br />

Neidhardt FC, Ingraham JL, Schaechter M (eds) (1990) Physiology of the bacterial cell.<br />

Sinauer, MA, USA<br />

Nohrstedt H-Ö,Arnebrandt K, Bååth E, Söderström B (1989) Changes in carbon content,<br />

respiration rate,ATP content, and microbial biomass in nitrogen-fertilized pine forest<br />

soils in Sweden. Can. J For Res 19:323–328


12 Interaction Between Soil Bacteria and Ectomycorrhiza-Forming Fungi 209<br />

Nurmiaho-Lassila E-L, Timonen S, Haahtela K, Sen R (1997) Bacterial colonialization<br />

patterns of intact Pinus sylvestris mycorrhizospheres in dry pine forest soil: an electron<br />

microscopy study. Can J Microbiol 43:1017–1035<br />

Oswald ET, Ferchau HA (1968) Bacterial associations of coniferous mycorrhizae. Plant<br />

Soil 28:187–192<br />

Pedersen EA, Reddy MS, Chakravarty P (1999) Effect of three species of bacteria on<br />

damping-off, root rot development, and ectomycorrhizal colonialization of lodgepole<br />

pine and white seedlings. Eur J For Pathol 29:123–134<br />

Perkins DN, Pappin DJ, Creasy D, Cotrell JS (1999) Probability-based protein identification<br />

by searching sequence databases using mass spectrometry data. Electrophoresis<br />

20:3551–3567<br />

Poole EJ, Bending GD, Whipps JM, Read DJ (2001) Bacteria associated with Pinus<br />

sylvestris-Lactarius rufus ectomycorrhizas and their effects on mycorrhiza formation<br />

in vitro. New Phytol 151:743–751<br />

Probanza A, Lucas JA, Guiterrez Mañero JF (1996) The influence of native rhizobacteria<br />

on European alder (Alnus glutinosa (L.) Gaertn.) growth. I. Characterization of<br />

growth promoting and growth inhibiting bacterial strains. Plant Soil 182:59–66<br />

Probanza A, Mateos JL, Lucas Garcia JA, Ramos B, de Felipe MR, Guiterrez Manero JF<br />

(2001) Effects of inoculation with PGPR Bacillus and Pisolithus tinctorius on Pinus<br />

pinea L. growth, bacterial rhizosphere colonialization, and mycorrhizal infection.<br />

Microb Ecol 41:140–148<br />

Sarand I, Timonen S, Nurmiaho-Lassila E-L, Koivula T, Haahtela K, Romantschuk M, Sen<br />

R (1998) Microbial biofilms and catabolic plasmid harbouring degradative fluorescent<br />

pseudomonads in Scots pine mycorrhizospheres developed on petroleum contaminated<br />

soil. FEMS Microbiol Ecol 27:115–126<br />

Sarand I, Timonen S, Koivula T, Peltola R, Haahtela K, Sen R, Romantschuk M (1999) Tolerance<br />

and biodegradation of m-toluate by Scots pine, a mycorrhizal fungus and fluorescent<br />

pseudomonads individually and under associative conditions. J Appl Microbiol<br />

86:817–826<br />

Schelkle M, Peterson RL (1996) Suppression of common root pathogens by helper bacteria<br />

and ectomycorrhizal fungi in vitro. Mycorrhiza 6:481–485<br />

Schelkle M, Ursic M, Farquhar M, Peterson RL (1996) The use of laser scanning confocal<br />

microscopy to characterize mycorrhizas of Pinus strobus L. and to localize associated<br />

bacteria. Mycorrhiza 6:431–440<br />

Sen R (2000) Budgeting for the wood-wide web. New Phytol 145:161–165<br />

Sheterline P, Handel SE, Molloy C, Hendry KAK (1992) The nature and regulation of<br />

actin filament turnover in cells. Acta Histochem 41:303–309<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis, 2nd edn. Academic Press, Cambridge<br />

Söderström B (1992) Ecological potential of ectomycorrhizal mycelium. In: Read DJ,<br />

Lewis DH, Fitter AH, Alexander IJ (eds) Mycorrhizas in ecosystems. Cambridge University<br />

Press, Cambridge, pp 77–83<br />

Söderström B, Bååth E, Lundgren B (1983) Decrease in soil microbial activity and biomass<br />

owing to nitrogen amendments. Can J Microbiol 29:1500–1506<br />

Tarkka MT, Vasara R, Gorfer M, Raudaskoski M (2000) Molecular characterization of<br />

actin genes from homobasidiomycetes: two different actin genes from Schizophyllum<br />

commune and Suillus bovinus. Gene 251:27–35.<br />

Timonen S, Jorgensen KS, Haahtela K, Sen R (1998) Bacterial community structure and<br />

defined locations of Pinus sylvestris–Paxillus involutus mycorrhizospheres in dry<br />

pine forest humus and nursery peat. Can J Microbiol 44:499–513<br />

Varese GC, Portinario S, Trotta A, Scannerini S, Luppi-Mosca AM, Martinotti MG (1996)<br />

Bacteria associated with Suillus grevillei sporocarps and ectomycorrhizae and their<br />

effects on in vitro growth of the Microbiont. Symbiosis 21:129–147


210<br />

Rüdiger Hampp and Andreas Maier<br />

Verhoef HA, Brussaard L (1990) Decomposition and nitrogen mineralization in natural<br />

and agro-ecosystems: the contribution of soil animals. Biogeochemistry 11:175–211<br />

Watteau F, Berthelin J (1990) Iron solubilization by mycorrhizal fungi producing<br />

siderophores. Symbiosis 9:59–67<br />

Whipps JM, Lynch JM (1986) The influence of rhizosphere on crop productivity. Adv<br />

Microb Ecol 9:187–244<br />

Williams KR, Stone KL (1997) Enzymatic cleavage and HPLC peptide mapping of proteins.<br />

In: Walker J (ed) Molecular biotechnology. Humana Press, Totowa, pp 155–167


13 The Surface of Ectomycorrhizal Roots and the<br />

Interaction with Ectomycorrhizal Fungi<br />

Ingrid Kottke<br />

1 Introduction<br />

Most of the trees in the temperate and alpine regions live in symbiosis with<br />

root fungi forming ectomycorrhizas. Ectomycorrhizas (ECM) display a very<br />

specified cellular organization.A fungal sheath covers the root <strong>surface</strong> and the<br />

hyphae invade intercellularly between the root cortical cells establishing the<br />

so-called Hartig net. The hyphal sheath is formed in a species-specific manner<br />

(Agerer 1998), but the architecture of the Hartig net is similar in all the<br />

ectomycorrhizas, independent of <strong>plant</strong> and fungal species (Blasius et al. 1986,<br />

Kottke and Oberwinkler 1987, 1989). Establishing the Hartig net, hyphal<br />

growth undergoes important changes. The hyphae invade as multi-branched,<br />

fan-like lobes in intimate juxtaposition, starting at the root <strong>surface</strong> and finally<br />

covering the root cortical cells in a dense mono-layer (Fig. 1; Jacobs et al. 1989;<br />

Brunner and Scheidegger 1992; Kottke et al. 1996).<br />

The Hartig net structure is only established in so-called short roots, a special<br />

root type of the ectomycorrhiza-forming <strong>plant</strong>s (Marks and Foster 1973;<br />

Wong et al. 1990). It was hypothesized that the <strong>surface</strong> of these rootlets might<br />

trigger the attachment of hyphae and the change of their growth characters<br />

(Jacobs et al. 1989; Brunner and Scheidegger 1992; Kottke 1997; Bonfante et<br />

al. 1998). Cysteine-rich, moderately hydrophobic proteins (“hydrophobins”)<br />

in the walls of the ectomycorrhizal fungus Pisolithus tinctorius (Pers.) Coker<br />

& Couch were shown to be highly expressed in the early stage of mycorrhiza<br />

formation and were considered to attach the hyphae to the root <strong>surface</strong> of<br />

Eucalyptus globulus ssp. bicostata Kirkp. (Tagu et al. 1996, 2000, 2001; Martin<br />

et al. 1999). A hydrophobic root <strong>surface</strong> was, therefore postulated and a cuticle-like<br />

layer on the <strong>surface</strong> of short roots may be the substrate for adhesion<br />

of ectomycorrhizal fungi (Kottke 1997). Recent ultrastructural studies comparing<br />

long and short roots have supported this hypothesis by revealing the<br />

origin of the cuticle-like layer on short, but not on long roots. Differences<br />

were also detected in the amounts of methyl-esterified pectins in the cortical<br />

cell walls of both the root types. Furthermore, when establishing the Har-<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


212<br />

Ingrid Kottke<br />

Fig. 1. Ectomycorrhiza formation by Laccaria amethystea and Picea abies. Longitudinal<br />

section through an early state mycorrhiza. Hyphal attachment to root hairs (arrowhead)<br />

without changes of hyphal morphology; hyphal attachment to root <strong>surface</strong> followed by<br />

hyphal enlargement (arrow) and lobe-like growth of hyphae (double arrow); typical<br />

Hartig net structure established between root cortical cells down to the endodermis<br />

(scale 15 mm). cc Cortical cell, e endodermis, hs hyphal sheath, Hn Hartig net, rh root hair<br />

tig net, the cuticle-like layer has to be penetrated by the hyphae. This process<br />

has not been shown before and may be considered as a locally restricted<br />

aggressive or saprophytic phase during ECM formation.<br />

2 Long and Short Roots of Ectomycorrhiza-Forming Plants<br />

Ectomycorrhizas are exclusively formed by perennial, woody <strong>plant</strong>s belonging<br />

to Pinaceae or to distinct families within the Rosidae (sensu “Angiosperm<br />

Phylogenetic Group”, Bremer et al. 1998). The root system of these ECMforming<br />

<strong>plant</strong>s is divided into main, or “long roots” of relatively fast and<br />

unlimited growth and secondary “short roots” of slow and limited growth<br />

(Noelle 1910; Clowes 1951; Marks and Foster 1973). Ectomycorrhizae are typically<br />

formed on short roots. However, long roots may become mycorrhizal<br />

after turning into a resting stage (Wilcox 1968b). It was speculated that the<br />

growth rate of hyphae might not compete with the growth rate of long roots,<br />

thus preventing mycorrhiza formation (Marks and Foster 1973). However, sig-


mrc<br />

rc<br />

rc<br />

a b<br />

c d<br />

13 Root Surface in Ectomycorrhizas 213<br />

met<br />

Fig. 2a–d. Root cap formation of several Pinaceae. a Long root of Picea abies with nonsuberized,<br />

decaying root cap cells; b long root of Larix decidua at resting stage with<br />

metacutization of root cap; c short root of Pinus sylvestris with suberized root cap cells<br />

containing phenols; d root budding in Picea abies from below a hyphal sheath, bud covered<br />

by suberized root cap cells (scale 15 mm). hs Hyphal sheath, met metacutization, mrc<br />

moribund root cap cell, rc root cap<br />

rc<br />

rc<br />

hs


214<br />

Ingrid Kottke<br />

nificant structural differences exist between the two root types and may be<br />

even more important for mycorrhiza formation or failure.<br />

Fast growing long root tips are covered by a conspicuous root cap consisting<br />

of non-suberized, rapidly decaying cells (Fig. 2a). Dormant long roots and<br />

short roots have in common that the root cap cells are few and become suberized<br />

(Fig. 2b, c). This type of root cap cells is also found in root buds even<br />

when emerging from below a hyphal sheath (Fig. 2d). The process was termed<br />

metacutization (“Metakutinisierung” Müller 1906) and was found to occur in<br />

a multitude of gymnosperm and angiosperm perennial species irrespectively<br />

of the epidermal or cortical cell type on the root <strong>surface</strong> of these two taxonomic<br />

groups (Plaut 1918). It was described in detail from light microscope<br />

studies of Fagus sylvatica L. (Clowes 1954), Betula alleghaniensis Britt., Alnus<br />

crispa (Ait.) Pursh, Eucalyptus pilularis Smith (Massicotte et al. 1986,<br />

1987a,b), Abies procera Rehder (Wilcox 1954), Picea abies [L.] Karst. (Kottke et<br />

al. 1986) and Pinus spp. (Hatch and Doak 1933).<br />

3 A Cuticle-Like Layer on the Surface of Short Roots<br />

Ultrastructural investigations yielded further details on the fate of the suberized<br />

root cap cells of short roots. Young root cap cells become suberized by a<br />

lamellar layer imposed on the inner side of the cell walls (Figs. 3a, b, 4a). As<br />

suberin is only weakly stained by osmium and lead, a suberin layer appears<br />

electron-translucent (Sitte 1975; Kottke and Oberwinkler 1990). Lamellae are<br />

visible in the suberin layer if waxes are present additionally (Fig. 4a; Sitte<br />

1975). The suberin layer progressively increases with ageing of the cells.<br />

Finally, these cells accumulate phenolic substances, become impermeable and<br />

moribund (Fig. 3a, b). Short roots proliferate slowly under the root cap cells<br />

(Fig. 2 c) and remain covered by their residues (Fig. 4b, c). The dead root cap<br />

cells progressively detach from the root (Figs. 3b, 4b, c), but the innermost,<br />

suberized root cap cell walls remain tightly connected to the root cortical cell<br />

layer (Figs. 3 c, 4b, c). Thus, the suberin layer of the innermost root cap cells<br />

covers the whole <strong>surface</strong> of short roots, similar to a fine cuticle. During the<br />

elongation of the root the suberin layer is thinned out (Fig. 4e). It fades away<br />

on the root hairs (Fig. 3d) covering only the root hair base (Fig. 4d). At the<br />

most proximal parts of the rootlets, the suberin layer may also fade away on<br />

the <strong>surface</strong> of cortical cells (Fig. 5a, c), but the cell junctions remain tightly<br />

covered by the suberin layer all along the rootlet (Fig. 5a, b). The whole situation<br />

is illustrated by a scheme (Fig. 6).<br />

The suberin layer is covered by a thin layer of electron-dense material. Phenols<br />

are strongly stained by osmium and lead and thus appear electron-dense.<br />

It is not always easy to discern if this material originates from insoluble phenolic<br />

residues of the former vacuole or from cell walls of the deceased root cap<br />

cells as vacuoles and moribund cell walls of root cap cells may contain high


ccw<br />

cc<br />

rccw<br />

cc<br />

rccw<br />

ccw<br />

ph<br />

rccw<br />

rccw<br />

13 Root Surface in Ectomycorrhizas 215<br />

rccw<br />

ph<br />

a b<br />

*<br />

rhcw<br />

c d<br />

Fig. 3. a, b Suberized (arrows) root cap cell layers close to the root apex of Picea abies.<br />

The outer layers detaching (*) and partly decomposed. Cell wall or vacuolar, phenolic<br />

residues form the superficial layer. Scale 0.5 mm. c Superficial layer on short root cortical<br />

cell formed by the suberized root cap cell wall lined by phenolic residues (arrow). Scale<br />

0.3 mm. d Cell wall of root hair with no suberin layer (scale 0.3 mm). cc Cortical cell, ccw<br />

cortical cell wall, ph phenolic residues, rccw root cap cell wall, rhcw root hair cell wall<br />

amounts of phenolic residues (Fig. 2 c). An attempt was undertaken to clarify<br />

the situation by carefully studying the cell layers (Fig. 3a, b). Additional hints<br />

for recognition of cell wall material were obtained by immunogold labelling<br />

(see below). At the final stage of root development, when only the innermost<br />

root cap cell wall and its suberin layer are preserved on the root cortical cell<br />

wall (Fig. 3 c), the thin, electron-dense layer on top of the suberin layer can<br />

only be interpreted as the phenolic residues of the former vacuole. Dehydration<br />

of mycorrhizas in alcohol and embedding in LRWhite resin may obscure<br />

the suberin layer (Kottke 1997; Bonfante et al. 1998), but high pressure cryofixation,<br />

dehydration by acetone and embedding in Araldite/Epon or embed-


216<br />

d<br />

Ingrid Kottke<br />

rcc<br />

ph<br />

rccw ccw<br />

a<br />

rccw<br />

cc<br />

ccw<br />

mrcc<br />

rccw<br />

cc<br />

rh cc<br />

ccw<br />

mrcc<br />

Fig. 4a–e. Cuticle-like layer on <strong>surface</strong> of short roots of Picea abies. a Lamellate structure<br />

of the suberin layer (arrowhead). Scale 0.1 mm. b, c Moribund root cap cells detaching<br />

from root cortical cell, suberized innermost root cap cell wall preserved in tight contact<br />

to cortical cell wall (arrowheads). Scale 0.5 mm. d Suberin layer fading away at root<br />

hair basis (arrowhead). Scale 0.5 mm. e Thinning of suberin layer (arrowhead) during<br />

elongation of cortical cells (scale 0.5 mm). cc Cortical cell, ccw cortical cell wall, ph phenolic<br />

residues, rcc root cap cell, rccw root cap cell wall, rh root hair<br />

ph<br />

e<br />

b<br />

ph


cc<br />

cc<br />

cc<br />

sl<br />

cc<br />

rcc<br />

13 Root Surface in Ectomycorrhizas 217<br />

a b<br />

cc<br />

cc<br />

cc<br />

cc<br />

rcc<br />

c d e<br />

Fig. 5a–e. Suberized root cap cell layer covering cell junctions of short roots, but not of<br />

long roots (Picea abies). a Residues of suberized root cap cells covering the cell junction<br />

at the proximal part of a short root, fading away on the cortical cell (arrow). Scale 3 mm.<br />

b Enlargement of cell junction displaying several suberin layers of moribund root cap<br />

cells. Scale 1 mm. c Enlargement of cortical cell displaying fading away of the suberin<br />

layer (arrow). Scale 1 mm. d No suberin layer on the cortical cell wall of a long root. Scale<br />

1 mm. e No suberin layer on top of the cell junction of a long root (scale 1 mm). cc Cortical<br />

cell, cj cell junction, rcc root cap cell, sl suberin layer<br />

ding in Spurr’s resin after fixation in glutaraldehyde yields clear results. The<br />

electron-dense layer on the root hair <strong>surface</strong> is no longer considered as a cuticle<br />

as was erroneously given in Kottke (1997).<br />

The root cap cells of long roots are not suberized and no cuticle-like layer<br />

exists on the <strong>surface</strong> of long roots and their cell junctions (Fig. 5d, e).<br />

cj<br />

cj<br />

sl


218<br />

Ingrid Kottke<br />

Fig. 6. Scheme illustrating the fate of<br />

the suberized root cap cells of short<br />

roots presumably in most ectomycorrhiza<br />

forming tree species. Figures<br />

refer to given micrographs<br />

4 Involvement of the Cuticle-Like Layer in Mycorrhiza<br />

Formation<br />

The cuticle-like, suberin layer covering short roots, by displaying a hydrophobic<br />

<strong>surface</strong>, appears to be involved in hyphal attachment at the beginning of<br />

ECM development. The suberin layer on the cell junctions is a barrier, however,<br />

that has to be penetrated when the hyphae invade between the root cortical<br />

cells establishing the Hartig net.<br />

5 Involvement of the Cuticle-Like Layer in Hyphal<br />

Attachment<br />

Using scanning electron microscopy, hyphae of P. tinctorius and Paxillus involutus<br />

(Batsch) Fr. attached to rootlets of Quercus acutissima Carruth or Betula<br />

spp., respectively, were found to be embedded in a mucilaginous material<br />

(Massicotte et al. 1987a, b; Brunner and Scheidegger 1992; Oh et al. 1995).<br />

Transmission electron microscopy revealed that adhesion of hyphae to the<br />

root <strong>surface</strong> was aided by polysaccharide fibrils and binding sites of mannose<br />

(Piché et al. 1983a; Thomson et al. 1989; Wong et al. 1990; Lei et al. 1991; Tagu<br />

et al. 2000). Laurent et al. (1999) identified cell-adhesion proteins in the cell


13 Root Surface in Ectomycorrhizas 219<br />

a b<br />

cc<br />

sl<br />

hy<br />

cc<br />

hy<br />

c ccw d<br />

walls of the ectomycorrhiza-forming fungus P. tinctorius. Investigation of the<br />

attachment of Laccaria amethystea (Bull.) Murrill to Picea abies short roots<br />

showed formation of an adhesion pad (Fig. 7a) which was strongly stained by<br />

the Swift reaction for cysteine-rich proteins (Lewis and Knight 1977; Kottke<br />

1997). The <strong>surface</strong> of the hyphae in contact to the suberin layer and to each<br />

other is stained similarly (Fig. 7b). Attachment of hyphae to the basis of root<br />

hairs by Swift-positive material was found previously for P. tinctorius and<br />

cc<br />

ph<br />

sl<br />

cj<br />

hy<br />

ccw<br />

Fig. 7. a Adhesion pad of Laccaria amethystea hyphae in contact with the suberin layer<br />

of a root cap cell on top of the cortical cell. Scale 0.5 mm. b Swift positive reaction of cysteine-rich<br />

proteins in the cell wall of hyphae in contact with the root and each other<br />

(Laccaria amethystea–Picea abies). Scale 1.5 mm. c Long root of Picea abies displaying no<br />

attachment of Laccaria amethystea hyphae. Scale 15 mm. d Immunogold labelling of<br />

methyl-esterified pectins by the monoclonal antibody JIM7. The cell junction is covered<br />

by a suberized root cap cell wall lined by phenolic residues (short root of Picea abies).<br />

Scale 0.5 mm. cc Cortical cell, ccw cortical cell wall, cj cell junction, ph phenolic residues,<br />

hy hypha, sl suberin layer


220<br />

Ingrid Kottke<br />

Picea mariana Mill. B.S.P. (Thomson et al. 1989). The superficial layer of the<br />

fungal wall may contain hydrophobins, cysteine-rich proteins, self-assembling<br />

at the wall/air interface (Wösten et al. 1994; Wessels 1997; Wösten and<br />

Vocht 2000). Hydrophobins were localized using antibodies in mycorrhizas<br />

formed by P. tinctorius and E. globulus and in mycorrhizas of Tricholoma terreum<br />

(Schaeff.) Quél. with the compatible host Pinus sylvestris L. (Mankel et<br />

al. 2000; Tagu et al. 2001). The cuticle-like layer may thus be considered as the<br />

hydrophobic <strong>surface</strong> appropriate for hyphal attachment by hydrophobins.<br />

Attachment to the tips of root hairs was observed (Kottke 1997), but<br />

occurred only within a defined, susceptible zone (Thomson et al. 1989).<br />

Hyphae may also attach to the <strong>surface</strong> of root cap cells (Bonfante et al. 1998).<br />

This kind of attachment differs from that to the short root <strong>surface</strong>. Staining<br />

for cysteine-rich proteins was found to be negative (Kottke 1997). Attachment<br />

was neither followed by enlargement of hyphae or lobed ramification, nor by<br />

any digesting process (Thomson et al. 1989; Kottke 1997). Instead, thickening<br />

of fungal wall has been observed and the appearance of b-1,3-glucans in the<br />

root cell wall was shown (Bonfante et al. 1998).<br />

No attachment to the <strong>surface</strong> of long roots was found. Hyphae grow along<br />

the long roots in acropetal direction without apparent changes (Fig. 7 c).<br />

6 Digestion of the Suberin Layer and the Cell Wall of the<br />

Root Cap<br />

The cuticle-like layer covers all the cell junctions of short roots (Figs. 5a, 6).<br />

The hyphae, therefore, must penetrate the suberin layer and the wall of the<br />

moribund root cap cell when establishing the Hartig net. Vesicles, probably<br />

containing a cutinase-like enzyme were frequently observed in hyphae dissolving<br />

the suberin (Fig. 8a). The hyphae split away the suberized root cap cell<br />

wall and proliferate below, on top of the <strong>surface</strong> of the cortical cell (Fig. 8b, c,<br />

d). This process may explain why finally, when the hyphal sheath covers the<br />

rootlet, the cuticle-like layer is no longer found. The suberin layer became<br />

integrated into the hyphal sheath (Fig. 8d).<br />

The hyphae digest the suberin layer locally and disrupt the root cap cell<br />

wall, but do not attack the wall of the live cortical cell (Fig. 8a, b). While the<br />

enzyme activity remains to be proven in situ, there are many indications for a<br />

controlled cell wall hydrolyzing activity of ECM fungi (for review, see Cairney<br />

and Burke 1994). ECM fungi digest cell wall material, including the suberin<br />

layer of the moribund root cap, but not material of live cells during mycorrhiza<br />

formation (Chilvers 1968; Piché et al. 1983b; Kottke and Oberwinkler<br />

1986). A strict spatial and temporal regulation of enzyme activity has, thus, to<br />

be expected when the hyphae contact alive cells.


ccw<br />

cc<br />

hy<br />

sl<br />

rccw<br />

hy<br />

hy<br />

7 Hartig Net Formation<br />

cc<br />

13 Root Surface in Ectomycorrhizas 221<br />

a b<br />

sl<br />

ccw<br />

sl<br />

rccw<br />

c cc<br />

d<br />

Fig. 8a–d. Cuticle-like suberin layer involved in mycorrhiza formation (Laccaria<br />

amethystea-Picea abies). a Local digestion of the suberin layer and root cap cell wall, no<br />

disturbance of cortical cell wall, vesicles probably containing enzymes (arrowhead,scale<br />

1 mm). b Hypha splitting off the suberin layer and proliferating beneath, on the <strong>surface</strong><br />

of the cortical cell wall. Scale 0.5 mm. c Hypha penetrating between cell junction, suberin<br />

layer partly digested (arrow) and partly preserved (arrowhead), lobed growth of hyphae<br />

visible (arrow). Scale 1 mm. d Hyphae proliferating under the suberin layer (arrowheads)<br />

show lobed branching typical of Hartig net structure (arrows). Scale 1 mm. cc Cortical<br />

cell, ccw cortical cell wall, hy hypha, rccw root cap cell wall, sl suberin layer<br />

Lobed growth of hyphae indicating the initialization of the Hartig net was<br />

found in connection with the digestion of the cuticle-like layer (Fig. 8 c, d).<br />

Thomson et al. (1989) described the formation of hyphal lobes at the base of<br />

root hairs. Lobed hyphal growth on the root <strong>surface</strong> has been observed by<br />

SEM (Jacobs et al. 1989; Brunner and Scheidegger 1992), but the connection to<br />

sl<br />

hy<br />

hy<br />

hy


222<br />

Ingrid Kottke<br />

suberin digestion is shown here for the first time. It remains to be elucidated<br />

if there is a direct signalling link between the digestion of the suberin and the<br />

change of hyphal growth characters.After digesting the suberin layer and disrupting<br />

the root cap cell wall, the hyphae come into direct contact with the live<br />

cortical cells. There may then be additional signals involved in triggering the<br />

hyphal growth changes at the root <strong>surface</strong> (Salzer et al. 1997, 2000).<br />

8 Pectins in the Cortical Cell Walls of Nonmycorrhizal Long<br />

and Mycorrhizal Short Roots<br />

Methyl-esterified pectins were localized in the root cell walls of Picea abies<br />

using the monoclonal antibody JIM7 (K. Roberts, John Innes Institute, Norwich<br />

UK; Fig. 7d). No difference in the amount of pectin was found between<br />

cortical cells in contact to hyphae and those lacking hyphal contact when<br />

short roots and mycorrhizas were compared (Fig. 9). The cortical cells of noncolonized,<br />

long roots, however, were significantly more densely marked by the<br />

antibody (Fig. 9). There was no difference between both root types in the<br />

amounts of methyl-esterified pectins in the cell walls of the meristems<br />

(Fig. 9). During differentiation, the amounts of methyl-esterified pectins obviously<br />

increase in cortical cell walls of long roots, but are reduced in cortical<br />

cell walls of short roots. There is no indication for a digestion of pectins by the<br />

hyphae as no changes in the amounts of pectins was found during early stages<br />

of Laccaria amethystea- Picea abies mycorrhiza formation. Balestrini et al.<br />

(1996) could not find any indication for polygalacturonase activity during<br />

ECM development between Coryllus avellana and Tuber magnatum either.<br />

The authors supposed de-esterification of the pectins according to increased<br />

labelling of de-esterified pectins after mycorrhiza formation. In the case of P.<br />

abies, however, immunogold labelling by the monoclonal antibody JIM5<br />

showed low amounts of de-esterified pectins and labelling decreased from<br />

inner cortical cells to outer cortical cells (not shown). High labelling of<br />

methyl-esterified pectins was detected in roots of Daucus carota L. and Avena<br />

sativa L. (Knox et al. 1990). This finding would support the view that fast<br />

growing roots contain high amounts of methyl-esterified pectins in cortical<br />

cells.<br />

It is unclear whether the amounts of methyl-esterified pectins have any<br />

influence on mycorrhiza formation. There is too little knowledge on the<br />

importance of methyl-esterified pectins for stability or plasticity of cell walls<br />

and cell-to-cell adhesion (Liners et al. 1994). Previously, reduction of the cell<br />

wall-bound ferulic acid, linking pectic substances in the cell wall matrix, was<br />

found to occur during mycorrhiza formation of Picea abies, Larix decidua and<br />

Arbutus menziesii (Münzenberger et al. 1990, 1995, Weiss et al. 1999). Less<br />

rigid cortical cell walls were considered a prerequisite for intercellular hyphal<br />

penetration during Hartig net establishment.


9 Conclusions<br />

13 Root Surface in Ectomycorrhizas 223<br />

Fig. 9. Amount of immunogold labelling by the monoclonal antibody JIM7 against<br />

methyl-esterified pectins. Counting of gold granules was carried out by means of image<br />

analysis in different compartments of ectomycorrhizas, short roots, and long roots.<br />

Material collected from in vitro cultures of Picea abies inoculated by Laccaria amethystea<br />

The <strong>surface</strong> of short roots appears to be important in ECM initialization. So<br />

far we have only started to understand the process. Further research is needed<br />

to clarify changes of cell wall components and signal exchanges involved.<br />

Some progress was, however, obtained by structural and molecular investigations<br />

during the last few years. Tight attachment of hyphae to the root <strong>surface</strong><br />

is established between hydrophobins on the hyphal <strong>surface</strong> and the hydrophobic<br />

root <strong>surface</strong>. The hydrophobic root <strong>surface</strong> derives from the residue of the<br />

suberized root cap of short roots. The lack of a suberized root cap might be<br />

involved in the lack of a Hartig net in long roots. Digestion of the suberin layer<br />

and the root cap cell wall may mean the occurrence of a slight, transient and<br />

locally restricted aggressive phase during ectomycorrhiza formation and may<br />

explain the slight, transient defence reactions in the early phase (Salzer et al.<br />

1997, 2000). The lack of pectin digestion by the fungus might avoid severe<br />

defence reactions of the root. The locally restricted digestion of the moribund,<br />

suberized root cap cell wall may, however, alternatively be looked upon<br />

as a saprophytic phase of interaction. Ectomycorrhizal fungi phylogenetically<br />

derive from saprophytes and not from parasites (Bresinsky et al. 1999) and<br />

many species have preserved saprophytic growth facilities.


224<br />

Ingrid Kottke<br />

When dissolving the suberin layer locally, the hyphae start lobed growth<br />

typical of Hartig net structure. It is unclear so far if the digestion of the cuticle-like<br />

layer has itself an inductive effect on hyphal growth. Signals obtained<br />

from the live cortical cells, reached after digestion of the suberized root cap<br />

layer, may be more decisive in change of hyphal growth characters. The<br />

described phenomena are unique in ECM formation and, as far is known, do<br />

not occur in any other <strong>plant</strong>-fungus interaction system.<br />

Acknowledgements. The valuable comments on the manuscript and the introduction to<br />

immunogold-labelling by Paola Bonfante is greatly appreciated. I also express my gratitude<br />

to Bettina Grüninger and Esther Strasdas for carrying out the JIM7/JIM5 labelling<br />

studies.<br />

References and Selected Reading<br />

Agerer R (1998) Colour atlas of ectomycorrhizae. Einhorn-Verlag, Schwäbisch Gmünd,<br />

140 pp<br />

Balestrini R, Hahn MG, Bonfante P (1996) Location of cell-wall components in ectomycorrhizae<br />

of Coryllus avellana and Tuber magnatum. Protoplasma 191: 55–69<br />

Blasius D, Feil W, Kottke I, Oberwinkler F (1986) Hartig net structure and formation in<br />

fully ensheathed ectomycorrhizas. Nordic J Bot 6: 837–842<br />

Bonfante P, Balestrini R, Martino E, Perotto S, Plassard C, Mousain D (1998) Morphological<br />

analysis of early contacts between pine roots and two ectomycorrhizal Suillus<br />

strains. Mycorrhiza 8:1–10<br />

Bremer K, Chase MW, Stevens PF (1998) An ordinal classification for the families of flowering<br />

<strong>plant</strong>s. Ann Mo Bot Gard 85:531–553<br />

Bresinsky A, Jarosch M, Fischer M, Schönberg I, Wittmann-Bresinsky B (1999) Phylogenetic<br />

relationship within Paxillus s. l. (Basidiomycetes, Boletales): Separation of a<br />

southern hemisphere genus. Plant Biol 1:327–333<br />

Brunner I, Scheidegger C (1992) Ontogeny of synthesized Picea abies(L.) Karst.- Hebeloma<br />

crustuliniforme (Bull. ex St Amans) Quél. ectomycorrhizas. New Phytol 120:359–<br />

369<br />

Cairney JW, Burke RM (1994) Fungal enzymes degrading <strong>plant</strong> cell walls: their possible<br />

significance in the ectomycorrhizal symbiosis. Mycol Res 98:1345–1356<br />

Chilvers GA (1968) Low power electron microscopy of the root cap region of eucalypt<br />

mycorrhizas. New Phytol 67:663<br />

Clowes FAL (1951) The structure of mycorrhizal roots of Fagus sylvatica. New Phytol<br />

50:1–16<br />

Clowes FAL (1954) The root-cap of ectotrophic mycorrhizas. New Phytol 53:525–9<br />

Hatch AB, Doak KD (1933) Mycorrhizal and other features of the root system of Pinus.J<br />

Arnold Arbor 14:85–99<br />

Jacobs PF, Peterson RL, Massicotte HB (1989) Altered fungal morphogenesis during early<br />

stage ectomycorrhiza formation in Eucalyptus pilularis. Scann Microsc 3:249–255<br />

Knox JP, Linstead PJ, King J, Cooper C, Roberts K (1990) Pectin esterification in spatially<br />

regulated both within cell walls and between developing tissues of root apices. Planta<br />

181:512–521<br />

Kottke I (1997 ) Fungal adhesion pad formation and penetration of root cuticle in early<br />

stage Picea abies-Laccaria amethystea mycorrhizas. Protoplasma 196:55–64


13 Root Surface in Ectomycorrhizas 225<br />

Kottke I, Oberwinkler F (1986) Root-fungus interactions observed on initial stages of<br />

mantle formation and Hartig net establishment in mycorrhizas of Amanita muscaria<br />

(L. ex Fr.) Hooker on Picea abies (L.) Karst. in pure culture. Can J Bot 64: 2348–2354<br />

Kottke I, Oberwinkler F (1987) Cellular structure and function of the Hartig net: coenocytic<br />

and transfer cell-like organization. Nordic J Bot 7:85–95<br />

Kottke I, Oberwinkler F (1989) Amplification of root-fungus interface in ectomycorrhizae<br />

by Hartig net architecture. Ann Sci For 46 Suppl:737s–740s<br />

Kottke I, Oberwinkler F (1990) Comparative investigations on the differentiation of the<br />

endodermis and the development of the Hartig net in mycorrhizae of Picea abies and<br />

Larix decidua. Trees 4:41–48<br />

Kottke I, Rapp C, Oberwinkler F (1986) Zur Anatomie gesunder und “kranker” Feinstwurzeln<br />

von Fichten: Meristem und Differenzierungen in Wurzelspitzen und Mykorrhizen.<br />

Eur J For Pathol 16:159–171<br />

Kottke I, Münzenberger B, Oberwinkler F (1996) Structural approach to function in<br />

ectomycorrhizas. In: Rennenberg H, Eschrich W, Ziegler H (eds) Trees – contribution<br />

to modern tree physiology. SPB Academic, The Hague, pp 3–22<br />

Laurent P, Voiblet C, Tagu D, de Carvalho D, Nehls U, De Bellis R, Ballestrini R, Bauw G,<br />

Bonfante P, Martin F (1999) A novel class of ectomycorrhiza-regulated cell wall<br />

polypeptides in Pisolithus tinctorius. Mol Plant Microbe Interact 12:862–871<br />

Lei J, Wong KK, Piché Y (1991) Extracellular Concanavalin A-binding sites during early<br />

interaction between Pinus banksiana and two closely related genotypes of the ectomycorrhizal<br />

basidiomycete Laccaria bicolor. Mycol Res 95:357–363<br />

Lewis PR, Knight DP (1977) Staining methods for sectioned material. North-Holland<br />

Publishing Company, Amsterdam, 311 pp<br />

Liners F, Gaspar T, Van Cutsem P (1994) Acetyl- and methyl-esterification of pectins of<br />

friable and compact sugar-beet calli: consequences for intercellular adhesion. Planta<br />

192:545–556<br />

Mankel A, Krause K, Genenger M, Kost G, Kothe E (2000) A hydrophobin accumulated in<br />

the Hartig net of ectomycorrhiza formed between Tricholoma terreum and its compatible<br />

host tree is missing in an incompatible association. J Appl Bot 74:95–99<br />

Marks GC, Foster RC (1973) Structure, morphogenesis and ultrastructure of ectomycorrhizae.<br />

In: Marks GC, Kozlowski TT (eds) Ectomycorrhizae. Their ecology and physiology.<br />

Academic Press, New York, London, pp 1–41<br />

Martin F, Laurent P, de Carvalho D,Voiblet C, Balestrini R, Bonfante P, Tagu D (1999) Cell<br />

wall proteins of the ectomycorrhizal basidiomycete Pisolithus tinctorius: identification,<br />

function, and expression in symbiosis. Fungal Gen Biol 27:161–174<br />

Massicotte HB, Peterson RL, Ackerley CA, Piche Y (1986) Structure and ontogeny of<br />

Alnus crispa-Alpova diplophloeus ectomycorrhizae. Can J Bot 64:177–192<br />

Massicotte HB, Peterson RL, Ackerly CA (1987a) Ontogeny of Eucalyptus pilularis-<br />

Pisolithus tinctorius ectomycorrhizae. I. Light microscopy and scanning electron<br />

microscopy. Can J Bot 65:1927–1939<br />

Massicotte HB, Peterson RL, Ackerly CA (1987b) Ontogeny of Eucalyptus pilularis-<br />

Pisolithus tinctorius ectomycorrhizae. II. Transmission electron microscopy. Can J Bot<br />

65:1940–1947<br />

Müller H (1906) Über die Metakutinisierung der Wurzelspitze und über die verkorkten<br />

Scheiden in den Achsen der Monokotyledonen. Bot Z 4:54–64<br />

Münzenberger B, Heilemann J, Strack D, Kottke I, Oberwinkler F (1990) Phenolics of<br />

mycorrhizas and non-mycorrhizal roots of Norway spruce. Planta 182:142–148<br />

Münzenberger B, Kottke I, Oberwinkler F (1995) Reduction of phenolics in mycorrhizas<br />

of Larix decidua Mill. Tree Physiol 15:191–196<br />

Noelle W (1910) Studien zur vergleichenden Anatomie und Morphologie der Koniferenwurzeln<br />

mit Rücksicht auf die Systematik. Bot Z 68:169–266


226<br />

Ingrid Kottke<br />

Oh KI, Melville LH, Peterson RL (1995) Comparative structural study of Quercus serrata<br />

and Q. acutissima formed by Pisolithus tinctorius and Hebeloma cylindrosporum.<br />

Trees 9:171–179<br />

Piché Y, Peterson RL, Ackerley KA (1983a) Early development of ectomycorrhizal short<br />

roots of pine (Pinus strobus). Scann Electron Microsc 111:1467–1474<br />

Piché Y, Peterson RL, Howarth MJ, Fortin JA (1983b) A structural study of the interaction<br />

between the ectomycorrhizal fungus Pisolithus tinctorius and Pinus strobus roots.<br />

Can J Bot 61:1185–119<br />

Plaut M (1918) Über die morphologischen und mikroskopischen Merkmale der Periodizität<br />

der Wurzel, sowie über die Verbreitung der Metakutisierung der Wurzelhaube<br />

im Pflanzenreich. Festschr. 100. j. Best. K. Württ. Landw. Hochsch. Hohenheim.<br />

Verlag Ulmer, Stuttgart, S 129–151<br />

Salzer P, Boller T (2000) Elicitor induced reactions in mycorrhizae and their suppression.<br />

In: Podila GK, Douds DD Jr (eds) Current advances in mycorrhizae research. Symposium<br />

Series, APS Press, The American Phytopathological Society, St. Paul, Minnesota,<br />

pp 1–10<br />

Salzer P, Münzenberger B, Schwacke R, Kottke I, Hager A (1997) Signalling in ectomycorrhizal<br />

fungus-root interactions. Trees – contribution to modern tree physiology.<br />

SPB Academic, The Hague, pp 339–356<br />

Sitte, P (1975) Die Bedeutung der molekularen Lamellenbauweise von Korkzellwänden.<br />

Biochem Physiol Pflanzen 168:287–297<br />

Tagu D, Martin T (1996) Molecular analysis of cell wall proteins expressed during the<br />

early steps of ectomycorrhiza development. New Phytol 133:73–85<br />

Tagu D, Lapeyrie F, Ditengou F, Lagrange H, Laurent P, Missoum N, Nehls U, Martin F<br />

(2000) Molecular aspects of ectomycorrhiza development. In: Podila GK, Douds DD Jr<br />

(eds) Current advances in mycorrhizae research. Symposium Series, APS Press, The<br />

American Phytopathological Society, St. Paul, Minnesota, pp 69–90<br />

Tagu D, De Bellis R, Balestrini R, De Vries OM, Piccoli G, Stocchi V, Bonfante P, Martin F<br />

(2001) Immunolocalization of hydrophobin HYDPt-1 from the ectomycorrhizal<br />

basidiomycete Pisolithus tinctorius during colonization of Eucalyptus globulus roots.<br />

New Phytol 149:127–135<br />

Thomson J, Melville IH, Peterson RL (1989) Interaction between the ectomycorrhizal<br />

fungus Pisolithus tinctorius and root hairs of Picea mariana (Pinaceae). Am J Bot<br />

76:632–636<br />

Weiss M, Schmidt J, Neumann D, Wray V, Christ R, Strack D (1999) Phenylpropanoids in<br />

mycorrhizas of Pinaceae. Planta 208:491–502<br />

Wessels JG (1997) Hydrophobins: Proteins that change the nature of the fungal <strong>surface</strong>.<br />

Adv Microbial Physiol 38:1–45<br />

Wilcox HE (1968) Morphological studies of the roots of Redpine, Pinus resinosa. II. Fungal<br />

colonisation of roots and the development of mycorrhizae. Am J Bot 55:688–700<br />

Wilcox HE (1954) Primary organization of active and dormant roots of noble-fir, Abies<br />

procera. Am J Bot 41:812–821<br />

Wösten HA, Asgeirdottir SA, Krook JH, Drenth JH, Wessels JG (1994) The fungal<br />

hydrophobin Sc3p self-assembles at the <strong>surface</strong> of aerial hyphae as a protein membrane<br />

constituting the hydrophobic rodlet layer. Eur J Cell Biol 63:122–129<br />

Wösten HA, de Vocht ML (2000) Hydrophobins, the fungal coat unravelled. Biochim Biophys<br />

Acta Rev Biomembr 1469:79–86<br />

Wong K, Montpetit D, Piché Y, Lei J (1990) Root colonization by four closely related genotypes<br />

of ectomycorrhizal basidiomycete Laccaria bicolor (Maire) Orton – comparative<br />

studies using electron microscopy. New Phytol 116:669–679


14 Cellular Ustilaginomycete – Plant Interactions<br />

Robert Bauer and Franz Oberwinkler<br />

1 Introduction<br />

The Ustilaginomycetes comprises more than 1300 species in ca. 80 genera of<br />

basidiomycetous <strong>plant</strong> parasites. They occur throughout the world, although<br />

many species are restricted to tropical, temperate or arctic regions. Some<br />

species of Ustilago and Tilletia, e.g., the barley, wheat or maize smut fungi, are<br />

well known because they are of economic importance (Trione 1982; Thomas<br />

1989). For example, from 1983 to 1988 the barley smut fungi reduced annual<br />

yields from 0.7 to 1.6 % in the prairie provinces in central Canada, causing<br />

annual losses of about US $8,000,000 (Thomas 1989). Tilletia contraversa is<br />

important in the international wheat trade (Trione 1982) and 2–5 % in a corn<br />

field are generally infected by Ustilago maydis, while up to 80 % of a field can<br />

be infected if conditions are good for the fungus. On the other hand, the galls<br />

of U. maydis are regarded as a delicacy in the Mesoamerican tradition. They<br />

are known in Mexico as “Huitlacoche” and in parts of the USA. as “maize<br />

mushroom”,“Mexican truffles” or “caviar azteca”.<br />

This chapter focuses predominantly on the cellular interaction of the Ustilaginomycetes<br />

that represents one of the three classes of the Basidiomycota<br />

(Begerow et al. 1997).<br />

2 The Term Smut Fungus<br />

Like the terms agaric, polypore, lichen etc., the term smut fungus circumscribes<br />

the organization and life strategy of a fungus, but it is not a taxonomic<br />

term.Smut fungi evolved in different fungal groups.Most smut fungi are in the<br />

Ustilaginomycetes. Other smut fungi, in the Microbotryales, are members of<br />

the Urediniomycetes (Bauer et al. 1997; Begerow et al. 1997). There are significant<br />

convergences between the urediniomycetous and the ustilaginomycetous<br />

phragmobasidiate smut fungi. Certain taxa of both groups are similar with<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


228<br />

respect to soral morphology,teliosporogenesis,life cycle,basidial morphology<br />

and host range.<br />

3 Life Cycle<br />

The species of the Ustilaginomycetes share an essentially similar life cycle<br />

with a saprobic haploid phase and a parasitic dikaryophase (e.g., Sampson<br />

1939). The haploid phase usually commences with the formation of<br />

basidiospores after meiosis of the diploid nucleus in the basidium and ends<br />

with the conjugation of compatible haploid cells to produce dikaryotic, parasitic<br />

mycelia. The dikaryotic phase ends with the production of basidia.<br />

Almost all Ustilaginomycetes sporulate on or in parenchymatic tissues of<br />

the hosts. In the ustilaginomycetous smut fungi, the young basidium becomes<br />

thick-walled and at maturity separates from the sorus, thus functioning as a<br />

dispersal agent, the teliospore. The teliospores are usually the most conspicuous<br />

stage in the smut’s life cycle. Most of the Ustilaginomycetes are dimorphic,<br />

producing a yeast or yeast-like phase in the haploid state.<br />

4 Hosts<br />

Robert Bauer and Franz Oberwinkler<br />

The Ustilaginomycetes are ecologically well characterized by their <strong>plant</strong> parasitism.<br />

Two species occur on spike mosses (Bauer et al. 1999), one on ferns,<br />

two on conifers, whereas all other Ustilaginomycetes parasitize angiosperms<br />

with a high proportion of species on monocots, especially on Poaceae and<br />

Cyperaceae. Thus, of the ca. 1300 species, ca. 42 % occurs on Poaceae and ca.<br />

15 % on Cyperaceae. Concerning the hosts two points are remarkable: (1) with<br />

a few exceptions the teliospore-forming species of the Ustilaginomycetes parasitize<br />

nonwoody herbs, whereas those without teliospores prefer woody<br />

trees or bushes. However, almost all species sporulate on parenchymatic tissues<br />

of the hosts. (2) Two of the angiosperm families with the highest number<br />

of species, the Orchidaceae with about 20,000 species and the Poaceae with<br />

about 9000 species, play quite a different role for the Ustilaginomycetes. There<br />

are no known species on Orchidaceae while the Poaceae are the most important<br />

host family of the Ustilaginomycetes. This can be tentatively explained by<br />

the completely different ecological strategies of the two families. Orchid<br />

species subsist with a few isolated individuals and are highly specialized for<br />

insect pollination. The Poaceae, however, disperse their dusty pollen by the<br />

wind and cover about a third of the land <strong>surface</strong> with numerous individuals.<br />

The ecology of the Ustilaginomycetes, with dusty teliospores or basidiospores<br />

dispersed by the wind and with the requirement of extensive host populations<br />

for successful infection, corresponds well to the ecology of the Poaceae.


5 Cellular Interactions<br />

14 Cellular Ustilaginomycete – Plant Interactions 229<br />

Information concerning the cellular interaction of the Ustilaginomycetes has<br />

come from only a few studies (Mims 1982, 1991; Mims and Nickerson 1986;<br />

Luttrell 1987; Nagler 1989; Nagler et al. 1990; Snetselaar and Tiffany 1990;<br />

Mims et al. 1992; Snetselaar and Mims 1994; Bauer et al. 1995, 1997; Martinez<br />

et al. 1999). Hyphae of the Ustilaginomycetes in contact with host <strong>plant</strong> cells<br />

possess zones of host–parasite interaction with fungal deposits resulting<br />

from exocytosis of primary interactive vesicles. These zones provide ultrastructural<br />

characters diagnostic for higher groups in the Ustilaginomycetes<br />

(Bauer et al. 1997). Initially, primary interactive vesicles with electron-opaque<br />

contents accumulate in the fungal cell (Fig. 1). Depending on the fungal<br />

species, the primary interactive vesicles may fuse with one another before<br />

being exocytosed from the fungal cytoplasm. Electron-opaque deposits also<br />

Fig. 1. Interactive<br />

vesicles in a hypha of<br />

Exobasidium pachysporum.<br />

Scale bar<br />

0.2 mm


230<br />

Robert Bauer and Franz Oberwinkler<br />

appear at the host side, opposite the point of contact with the fungus. Detailed<br />

studies indicate that these deposits at the host side originate from the exocytosed<br />

fungal material by transfer towards the host plasma membrane (Fig. 2;<br />

Bauer et al. 1995, 1997).<br />

The following major types, minor types and variations were recognized by<br />

Bauer et al. (1995, 1997).<br />

5.1 Local Interaction Zones<br />

Fig. 2. Transfer stage<br />

between Mycosyrinx cissi<br />

(upper cell) and its host<br />

(lower cell). Note the infiltrated<br />

host cell wall<br />

(between the arrows) and<br />

the electron-opaque<br />

deposit at the host side<br />

(arrrowhead). Scale bar<br />

0.1 mm<br />

Short-term production of many primary interactive vesicles per interaction<br />

site results in local interaction zones. This type of cellular interaction characterizes<br />

the Entorrhizomycetidae and Exobasidiomycetidae (Bauer et al. 1997).


Fig. 3. Local interaction<br />

zone without interaction<br />

apparatus between Conidiosporomyces<br />

ayresii<br />

(upper cell) and its host<br />

(lower cell) showing the<br />

secretion profile of one<br />

interactive vesicle (arrow).<br />

Note the electron-opaque<br />

deposit at the host side<br />

(arrowhead). Host<br />

response to infection is<br />

visible at R. Scale bar<br />

0.5 mm<br />

14 Cellular Ustilaginomycete – Plant Interactions 231<br />

5.1.1 Local Interaction Zones without Interaction Apparatus<br />

Primary interactive vesicles fuse individually with the fungal plasma membrane<br />

(Fig. 3). Depending upon the species, local interaction zones without<br />

interaction apparatus are present in intercellular hyphae or in haustoria. This<br />

type of cellular interaction characterizes the Entorrhizomycetidae, Georgefischeriales,<br />

Tilletiales and Microstromatales (Bauer et al. 1997).<br />

5.1.2 Local Interaction Zones with Interaction Apparatus<br />

Fusion of the primary interactive vesicles precedes exocytosis. This type of<br />

cellular interaction characterizes the Exobasidianae (Bauer et al. 1997).<br />

5.1.2.1 Local Interaction Zones with Simple Interaction Apparatus<br />

Primary interactive vesicles fuse to form one large secondary interactive vesicle<br />

per interaction site (Fig. 4). Interaction zones of this type are only located<br />

in intercellular hyphae. This type of cellular interaction characterizes the<br />

Entylomatales (Bauer et al. 1997).<br />

5.1.2.2 Local Interaction Zones with Complex Interaction Apparatus<br />

Numerous primary interactive vesicles fuse to form several secondary interactive<br />

vesicles per interaction site. Fusion of the secondary interactive vesicles


232<br />

Robert Bauer and Franz Oberwinkler<br />

Fig. 4. Local interaction zone between Entyloma hieracii (upper cell) and its host (lower<br />

cell) showing the exocytosis profile of a simple interaction apparatus (arrow). Note the<br />

electron-opaque deposit at the host side (arrowhead). Host response to infection is visible<br />

at R. Scale bar 0.5 mm<br />

then results in the formation of a complex cisternal net. This type of cellular<br />

interaction characterizes the Doassansiales and Exobasidiales (Bauer et al.<br />

1997).<br />

1. Local interaction zones with complex interaction apparatus containing<br />

cytoplasmic compartments (Fig. 5)<br />

The intercisternal space of the cisternal net finally becomes integrated in<br />

the interaction apparatus. Depending upon the species, interaction zones<br />

of this type are formed by intercellular hyphae or haustoria. This type of<br />

cellular interaction characterizes the Doassansiales (Bauer et al. 1997).<br />

2. Local interaction zones with complex interaction apparatus producing<br />

interaction rings (Fig. 6)


14 Cellular Ustilaginomycete – Plant Interactions 233<br />

Fig. 5. Local interaction zone between Doassinga callitrichis (upper cell) and its host<br />

(lower cell) showing the exocytosis profile of a complex intercisternal interaction apparatus<br />

(arrow). The interaction apparatus and its intercisternal space is excluded from the<br />

cytoplasm. Note the electron-opaque deposit at the host side (arrowhead). Host<br />

response to infection is visible at R. Scale bar 0.5 mm<br />

Fig. 6. Local interaction<br />

zone between Exobasidium<br />

pachysporum (upper cell)<br />

and its host (lower cell)<br />

showing the exocytose<br />

profile of a complex interaction<br />

apparatus (arrows)<br />

and the sectioned interaction<br />

ring (double arrowheads).<br />

Note the electronopaque<br />

deposit at the host<br />

side (arrow). Initial host<br />

response to infection is<br />

visible at R. Scale bar<br />

0.5 mm


234<br />

Robert Bauer and Franz Oberwinkler<br />

The intercisternal space does not become integrated in the interaction<br />

apparatus. The transfer of fungal material towards the host plasma membrane<br />

occurs in two or three steps. The first transfer results in the deposition<br />

of a ring at the host plasma membrane. Depending upon the species,<br />

interaction zones of this type are located in intercellular hyphae or haustoria.<br />

This type of cellular interaction characterizes the Exobasidiales (Bauer<br />

et al. 1997).<br />

5.2 Enlarged Interaction Zones<br />

Continuous production and exocytosis of primary interactive vesicles results<br />

in the continuous deposition of fungal material at the whole contact area with<br />

the host cell. Depending upon the species, this type of interaction zone is<br />

located in intercellular hyphae (Fig. 2), intracellular hyphae or haustoria<br />

(Fig. 7). This type of cellular interaction characterizes the Ustilaginomycetidae<br />

(Bauer et al. 1997).<br />

Fig. 7. Haustorial apex (h)<br />

of Ustacystis waldsteiniae<br />

encased by an electronopaque<br />

vesicular matrix<br />

(arrows). Scale bar 0.5 mm


6 Conclusions<br />

14 Cellular Ustilaginomycete – Plant Interactions 235<br />

Similar development of the different interaction types occurring in the Ustilaginomycetes<br />

reveals that these interaction types are homologous to one<br />

another, thus reflecting variations of a common ancestral type. Accordingly,<br />

during the phylogenetic history the cellular interactions gradually specialized<br />

and optimized. An apomorphy for the Ustilaginomycetes is the presence of<br />

interaction zones with fungal deposits resulting from exocytosis of primary<br />

interactive vesicles. The contents of the primary interactive vesicles are transferred<br />

towards the host plasma membrane by different mechanisms in the<br />

various taxa. This parasitic process is unique among the basidiomycetes (e.g.,<br />

see Littlefield and Heath 1979). Interestingly, a similar parasitic process may<br />

occur in the downy mildews (Hickey and Coffey 1977; Coffey and Wilson<br />

1983; Wetherbee et al. 1985). The similarities include the presence of densely<br />

stained vesicles at the penetration region, the localized increase in the electron<br />

opacity of the host cell, and the deposition of electron-opaque material<br />

between host cell wall and host plasma membrane. Because of numerous fundamental<br />

differences between the downy mildews and the Ustilaginomycetes,<br />

these similarities must be interpreted as a result of convergent evolution.<br />

The transfer of fungal material towards the host plasma membrane<br />

appears to be unusual and its function is basically unknown. Bauer et al.<br />

(1995) studied the cellular interaction of the ustilaginomycete Ustacystis<br />

waldsteiniae in detail and hypothesized the following scenario for this fungus:<br />

the transferred fungal material stabilizes and binds the associated host<br />

plasma membrane and, thus prevents on the one hand, membrane recycling<br />

via endocytosis. On the other hand, exocytosis of Golgi products of the host<br />

cell at this point results in the formation of coralloid vesicular buds extending<br />

into the fungal deposit. Finally, the vesicular buds separate from the host cytoplasm.<br />

Bauer et al. (1995) assumed that in this interaction scenario the following<br />

three characteristics are advantageous for the parasite: (1) the Golgi products<br />

extruded via exocytosis could serve as direct nutriment for the parasite,<br />

(2) the formation of the coralloid vesicular buds extending into the fungal<br />

deposits results in a greatly increased transfer-like host – parasite contact <strong>surface</strong>,<br />

and (3) the content of the vesicular buds could also serve as direct nutriment<br />

for the parasite.<br />

Acknowledgements. We thank Uwe Simon for critically reading the manuscript, and the<br />

Deutsche Forschungsgemeinschaft for financial support.


236<br />

Robert Bauer and Franz Oberwinkler<br />

References and Selected Reading<br />

Bauer R, Mendgen K, Oberwinkler F (1995) Cellular interaction of the smut fungus Ustacystis<br />

waldsteiniae. Can J Bot 73:867–883<br />

Bauer R, Oberwinkler F, Vánky K (1997) Ultrastructural markers and systematics in<br />

smut fungi and allied taxa. Can J Bot 75:1273–1314<br />

Bauer R, Vánky K, Begerow D, Oberwinkler F (1999) Ustilaginomycetes on Selaginella.<br />

Mycologia 91:475–484<br />

Begerow D, Bauer R, Oberwinkler F (1997) Phylogenetic studies on large subunit ribosomal<br />

DNA sequences of smut fungi and related taxa. Can J Bot 75:2045–2056<br />

Coffey MC, Wilson UE (1983) An ultrastructural study of the late-blight fungus Phytophthora<br />

infestans and its interaction with the foliage of two potato cultivars possessing<br />

different levels of general (field) resistance. Can J Bot 61:2669–2685<br />

Hickey EL, Coffey MD (1977) A fine-structural study of the pea downy mildew fungus<br />

Peronospora pisi in its host Pisum sativum. Can J Bot 55:2845–2858<br />

Littlefield LJ, Heath MC (1979) Ultrastructure of rust fungi. Academic Press, New York<br />

Luttrell ES (1987) Relations of hyphae to host cells in smut galls caused by species of<br />

Tilletia, Tolyposporium, and Ustilago. Can J Bot 65:2581–2591<br />

Martinez C, Roux C, Dargent R (1999) Biotrophic development of Sporisorium reilianum<br />

f. sp. zeae in vegetative shoot apex of maize. Phytopathology 89:247–253<br />

Mims CW (1982) Ultrastructure of the haustorial apparatus of Exobasidium camelliae.<br />

Mycologia 74:188–200<br />

Mims CW (1991) Using electron microscopy to study <strong>plant</strong> pathogenic fungi. Mycologia<br />

83:1–19<br />

Mims CW, Nickerson NL (1986) Ultrastructure of the host-pathogen relationship in the<br />

red leaf disease of lowbush blueberry caused by the fungus Exobasidium vaccinii. Can<br />

J Bot 64:1338–1343<br />

Mims CW, Snetselaar KM, Richardson EA (1992) Ultrastructure of the leaf stripe smut<br />

fungus Ustilago striiformis: host-pathogen relationship and teliospore development.<br />

Int J Plant Sci. 153:289–290<br />

Nagler A, Oberwinkler F (1989) Haustoria in Urocystis (Tilletiales). Plant Syst Evol<br />

165:17–28<br />

Nagler A, Bauer R, Oberwinkler F, Tschen J (1990) Basidial development, spindle pole<br />

body, septal pore, and host-parasite-interaction in Ustilago esculenta. Nordic J Bot<br />

10:457–464<br />

Sampson K (1939) Life cycles of smut fungi. Trans Br Mycolog Soc 23:1–23<br />

Snetselaar KM, Tiffany LH (1990) Light and electron microscopy of sorus development<br />

in Sorosporium provinciale, a smut of big bluestem. Mycologia 82:480–492<br />

Snetselaar KM, Mims CW (1994) Light and electron microscopy of Ustilago maydis<br />

hyphae in maize. Mycolog Res 98:347–355<br />

Thomas PL (1989) Barley smuts in the prairie provinces of Canada, 1983–1988. Can J<br />

Phytopathol 11:133–136<br />

Trione EJ (1982) Dwarf bunt of wheat and its importance in international wheat trade.<br />

Plant Dis 66:1083–1088<br />

Wetherbee R, Hinch JM, Clarke AE (1985) Response of Zea mays roots to infection with<br />

Phytophthora cinnamomi II. The cortex and stele. Protoplasma 126:188–197


15 Interaction of Piriformospora indica<br />

with Diverse Microorganisms and Plants<br />

Giang Huong Pham, Anjana Singh, Rajani Malla, Rina Kumari,<br />

Ram Prasad, Minu Sachdev, Karl-Heinz Rexer, Gerhard Kost,<br />

Patricia Luis, Michael Kaldorf, François Buscot,<br />

Sylvie Herrmann, Tanja Peskan, Ralf Oelmüller,<br />

Anil Kumar Saxena, Stephané Declerck, Maria Mittag,<br />

Edith Stabentheiner, Solveig Hehl, and Ajit Varma<br />

1 Introduction<br />

An axenically cultivable Mycorrhiza-like-fungus has been described by<br />

Varma and his collaborators. The fungus was named Piriformospora indica<br />

based on its characteristic pear-shaped chlamydospores (Verma et al. 1998). P.<br />

indica tremendously improves the growth and overall biomass production of<br />

diverse hosts, including legumes (Varma et al. 1999, 2001; Singh et al. 2002a),<br />

medicinal and other <strong>plant</strong>s of economic importance (Rai et al. 2001; Singh et<br />

al. 2003a, b). Interestingly, the host spectrum of P. indica is very much like<br />

arbuscular mycorrhizal fungi (AMF). In addition, a pronounced growth promotional<br />

effect was seen with terrestrial orchids (Blechert et al. 1999; Singh<br />

and Varma 2000; Singh et al. 2000, 2002b). The fungus also provides protection<br />

when inoculated into the tissue culture-raised <strong>plant</strong>lets by overcoming the<br />

‘transient trans<strong>plant</strong> shock’ on transfer to the field and renders almost 100 %<br />

survival (Sahay and Varma 1999, 2000). The fungus has great potential in<br />

forestry, horticulture, agriculture, viticulture and especially for better establishment<br />

of tissue culture-raised <strong>plant</strong>s much needed in the <strong>plant</strong> industry<br />

(Singh et al. 2003). This would open up numerous opportunities for the optimization<br />

of <strong>plant</strong> productivity in both managed and natural ecosystems,<br />

while minimizing the risk of environmental damage. The properties of the<br />

fungus, Piriformospora indica, have been patented (Varma and Franken 1997,<br />

European Patent Office, Muenchen, Germany. Patent No. 97121440.8–2105,<br />

Nov. 1998). The culture has been deposited at Braunschweig, Germany (DMS<br />

No.11827). An 18S rDNA fragment was deposited at EMBL under the accession<br />

number AF 014929.<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


238<br />

Giang Huong Pham et al.<br />

The fungus forms inter- and intracellular hyphae in the root cortex, often<br />

differentiating into dense hyphal coils and chlamydospores. Like AM fungi,<br />

hyphae multiply within the host cortical tissues and never traverse through<br />

the endodermis. Likewise, they also do not invade the aerial portion of the<br />

<strong>plant</strong> (stem and leaves).<br />

This chapter details the interaction of P. indica with various groups of<br />

microorganisms and higher <strong>plant</strong>s.<br />

2 Interaction with Microorganisms<br />

2.1 Rhizobacteria<br />

Piriformospora indica and the respective bacteria Pseudomonas fluorescence<br />

and Azotobacter chroococcum were placed on defined modified Aspergillus<br />

medium (see Chap. 30). After 7-day incubation at 25 °C, it was found that Ps.<br />

fluorescence completely blocked the growth of the fungus. P. indica acquired<br />

immense, but tiny chlamydospores, perhaps to overcome the stress (Fig. 1).<br />

Plausible reasons for the inhibition could be the production of ammonia,<br />

HCN, siderophores, antibiotics or chitinase. In contrast, Az. chroococcum promoted<br />

the growth of the fungus which produced extensive mycelium with low<br />

and delayed sporulation. The strains of Pseudomonas sp. and Ps. putrida also<br />

Fig. 1. Interactions with Pseudomonas fluorescens (left) and Bradyrhizobium sp. (right).<br />

P. indica was grown in the center of plates with modified Aspergillus medium for 48 h.<br />

Then freshly grown (early log phase) bacteria were inoculated four times at an equal distance<br />

close to the margin of the plate. Incubation was done at 25±2 °C. Photographed<br />

after 5 days. The growth of P. indica was strongly suppressed by Ps. fluorescens and promoted<br />

by Bradyrhizobium sp.


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 239<br />

initially blocked the growth of P. indica, but the fungus rapidly recovered.<br />

Bacillus subtilis had neutral effects, whereas strains of Azospirillum and<br />

Bradyrhizobium promoted the growth of the fungus.<br />

2.2 Chlamydomonas reinhardtii<br />

Ch. reinhardtii and P. indica were allowed to grow on MMN 1/10 medium. The<br />

alga was inoculated as a streak either only on one side or on both sides of a<br />

fungal disc. Both microorganisms grew well in both experiments, but the<br />

growth of the alga was more intense and the color of the colony was a much<br />

darker green if there was only one streak (Fig. 2). At this stage, to our knowledge,<br />

no suitable explanation can be offered for these phenomena.<br />

2.3 Sebacina vermifera<br />

One disc each of P. indica and S. vermifera were placed on Aspergillus<br />

medium. The distance between the two inocula was 4 cm. Both fungi grew<br />

normally without inhibiting the growth of each other. The most interesting<br />

part was that after 7 days at the intersection of two colonies, hyphae turned<br />

highly intertwined, inflated and produced a large number of chlamydospores.<br />

Therefore, both strains were able to block each other with a typical deadlock<br />

Fig. 2. Interaction with Chlamydomonas reinhardtii. P. indica was grown on MS<br />

medium for 48 h. Thereafter, the green alga was streaked on one side (left) or on both<br />

sides (right) of the mycelium. Incubation was carried out under 52 µmol/m light and<br />

24±2 °C temperature. Left Dark green strongly grown algal colonies


240<br />

Giang Huong Pham et al.<br />

reaction. No evidence for the production of basidia and basidiospores was<br />

recorded (Fig. 3).<br />

2.4 Other Soil Fungi<br />

Several commonly occurring soil fungi were tested for the interaction with P.<br />

indica. The results were highly diverse (Fig. 4). The growth of Aspergillus<br />

sydowii, Rhizopus stolonifer, andAspergillus niger was completely blocked by<br />

P. indica. The growth of Cunninghamella echinulata was reduced, whereas<br />

Rhizopus oryzae, As. flavus and Aspergillus sp. completely blocked the growth<br />

of P. indica. The data indicated that P. indica divulges a wide range of interaction<br />

types with diverse soil fungi.<br />

2.5 Gaeumannomyces graminis<br />

Fig. 3. Interaction with Sebacina vermifera.<br />

The fungal inocula were placed<br />

on Aspergillus medium about 3 cm<br />

apart and incubated for 5 days. The<br />

mycelia formed a sharp demarcation<br />

line where they touched<br />

In his pioneering work, Dehne (1982) was able to show that AM fungi are able<br />

to reduce soil-borne diseases and/or the severity of diseases caused by root<br />

pathogens. P. indica was challenged with a virulent root and seed pathogen G.<br />

graminis (Fig. 5). In a confrontational experiment, initially the mycelia were<br />

not able to overcome each other, resulting in sharp borderlines between the<br />

colonies. After prolonged incubation, P. indica started to invade into the area<br />

of G. gramins and caused a lysis of the root pathogen hyphae.<br />

In another experiment, when the P. indica was allowed to grow earlier and<br />

the pathogen was inoculated later in the center of the solidified Aspergillus<br />

medium, the pathogen growth was completely blocked. A culture filtrate of P.<br />

indica also completely inhibited the growth of the pathogen. These experi-


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 241<br />

Fig. 4a–f. Interactions with soil fungi. One disc of P. indica and a soil fungus each were<br />

placed on the solidified Aspergillus medium. Closed arrows indicate where P. indica was<br />

inoculated, open arrows indicate the inoculation placement of the respective fungus. a<br />

Aspergillus sydowii, b Rhizopus stolonifer, c Aspergillus niger, d Rhizopus oryzae, e<br />

Aspergillus flavus, f Aspergillus sp. P. indica strongly suppressed the growth of some<br />

fungi studied (a–c) or was itself suppressed (f)


242<br />

Giang Huong Pham et al.<br />

Fig. 5. Interaction with Gaeumannomyces graminis (G). Two discs of P. indica (P) and<br />

the root pathogen were each placed at equal distances. Incubation was done for 7 days on<br />

Aspergillus medium in the dark at 25±2 °C. Right Top view of the mycelia, left bottom<br />

view of the mycelia shining through the agar. The mycelia formed a sharp demarcation<br />

line where they made contact; after prolonged incubation, P. indica invaded the hyphal<br />

mat of G. graminis<br />

ments demonstrated that P. indica is able to act as a potential agent for biological<br />

control of root diseases; however, the chemical nature of the inhibitory<br />

factor is still unknown.<br />

3 Interaction with Bryophyte<br />

To test the ability of P. indica to interact with different kinds of moss and liverwort,<br />

the <strong>plant</strong>s were first grown in axenic culture. In co-culture with the<br />

fungus, Eurhynchium praelongum and Cephalozia bicuspidata were weakly<br />

colonized without causing severe symptoms to the gametophyte. In Riccardia<br />

incurvata heavy colonization took place, but the growth promotional effect<br />

was hardly significant. In this liverwort the interaction was intense and the<br />

fungus entered deeply into the thallus. In a further study, the interaction<br />

found in this species will be compared to the interaction found in Aneura pinguis,<br />

a liverwort of the same family Aneuraceae.<br />

4 Interaction with Higher Plants<br />

A large number of diverse higher <strong>plant</strong>s (mono- and dicots) interacted with P.<br />

indica (Table 1). This included terrestrial, annual and perennial herbs, and<br />

woody <strong>plant</strong>s. Interestingly, P. indica mimics a number of symbiotic proper-


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 243<br />

Table 1. Host spectrum tested for P. indica<br />

Hosts<br />

Abrus precatorius L.<br />

Acacia catechu (L.) Willd.<br />

A. nilotica (L.) Del.<br />

Adhatoda vasica Nees<br />

Aneura pinguis (L.) Dumort.<br />

Arabidopsis thaliana (L.) Heynh.<br />

Artemisia annua L.<br />

Azadirachta indica A. Juss.<br />

Bacopa monnieria (L.) Wett.<br />

Cassia angustifolia Vahl<br />

Chlorophytum borivilianum Santapau & R.R. Fernandez<br />

Ch. tuberosum Baker<br />

Cicer arietinum L.<br />

Coffea arabica L.<br />

Cymbopogon martini (Roxb.) W. Wats.<br />

Dactylorhiza fuchsii (Druce) Soo’<br />

D. incarnata (L.) Soo’<br />

D. maculata (L.) Verm.<br />

D majalis (Rchb.) P.F. Hunt & Summerh.<br />

D. purpurella (Steph’s) Soo’<br />

Daucus carota L.<br />

Delbergia sissoo Roxb.<br />

Glycine max (L.) Merr.<br />

Lycopersicon esculentum Mill.<br />

Nicotiana attenuata Torr. ex S. Wats L.<br />

N. tabaccum L.<br />

Oryza sativa L.<br />

Petroselinum crispum (Mill.) A. W. Hill<br />

Pisum sativum L.<br />

Populus tremula L.<br />

P. tremuloides Michx. (clone Esch5)<br />

Prosopis chilensis (Mol.) Stuntz<br />

P. juliflora (Sw.) DC.<br />

Quercus robur L. (clone oak DF 159)<br />

Setaria italica (L.) P. Beauv.<br />

Solanum melongena L.<br />

Sorghum vulgare Pers.<br />

Spilanthes calva DC.<br />

Tagetes erecta L.<br />

Tectona grandis L.<br />

Terminalia arjuna Wight & Arn.<br />

Tephrosia purpurea (L.) Pers.<br />

Vigna mungo (L.) Hepper<br />

V. radiata (L.) R. Wilczek<br />

Withania somnifera (L.) Dunal<br />

Zea mays L.<br />

Zizyphus nummularia Burm. fil.<br />

Data are based on the root colonization analysis in vivo and in vitro (cf.Varma et al.<br />

2001; Singh et al. 2003a, b)


244<br />

Giang Huong Pham et al.<br />

Fig. 6. Interactions with Zea mays and Setaria italica. The substratum was sterilized and<br />

filled into pots (1 kg). Fungal inoculum (1 % w/v) was thoroughly mixed with the soil.<br />

Plants were irrigated with tap water on alternate days to maintain about 70 % soil moisture.<br />

They were grown under greenhouse conditions maintained at 25±2 °C, 16 h<br />

light/8 h dark with fluorescent light intensity 1000 lux and relative humidity 70 %. P.<br />

indica promoted the growth of both monocots


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 245<br />

ties, which are characteristics of AM fungi (Singh et al. 2002a, b; Singh et al.<br />

2003a, b; Varma et al. 2001). It colonizes the root cortex in a variety of host<br />

<strong>plant</strong>s and improves their overall biomass production.<br />

4.1 Monocots<br />

Recently, we have selected the model <strong>plant</strong>s, Zea mays L. and Setaria italica<br />

(L.) Beauv. for in-depth studies. The roots were colonized and the growth of<br />

the <strong>plant</strong>s was highly promoted as a result of interaction with the fungus<br />

(Fig. 6). The phytopromotional influence was evident from early stages of the<br />

interaction.<br />

4.2 Legumes<br />

P. indica promotes the growth and survival of tissue culture raised-tropical<br />

legumes like Cicer arietinum L., Vigna radiata (L.) R. Wilczek, Pisum sativum<br />

L., Vigna mungo L. Hepper (Fig. 7) and Glycine max (L.) Merr. The fungal colonization<br />

resulted in 100 % survival of the in vitro raised <strong>plant</strong>s, whereas it<br />

was less than 50 % in uninoculated <strong>plant</strong>s.<br />

A dramatic increase in the <strong>plant</strong> growth was observed in C. arietinum and<br />

V. mungo as compared to their corresponding controls. The percent increase<br />

in <strong>plant</strong> height was 35.7 and 14.2 %, respectively, and the increase in fresh<br />

weight was 90 and 11 %, respectively, as compared to corresponding controls.<br />

Fig. 7. Interactions with Pisum sativum<br />

(left) and Vigna mungo (right). Surfacesterilized<br />

seeds of the legumes were<br />

germinated on water agar. Approximately<br />

3-cm-long young seedlings were<br />

placed on MS agar slants and incubated<br />

with P. indica (P). Control <strong>plant</strong>s (C)<br />

did not receive any fungus. Tubes were<br />

incubated at 25±2 °C and 1000 lux.<br />

Photographs were taken after 6 days.<br />

The fungus promoted the growth of<br />

both legumes


246<br />

Treated roots were colonized by the fungus and produced extramatrical, interand<br />

intracellular hyphae. Chlamydospores were observed at maturity.<br />

4.3 Orchids<br />

Giang Huong Pham et al.<br />

Seeds of Dactylorhiza purpurella (Steph’s.) Soó and D. majalis (Rchb. F.) Hunt<br />

and Summerh. were <strong>surface</strong>-sterilized and inoculated with P. indica (Fig. 8).<br />

After 2 weeks, seeds of D. purpurella started germination. After the appear-<br />

Fig. 8a–d. Interaction with Dactylorhiza majalis. Seeds of the orchid were <strong>surface</strong>-sterilized<br />

and germinated on oat agar. When some of the seeds started to swell, P. indica was<br />

added. The plates were incubated in the dark at room temperature. a Hyphae penetrating<br />

into the protocorm testa without traversing into the epidermis. b Hypha penetrating<br />

into a rhizoid (arrowhead), growing towards the protocorm and then entering into the<br />

cortex. c Semithin section of a peloton formed in a living cortical cell. d SEM picture of<br />

a peloton formed in a cortical cell, arrowhead pointing at starch grana (Blechert et al.<br />

1999; Varma et al. 2001)


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 247<br />

ance of the rhizoids the fungus penetrated into them, growing towards the<br />

protocorm. Inter- and intracellular hyphae spread from the basal swelling of<br />

the rhizoids and typical pelotons were formed in living cortical cells. Digestion<br />

of the pelotons started 17 days after inoculation. Differences in the<br />

growth of the protocorms inoculated with P. indica and the corresponding<br />

controls were obvious after the intracellular interactions began. P. indica was<br />

found to be a typical orchid mycorrhizal fungus in vitro, promoting growth in<br />

all tested Dactylorhiza species (Blechert et al.1999; Singh et al. 2000, 2002b;<br />

Varma et al. 2001).<br />

4.4 Medicinal Plants<br />

The tissue culture-raised <strong>plant</strong>s and seedlings from <strong>surface</strong>-sterilized seeds<br />

of medicinal <strong>plant</strong>s, like Spilanthes calva DC, Withania somnifera (L.) Dunal,<br />

Bacopa monnieria (L.) Wett., Adhatoda vasica L., Azadirachta indica A.Juss.<br />

(neem), Artemisia annua L., Chlorophytum tuberosum Baker, C. borivilianum<br />

Santapau & R. R. Fernandez (musli), and Termnalia arjuna L. were inoculated<br />

with P. indica in mist chambers and nurseries before being transferred<br />

to the field (Fig. 9).<br />

Significant increases in growth and yield of the <strong>plant</strong> species were recorded<br />

relative to uninoculated controls. Shoot and root length, biomass, basal stem,<br />

leaf area, overall size, inflorescence number, flower and seed production were<br />

all enhanced in the presence of the fungus. Net primary productivity was also<br />

higher than in control <strong>plant</strong>s. The results clearly indicate the commercial<br />

potential of P. indica for large-scale cultivation of medicinal <strong>plant</strong>s. The differences<br />

in growth observed may have been caused by a greater absorption of<br />

water and mineral nutrients due to extensive colonization of roots and the<br />

proliferation of the mycelium into the soil.<br />

In another pot trial experiment, neem seedlings were inoculated with Glomus<br />

mosseae, Scutellospora gilmorei, and P. indica. The treatment was conducted<br />

using pots with sterile and natural soils. Plant growth of P. indicatreated<br />

<strong>plant</strong>s was found to be drastically improved compared to those <strong>plant</strong>s<br />

treated with AM fungi and controls. P. indica-treated <strong>plant</strong>s attained maximum<br />

height, healthier foliage and a well developed subterranean.<br />

Bacopa monnieria (L.) Wett. is considered to be important because the<br />

whole <strong>plant</strong> has medicinal value. P. indica colonizes the roots of tissue culture-raised<br />

<strong>plant</strong>s and promoted the overall <strong>plant</strong> biomass. A biological<br />

hardening rendered almost 100 % survival on transfer from the laboratory to<br />

the field.<br />

S. calva and W. somnifera were treated with P. indica in a field trial. A pronounced<br />

growth response following the P. indica inoculation was observed.<br />

The basal stem and leaf areas of treated <strong>plant</strong>s were enhanced. Interestingly,<br />

large kidney-shaped inflorescences were observed on inoculated S. calva


248<br />

Giang Huong Pham et al.<br />

Fig. 9a–f. Interactions with medical <strong>plant</strong>s. P. indica was used for the inoculation of<br />

young seedlings of a Adhatoda vasica, b Azadirachta indica, c Terminalia arjuna, d Spilanthus<br />

calva, e Withania somnifera and f Chlorophytum borivillianum growing in pots.<br />

After the establishment of the interaction, the latter three <strong>plant</strong> species were tranferred<br />

to the field where the pictures were taken. In a–c, e, the control is on the left, inoculated<br />

<strong>plant</strong>s are on the right. In all experiments growth and flowering of the treated <strong>plant</strong>s<br />

were obviously promoted


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 249<br />

<strong>plant</strong>s, however, these kidney-shaped inflorescences were never observed in<br />

control <strong>plant</strong>s. The length of the inflorescence and the number of flowers on<br />

inoculated S. calva <strong>plant</strong>s also increased compared to the controls. Similarly,<br />

the number of flowers of the inoculated W. somnifera was higher than in the<br />

controls.For both inoculated medicinal <strong>plant</strong> species,the number of seeds was<br />

higher than for the controls (Rai et al.2001),and the overall root biomass of the<br />

inoculated <strong>plant</strong>s was higher than that of the corresponding controls.The fresh<br />

and dry weights of both underground and aboveground parts of S.calva and W.<br />

somnifera-inoculated <strong>plant</strong>s were higher than in the controls.<br />

4.5 Economically Important Plants<br />

Plants of economic importance tested in vivo and in vitro were Tagetes erecta<br />

L. (marigold), Nicotiana tabaccum L. (tobacco), Lycopersicon esculentum Mill.<br />

(tomato) and Solanum melongena L. (bringal). In a pot trial marigold inoculated<br />

with P. indica showed healthier <strong>plant</strong>s with early bud formation and<br />

enlarged flowers compared to the control (Fig. 10).<br />

Hypocotyl of germinated seeds of tobacco were taken as an ex<strong>plant</strong> for callus<br />

development. Callusing regeneration of the shoot was established on MS<br />

medium (Murashige and Skoog 1962). Biological hardening of the regenerated<br />

<strong>plant</strong>lets with P. indica recorded the maximum capacity for regaining the<br />

tensile strength of the stem (Fig. 11). Plants treated with G. mosseae possessed<br />

less tensile strength, but more than the control <strong>plant</strong>lets.<br />

Early root induction was recorded in the bringal root organ culture interacting<br />

with P. indica (Tables 2, 3). This study indicated that P. indica is a potent<br />

Fig. 10. Interaction with Tagetes erecta.<br />

The experiment was conducted as<br />

described in Fig. 6. P. indica-inoculated<br />

<strong>plant</strong>s (P) were extensive in growth and<br />

had bigger flowers compared to the<br />

control (C)


250<br />

Giang Huong Pham et al.<br />

Fig. 11a–d. Interaction with tissue culture-raised Nicotiana tabacum. Surface-sterilized<br />

seeds of tobacco were germinated on 1/2 strength MS medium. Fifteen days after germination,<br />

hypocotyl was transferred for callus formation on MS media supplemented with<br />

NAA 2 mg/l and BAP 0.5 mg/l. Cultures were grown in a controlled tissue culture laboratory.<br />

Mycelia of P. indica were grown in culture bottles on minimal medium. Regenerated<br />

shoots were transferred to these bottles. Observations were made after 15 days of treatment;<br />

root fragments were stained with Trypan blue.After 3 weeks the <strong>plant</strong>s were transferred<br />

to pots with sterile substratum and grown in a mist chamber. a Massive callus formation<br />

on 1/2 MS medium of the P. indica-treated <strong>plant</strong>s (right) compared to the control<br />

(left). b Root fragment of inoculated tobacco <strong>plant</strong>let colonized by the fungus. c Differentiation<br />

of the callus cultures after 15 days of inoculation on regeneration medium; left<br />

control, right treated <strong>plant</strong>let. d Tobacco <strong>plant</strong>s after 8 weeks in a mist chamber, left control,<br />

right treated <strong>plant</strong>


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 251<br />

<strong>plant</strong> growth-promoting fungus. It is not only the mycelium in association<br />

with the root which exerts this effect, fungus culture filtrate containing fungal<br />

exudates (these may be hormones, proteins, enzymes, polyamines,<br />

amino acids, etc.) also exhibited almost the same effect.<br />

P. indica-inoculated tomato seedlings grown in glass tubes on MS<br />

medium were about three times higher than the control and proliferate root<br />

biomass was observed. Soon after inoculation adventitious roots appeared<br />

high above the <strong>surface</strong> of the substrate. As a result of prolonged incubation,<br />

the subapical region of the shoot became chlorotic and finally the leaves<br />

wilted, probably due to the restricted nutrition. However, adventitious roots<br />

started developing again from the apex, thus maintaining a green tip.<br />

Table 2. Morphological features of tobacco <strong>plant</strong>lets after treatment with different<br />

mycobionts (biological hardening)<br />

Mycobiont 2 Weeks 4 Weeks<br />

Control Leaves lost turgor pressure Some leaves turned yellow, stem<br />

and stem the tensile strength turned brown<br />

G. mosseae Leaves lost turgor pressure Some leaves dried, stem regained<br />

and stem the tensile strength tensile strength<br />

P. indica Leaves lost turgor pressure and Plants were healthy, stem<br />

stem the tensile strength regained tensile strength<br />

cf. Sahay and Varma (1999)<br />

Morphological features of micropropagated tobacco <strong>plant</strong>lets subjected to biological<br />

hardening with Glomus mosseae and Piriformospora indica. Soil substrata were sterilized<br />

soil:sand mixture (3:1). Inocula (spores, hyphae, colonized root pieces, etc.) were<br />

included at 1 % (w/v) to each pot (10¥6 cm)<br />

Table 3. Plant biomass and percent-colonization as a result of interaction of the<br />

<strong>plant</strong>lets with mycobionts (biological hardening)<br />

Mycobionts Fresh weight (g/<strong>plant</strong>) Colonization (%)<br />

Control 2.18±0.199 nd<br />

Glomus mosseae 2.69±0.145 64±15.16<br />

Piriformospora indica 3.08±0.266 76±25.10<br />

Plants were harvested and the total fresh weight was recorded. Fungal root colonization<br />

was estimated after staining with Trypan blue. RM ANOVA ON RANKS test shows<br />

c 2 =8.40 with 3 degrees of freedom. P (est) =0.0384, P (exact) =0.0190. The differences in the<br />

median value among the treatment groups are greater than would be expected by<br />

chance, i.e., there is a statistically significant difference (P=0.0190). Data represent mean<br />

±SD, nd, not detected


252<br />

Giang Huong Pham et al.<br />

4.6 Timber Plants<br />

Young seedlings of Populus tremula L., Quercus robur L. and Dalbergia sissoo<br />

Roxb. ex DC. were tested in the green house and followed by field trials. In a<br />

pot culture experiment, P. indica-inoculated <strong>plant</strong>s of Po. tremula were<br />

strongly promoted according to their biomass production compared to the<br />

controls (Fig. 12). Qu. robur (clone oak DF 159) was micro-propagated and<br />

rooted as described by Herrmann et al. (1998). P. indica was pre-cultivated for<br />

7 days on Aspergillus medium (Kaefer 1977). Co-culture was performed on<br />

MMN 1/10 medium (Marx 1969) in Petri dishes. The oak shoots grew out of<br />

the dish and to avoid rapid wilting, the inoculated dishes were grown in large<br />

Petri dishes (radius of 145 mm) with moistened absorbent paper sheets. Cocultivation<br />

was carried out at 25 °C and a photoperiod of 16 h illumination<br />

(97 W m –2 ; OSRAM L 115 W/20SA cool white) for 10 weeks. At the end of the<br />

experiment, the roots were colonized by the fungus (Fig. 12).<br />

Precedent results (Herrmann et al. 1998) showed that an ectomycorrhizal<br />

fungus Piloderma croceum Erikss. & Hjortst. was able to enhance <strong>plant</strong><br />

growth of oaks before any mycorrhizal formation occurred. Hence, P. indica<br />

and P. croceum were able to enhance root development. In further investigations<br />

it would be of interest to compare the effects of both fungi separately<br />

and in combination on root initiation and elongation, and analyse in which<br />

Fig. 12. Interactions with ectomycorrhizal <strong>plant</strong>s. Left Seedlings of Populus tremula<br />

(obtained from Köln, Germany) were treated with P. indica. The experimental design<br />

was the same as described in Fig. 6. The larger <strong>plant</strong> was treated with the fungus, the<br />

smaller <strong>plant</strong> without fungus. Right Quercus robur (clone oak DF 159) seedlings were<br />

treated with P. indica under laboratory conditions. It was micro-propagated and rooted<br />

as described by Herrmann et al. (1998). P. indica was pre-cultivated for 7 days on<br />

Aspergillus medium. Co-culture was performed on 1/10 MMN medium in Petri dishes.<br />

Co-cultivation was conducted at 25 °C and a photoperiod of 16 h illumination (97 W/m)<br />

over a duration of 10 weeks. Picture shows hyphae and spores formed within and around<br />

the roots


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 253<br />

manner diffusing substances may be involved in the phenomena. A similar<br />

positive response was recorded for P. tremula and D. sissoo.<br />

4.7 Unexpected Interactions with Wild-Type and Genetically Modified<br />

Populus Plants<br />

Hybrid aspen Populus tremula x P. tremuloides Michx. (clone Esch5, kindly<br />

supplied by Dr. M. Fladung, Federal Research Centre for Forestry and Forest<br />

Products, Grosshansdorf, Germany) were cultivated in vitro at 25 °C in permanent<br />

light. Four <strong>plant</strong>s were grown in each Magent GA7 vessel containing<br />

80 ml of WPM medium (see Chap. 30). When 2-cm cuttings of the shoot<br />

including the tip were transferred to fresh medium, rooting was initiated at<br />

the cutting site in most trans<strong>plant</strong>s after 5–6 days. When Populus was inoculated<br />

with P. indica after rooting of trans<strong>plant</strong>s had commenced, stimulation<br />

in the root growth was observed after 5 days. A clear stimulation of root<br />

branching and an apparent increase in root length were observed (Fig. 13).<br />

This result confirms the observations made for other <strong>plant</strong>s. No significant<br />

changes were observed in <strong>plant</strong> shoot growth.<br />

In a second experiment, the fungus was allowed to grow in the WPM<br />

medium 1 week prior to the Populus trans<strong>plant</strong>s. Interestingly, <strong>plant</strong> growth<br />

and rooting pattern changed under these conditions. The salient changes<br />

recorded were:<br />

– inhibition of root formation at the cutting site where rooting normally<br />

occurs without inoculation with fungus.<br />

– aerial root formation was induced.<br />

– deformations occurred in aerial roots when they came into contact with<br />

the <strong>surface</strong> of the fungus-inoculated medium, and they failed to grow into<br />

the medium.<br />

Shoot growth of inoculated <strong>plant</strong>s was suppressed compared to the control.<br />

After 6 weeks of cultivation, a profuse fungal mat appeared on the <strong>surface</strong> of<br />

the medium, however, <strong>plant</strong>s were not killed. Observed under the light microscope,<br />

fungal infection was not detected in aerial roots.<br />

We speculated that one or more chemical compounds were produced by<br />

the mycelium which were responsible for the changes mentioned above. To<br />

prepare a crude extract, <strong>plant</strong> material was removed from the cultures. The<br />

remaining medium (with or without fungus) was autoclaved for 30 min at<br />

121 °C. These extracts were mixed with the same volume of double strength<br />

WPM medium and filled into culture vessels. After solidification, four trans<strong>plant</strong>s<br />

without roots were transferred into these media. After 5 days in the<br />

medium prepared with the <strong>plant</strong> extract, the rooting was initiated at the cutting<br />

site. In contrast, the <strong>plant</strong>s incubated with the extract prepared from both<br />

types of media did not form any roots (neither in the medium nor in the air).


254<br />

Giang Huong Pham et al.<br />

Fig. 13a–d. Interaction with in vitro grown Populus clone Ech5 on WPM. Ex<strong>plant</strong>s with<br />

a length of 2 cm, including one terminal and 5–6 side buds were transferred to Magenta<br />

GA7 vessels containing 80 ml of WPM medium. a Top view of a noninoculated control<br />

after 4 weeks of cultivation at 25 °C in constant light; b four discs of P. indica culture<br />

(snowflake) were placed onto the medium and incubated for 7 days at 25 °C prior to the<br />

introduction of the <strong>plant</strong> cuttings; c normal rooting started at the cutting site (arrow)<br />

after 1 week. The photo shows the typical rooting pattern after 4 weeks; d rooting pattern<br />

of an inoculated <strong>plant</strong>let after 4 weeks of co-cultivation with P. indica; rooting at the cutting<br />

site was completely blocked (arrow). Instead, aerial rooting appeared above the<br />

medium, ca. 1 cm apart from the cutting site (arrow). Root tips, when in contact with the<br />

medium, showed a modified morphology (see circles). Inset shows a magnified view of<br />

the modified root tips


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 255<br />

Water extracts of fungus and <strong>plant</strong>-grown medium prepared at room temperature<br />

had no influence on rooting.<br />

4.8 Non Mycorrhizal Plants<br />

Most species of the <strong>plant</strong>s are normally infected by mycorrhizal fungi, but<br />

some <strong>plant</strong> taxa do not usually form generally recognizable mycorrhizas<br />

(Tester et al. 1987). Members of, e.g., Brassicaceae, Chenopodiaceae and Amaranthaceae<br />

(Read 1999; Singh et al. 2003a, b; Varma 1998, 1999; Varma et al.<br />

2001), belong to this exceptional group of nonmycorrhizal <strong>plant</strong>s. The mechanism<br />

which determines the nonhost nature of <strong>plant</strong> species preventing the<br />

establishment of a functional symbiosis is not known. Present knowledge of<br />

the sequence of fungal development leading to the establishment of functional<br />

mycorrhiza suggests that the nonhost nature of <strong>plant</strong>s lies in their<br />

inability to trigger expression of fungal genes involved in hyphal commitment<br />

to the symbiotic status.<br />

It would be useful to assess these <strong>plant</strong>s with respect to their interaction<br />

with P. indica. In vitro studies with P. indica and S. vermifera recorded that<br />

these two symbiotic fungi profusely interacted with the root system of the<br />

crucifer <strong>plant</strong>s, Brassica juncea (L.) Czern. et Coss. (mustard), Brassica oleracea<br />

var. capitataL. (cabbage), and the Chenopodiaceae Spinacia oleracea L.<br />

(spinach). Although some of these <strong>plant</strong>s were said to be able to form AM<br />

(Tester et al. 1987), no mycorrhizal interactions with Glomus mosseae were<br />

found in the pot trials we conducted. Instead, all the <strong>plant</strong>s inoculated with P.<br />

indica were colonized by the fungus and recorded phytopromotional effects<br />

in comparison to the control. However, a high degree of variation was<br />

recorded in with respect to biomass and their length. Cabbage responded<br />

most positive with P. indica. Different results were recorded in root systems.<br />

In cabbage, the fungi profusely colonized inter- and intracellularly the root<br />

cortex cells. Colonization in mustard was less in comparison to cabbage and<br />

followed by spinach.<br />

Further experiments indicated that P. indica did not invade the root of myc –<br />

mutants of pea (Pisum sativum L.) and soyabean (Glycine max (L.) Merr.).<br />

When the fungus was confronted with these mutants, <strong>plant</strong> growth was suppressed<br />

and the fungal morphology was severely affected. The sporulation of<br />

P.indicainteracting with wild types was homogenous, while it was heterogeneous<br />

in myc – mutants (Fig. 14). During the co-cultivation the mycelia turned<br />

brown and produced a copious amount of mucilage.


256<br />

Giang Huong Pham et al.<br />

Fig. 14. Interaction with myc – mutant of Pisum sativum. Wild-type and myc – mutant of<br />

pea were inoculated with P. indica. Growth conditions were as described in Fig. 7. Left<br />

SEM-picture of the <strong>surface</strong> of colonized roots (wild-type) and mycelial mats of P. indica<br />

showing dense masses of well differentiated chlamydospores. Right SEM-picture of the<br />

root <strong>surface</strong> and the mycelial mat after interaction with myc – mutant and 14 days of<br />

incubation with a low amount of morphologically heterogeneous chlamydospores<br />

4.9 Arabidopsis thaliana<br />

A. thaliana (L.) Heynh. seedlings were pre-germinated for 2 weeks on MS and<br />

then transferred to Aspergillus medium. Plants were further cultivated with<br />

or without the fungus P. indica at 22 °C and under short day conditions. Control<br />

<strong>plant</strong>s (without fungus) remained small with limited root growth and<br />

branching. In contrast, the co-cultivation with P. indica resulted in promotion<br />

of the <strong>plant</strong> growth and extensive root proliferation and elongation.An observation<br />

of the inoculated roots under the light microscope revealed that the<br />

fungus colonized the root <strong>surface</strong> and the cortical zone. Chlamydospores were<br />

produced by external hyphae (extramatrical) and within the root cortex and<br />

root hairs (intracellular). The fungal colonization reduced the root hair formation<br />

(Fig. 15).<br />

In another independent study, A. thaliana was cultivated on MYP-agar.A 1month-old<br />

culture was flooded with 10 ml sterilized water to gain a chlamydospore<br />

suspension for inoculation. Three-day-old A. thaliana seedlings were<br />

inoculated each with 10 ml of chlamydospore suspension. After 5, 10, 17, and<br />

31 days of co-cultivation, <strong>plant</strong>s were harvested and the roots examined with<br />

the light microscope and SEM. Moreover, FDA (fluoresceindiacetate) was used<br />

to discriminate between living and dead cells. At the latest, 17 days after inoculation,<br />

the whole root <strong>surface</strong> of A. thaliana was covered with mycelium.<br />

Most of the hyphae were growing between root hairs and some were closely<br />

attached to the rhizodermis. Several of these closely attached hyphae were following<br />

the anticlinal, axial cell walls of the rhizodermal cells. Keijer (1996)<br />

found that this is an indication of the beginning of an interaction between<br />

Rhizoctonia solani and its hosts.


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 257<br />

Fig. 15a–d. Interaction with Arabidopsis thaliana. Seedlings were germinated for<br />

2 weeks on MS and then transferred to Aspergillus medium. Plants were cultivated at<br />

22 °C and under short day conditions. a Control <strong>plant</strong>s remained small with limited root<br />

growth and branching; b in contrast, the co-cultivaton with P. indica resulted in the promotion<br />

of <strong>plant</strong> growth and extensive root proliferation; c control roots produced a large<br />

number of root hairs growing uniformly from the top to the base; d inoculated roots as<br />

seen under the light microscope after staining with cotton blue: P. indica colonized the<br />

root <strong>surface</strong> and the cortical zone, spores were produced by external hyphae and in the<br />

roots and root hairs (inset)


258<br />

Giang Huong Pham et al.<br />

Fig. 16a–e. Interaction with Arabidopsis thaliana. a SEM picture of hyphae closely<br />

attached to the root <strong>surface</strong>. Besides normal hyphae (arrowhead) P. indica also forms<br />

coralloid hyphae (arrow) and chlamydospores (asterisk, collapsed due to preparation);<br />

b SEM picture of a hypha probably forming an appressorial swelling (arrow), which has<br />

caused an imprint (arrowhead) on the <strong>surface</strong> of a rhizodermal cell; c epifluorescence<br />

LM-picture of a hypha entering a root hair (arrow). Staining: aniline blue; d root stained<br />

with cotton blue shows intracellular chlamydospores in the rhizodermis (arrows); e a<br />

segment of the root stained with FDA and observed in epifluorescence showing lower<br />

FDA-fluorescence (arrowhead) than adjacent regions, indicating less vitality. In bright<br />

field it was clearly visible that this region was covered with hyphae (arrow chlamydospore)


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 259<br />

From 10 days after inoculation on, some hyphae formed irregular, globular<br />

swellings, which were called coralloid hyphae (Fig. 16). These hyphae preceded<br />

the production of chlamydospores, which were produced terminally.<br />

Mature spores could be found from 17 days after inoculation on. On the <strong>surface</strong><br />

of rhizodermal cells occasionally slightly swollen hyphal tips could be<br />

observed. In some cases an imprint in the host cell wall was visible, probably<br />

caused by mechanical pressure in combination with enzymes, indicating an<br />

appressorial function of these hyphal tips (Fig. 16). In some host cells intracellular<br />

hyphae could be found and at the penetration point, the hyphae had<br />

generally reduced their diameter. Host cell wall thickenings as an answer to<br />

the penetration could never be observed. Like hyphae of the external<br />

mycelium, intracellular hyphae formed coralloid swellings and sometimes<br />

also produced chlamydospores. A necrotrophic potential of P. indica could be<br />

demonstrated by vitality tests. Control roots and regions, which were free<br />

from mycelium were always stained bright green, which indicated the vitality<br />

of the corresponding cells. Root areas that were covered with hyphae often<br />

showed weaker or no fluorescence (Fig. 16). This indicates that the fungus is<br />

able to cause local damage to the root cortex of this host.<br />

4.10 Root Organ Culture<br />

Root organ culture of Daucus carota L. (carrot) was prepared as described by<br />

Bécard and Piche (1992). P. indica interacted with the root organ culture of<br />

carrot in the same way as was found in other <strong>plant</strong>s tested. The infection rate,<br />

as a portion of infected root length, has been calculated to be 17 % 9 weeks<br />

after inoculation, 50 % in the most successful culture and 40 % after pro-<br />

Fig. 17a–c. Interaction with transformed Daucus carota (Queen Anne’s-lace) root. Root<br />

organ cultures were inoculated with P. indica and grown for 20 days. a Dark circles represent<br />

the place for inocula; b hyphae and chlamydospores on the <strong>surface</strong> of the roots; c<br />

intracellular sporulation as seen with the LM


260<br />

Giang Huong Pham et al.<br />

longed incubation. Newly developed lateral roots were preferred infection<br />

sites and most infections started 0.5–1 cm behind the root tips. Bécard and<br />

Fortin (1988) observed a similar pattern for AMF. The preferential site for primary<br />

infection by the germ tubes of germinating AM spores was the elongation<br />

zone of the main root, where lateral root primordia formed. (Fig. 17).<br />

5 Cell Wall Degrading Enzymes<br />

The exo-oxidative enzyme laccase has been detected in a large number of<br />

basidiomycete ectomycorrhizal fungi, in a few ectomycorrhizal ascomycetes<br />

and only in one endomycorrhizal species (Gramss et al. 1998). All the<br />

ascomycetes tested showed the presence of laccase, but in basidiomycetes only<br />

33 out of 44 species were found to be active (Table 4). However, to our knowl-<br />

Table 4. Laccase activity in mycorrhizal fungi<br />

Fungal species Systematic positions Laccase References<br />

activities<br />

Ectomycorrhizal<br />

basidiomycetes<br />

Amanita gemmata Agaricales, Amanitaceae (+) Gramss et al. (1998)<br />

Amanita muscaria Agaricales, Amanitaceae (+) Gramss et al. (1998)<br />

Amanita rubescens Agaricales, Amanitaceae (+) Gramss et al. (1998)<br />

Amanita spissa Agaricales, Amanitaceae (+) Gramss et al. (1998)<br />

Amanita strobiliformis Agaricales, Amanitaceae (+) Gramss et al. (1998)<br />

Boletinus cavipes Boletales, Gyrodontaceae (–) Gramss et al. (1998)<br />

Boletus edulis Boletales, Boletaceae (–) Gramss et al. (1998)<br />

Boletus erythropus Boletales, Boletaceae (–) Gramss et al. (1998)<br />

Boletus luridus Boletales, Boletaceae (–) Gramss et al. (1998)<br />

Boletus piperatus Boletales, Boletaceae (–) Gramss et al. (1998)<br />

Cortinarius varius Agaricales, Cortinariaceae (+) Gramss et al. (1998)<br />

Hebeloma crustuliniforme Agaricales, Cortinariaceae (+) Gramss et al. (1998)<br />

Hebeloma edurum Agaricales, Cortinariaceae (–) Gramss et al. (1998)<br />

Hebeloma hiemale Agaricales, Cortinariaceae (+) Gramss et al. (1998)<br />

Hebeloma sinapizans Agaricales, Cortinariaceae (+) Gramss et al. (1998)<br />

Laccaria amethystina Agaricales, Tricholomataceae (+) Muenzenberger et<br />

al. (1997)<br />

Lactarius deliciosus Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Lactarius deterrimus Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Lactarius necator Agaricales, Russulaceae (–) Gramss et al. (1998)<br />

Lactarius rufus Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Lactarius torminosus Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Leccinum scabrum Boletales, Boletaceae (+) Gramss et al. (1998)<br />

Leccinum versipelle Boletales, Boletaceae (+) Gramss et al. (1998)<br />

Paxillus involutus Boletales, Paxillaceae (+) Gramss et al. (1999)


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 261<br />

edge, the presence of laccase genes in these fungi was shown only in Lactarius<br />

rufus (Chen et al. 2003). Most fungi tested showed a strong laccase activity. P.<br />

indica and S. vermifera ss. Warcup and Talbot also showed a positive reaction<br />

to the ABTS test (oxidation of 2,2¢- azino-bis (3-ethylthiazoline-6-sulfonate),<br />

i.e., presence of laccase activity (Fig. 18). However, the reaction was faster in<br />

the former fungus than in the latter. Laccase activity was observed in the very<br />

young culture (5 days old), whereas in S. vermifera the reaction appeared<br />

stronger after 10–12 days.<br />

Other cell wall-degrading enzymes detected in P. indica are given in<br />

Table 5.All major cell wall degrading enzymes were present in P. indica except<br />

monoxygenase and phenoloxidase. Table 6 shows the activities of CMC-ase,<br />

xylanase and polygalacturonase in the <strong>plant</strong>s incubated with P. indica. In the<br />

case of polygalacturonase, the enzyme activity was higher at the initial stage<br />

Table 4. (Continued)<br />

Fungal species Systematic positions Laccase References<br />

activities<br />

Pisolithus tinctorius Boletales, Sclerodermataceae (–) Gramss et al. (1998)<br />

Russula aeruginea Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Russula foetens Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Russula violeipes Agaricales, Russulaceae (+) Gramss et al. (1998)<br />

Scleroderma citrinum Boletales, Sclerodermataceae (–) Gramss et al. (1998)<br />

Suillus aeruginascens Boletales, Boletaceae (+) Gramss et al. (1998)<br />

Suillus granulatus Boletales, Boletaceae (low) Gramss et al. (1998)<br />

Suillus grevillei Boletales, Boletaceae (+) Gramss et al. (1998)<br />

Suillus luteus Boletales, Boletaceae (+) Gramss et al. (1998)<br />

Suillus variegatus Boletales, Boletaceae (low) Gramss et al. (1998)<br />

Tricholoma fulvum Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Tricholoma imbricatum Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Tricholoma lascivum Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Tricholoma scalpturatum Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Tricholoma subannulatum Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Tricholoma terreum Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Tricholoma ustaloides Agaricales, Tricholomataceae (+) Gramss et al. (1998)<br />

Xerocomus badius Boletales, Boletaceae (low) Gramss et al. (1999)<br />

Xerocomus chrysenteron Boletales, Boletaceae (–) Gramss et al. (1998)<br />

Xerocomus subtomentosus Boletales, Boletaceae (–) Gramss et al. (1998)<br />

Ectomycorrhizal ascomycetes<br />

Morchella conica Pezizales (+) Gramss et al. 1998<br />

Morchella elata Pezizales (+) Gramss et al. 1998<br />

Morchella esculenta Pezizales (+) Gramss et al. 1998<br />

Tuber sp.<br />

Endomycorrhizal fungi<br />

Pezizales (+) Miranda et al. 1992<br />

Glomus etunicatus Glomales (+) Nemec, 1981


262<br />

Giang Huong Pham et al.<br />

Fig. 18. Laccase activity in P. indica. The fungus was grown for 5 days on Aspergillus<br />

medium, then a small hole (arrow) was cut into the agar at the border of the growing<br />

mycelial mat. Three drops (30 ml) of ABTS were added. Green coloration appeared and<br />

was monitored at different time intervals. The evolution of the coloration is given with<br />

time, but the coloration was immediately obtained after a few seconds. Snowflake indicates<br />

the fungal growth<br />

Table 5. Cell wall degrading enzymes from P. indica<br />

Compound Detection Possible enzymes Important for<br />

the degradation of<br />

ABTS + Laccase Lignin<br />

Ferulic acid + Ferulase Lignin<br />

Vanillin – Monoxygenase Lignin<br />

Tannin – Phenoloxidase Phenols<br />

Starch + Amylase Plant storage polysaccharides<br />

Cellulose + Cellulase Cellulose<br />

Gelatine + Protease Proteins<br />

Pectin + Pectinase Pectin<br />

Lipid + Lipase Fat<br />

Xylan + Xylanase Hemicellulose<br />

Chitin + Chitinase Chitin<br />

cf. Bütehorn (1999) and Varma et al. (2001)<br />

(after 20 min) of incubation, i.e., up to 0.058 mmol ml –1 min –1 , and declined to<br />

0.041 mmol ml –1 min –1 after 80 min of incubation.<br />

Fungal hyphae entered the cells randomly through the cell wall. It seems<br />

the entry was facilitated by the combined action of wall degrading enzymes<br />

and mechanical pressure.<br />

Under normal conditions AMF do not invade vascular systems and the aerial<br />

parts of their hosts. Despite heavy root colonization this is also true for P.<br />

indica and S. vermifera, although these fungi were able to produce heavy<br />

amounts of cell wall-degrading enzymes.


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 263<br />

6 Conclusions<br />

Table 6. Extracellular hydrolytic enzymes produced in vitro<br />

by P. indica<br />

Piriformospora indica is a wide host range organism. It interacts with <strong>plant</strong><br />

growth-promoting rhizobacteria (PGPRs), terrestrial mycobionts, green<br />

algae, lower and higher <strong>plant</strong>s. Among the PGPRs, Pseudomonas fluorescence<br />

inhibited the fungus, while strains of Azotobacter, Bradyrhizobium and<br />

Azospirillum over all enhanced the fungal growth. In vitro and in vivo studies<br />

as well as field trials have proved phytopromotional effects on most <strong>plant</strong>s<br />

tested. Exceptions were myc – mutants of pea and soyabean, where the hyphae<br />

did not invade and the <strong>plant</strong> growth was negatively influenced. Results<br />

obtained from the interaction with Arabidopsis thaliana and Hybrid aspen<br />

(Populus tremula x P. tremuloides) were interesting as they opened new vistas<br />

to understand the mechanism and molecular basis of <strong>plant</strong>-fungus symbiosis.<br />

There are still lots of unanswered questions: how does the fungus promote<br />

the growth of the <strong>plant</strong>s and why is the growth of nonhosts reduced? what is<br />

the mechanism of root pathogen suppression? these are only two examples of<br />

such questions to be answered in the future.<br />

Acknowledgments. The Indian authors are thankful to DBT, DST, CSIR, UGC, and the<br />

Government of India for partial financial assistance. We are thankful to Dr. Michael<br />

Weiss, Germany for providing 28 s rDNA analysis of P. indica.<br />

IU a<br />

CMCase 0.013<br />

Xylanase 0.062<br />

Polygalacturonase 0.017<br />

cf.Varma et al. (2001)<br />

a 0.5 % Na-polypectate (Sigma) was used as the substrate.<br />

One unit activity (IU) of Pgase is defined as the amount of<br />

enzyme which releases 1 mol of carboxyl group as equivalent<br />

to the amount of Na-thiosulphate added to neutralize<br />

the residual iodine. Polymethylgalacturonase (PMG) was<br />

completely absent


264<br />

Giang Huong Pham et al.<br />

References and Selected Reading<br />

Bécard G, Fortin JA (1988) Early events of vesicular-arbuscular mycorrhiza formation on<br />

Ri T-DNA transformed roots. New Phytol 108:211–218<br />

Bécard G, Piche Y (1992) Establishment of vesicular – arbuscular mycorrhiza in root<br />

organ culture: Review and proposed methodology. Methods Microbiol 24:89–108<br />

Blechert O, Kost G, Hassel A, Rexer R-H, Varma A (1999) First remarks on the symbiotic<br />

interactions between Piriformospora indica and terrestrial orchids. In: Varma A,<br />

Hock B (eds) Mycorrhizae 2nd edn. Springer, Berlin Heidelberg New York, pp 683–688<br />

Bütehorn B (1999) Erste Zytologische und molekulare Untersuchungen zu Piriformospora<br />

indica, einem pflanzenwachstumsfördernden Endophyten. PhD Thesis, Marburg,<br />

Germany<br />

Chen DM, Bastias BA, Taylor AFS, Cairney JWG (2003) Identification of laccase-like<br />

genes in ectomycorrhizal basidiomycetes and transcriptional regulation by nitrogen<br />

in Piloderma byssinum. New Phythol 157:547–554<br />

Dehne HW (1982) Interaction between VAM fungi and <strong>plant</strong> pathogens. Phytopathology<br />

72:1115–1119<br />

Gramss G, Kirsche B, Voigt K-D, Günther T, Fritsche W (1998) Conversion rates of five<br />

polycyclic aromatic hydrocarbons in liquid cultures of fifty-eight fungi and the concomitant<br />

production of oxidative enzymes. Mycol Res 103:1009–1018<br />

Gramss G, Günther T, Fritsche W (1999) Spot tests for oxidative enzymes in ectomycorrhizal,<br />

wood-, and litter decaying fungi. Mycol Res 102:67–72<br />

Herrmann S, Munch J-C, Buscot F (1998) A gnotobiotic system with oak micro-cuttings<br />

to study specific effects of mycobionts on <strong>plant</strong> morphology before, and in the early<br />

phase of ectomycorrhiza formation by Paxillus involutus and Piloderma croceum.<br />

New Phytol 138:203–212<br />

Kaefer E (1977) Meiotic and mitotic recombination in Aspergillus and its chromosomal<br />

aberrations. Adv Genet 19:33–131<br />

Keijer J (1996) The initial steps of the interaction process in Rhizoctonia solani.In:Sneh<br />

B, Jabaji-Hare S, Neate S, Dijst G (eds) Rhizoctonia species: taxonomy, molecular biology,<br />

ecology, pathology and disease control. Kluwer, Dordrecht, pp 149–162<br />

Marx DH (1969) The influence of ectotrophic mycorrhizal fungi on the resistance of pine<br />

roots to pathogenic infections. I.Antagonism of mycorrhizal fungi to root pathogenic<br />

fungi and soil bacteria. Phytopathology 59:153–163<br />

Miranda M, Bonfigli A, Zarivi O, Ragnelli AM, Pacioni G, Botti D (1992) Truffle tyrosinase:<br />

properties and activity. Plant Sci 81:175–182<br />

Münzenberger B, Otter T, Wustrich D, Polle A (1997) Peroxidase and laccase activities in<br />

mycorrhizal and non-mycorrhizal fine roots of Norway spruce (Picea abies) and larch<br />

(Larix decidua). Can J Bot 75:932–938<br />

Murashige T, Skoog F (1962) A revised medium for rapid growth and bioassays with<br />

tobacco tissue cultures. Physiol Plant 15:473–497<br />

Nemec S (1981) Histochemical characteristics of Glomus etunicatus infection on Citrus<br />

limon fibrous roots. Can J Bot 59:609–617<br />

Rai M, Acharya D, Singh A, Varma A (2001) Positive growth responses of the medicinal<br />

<strong>plant</strong>s Spilanthes calva and Withania somnifera to inoculation by Piriformospora<br />

indica in a field trial. Mycorrhiza 11:123–128<br />

Read DJ (1999) Mycorrhiza – The state of art. In: Varma A, Hock B (eds) Mycorrhizae 2nd<br />

edn. Springer, Berlin Heidelberg New York, pp 3–34<br />

Sahay NS, Varma A (1999) Piriformospora indica; a new biological hardening tool for<br />

micropropagated <strong>plant</strong>s. FEMS Microbiol Lett 181:297–302<br />

Sahay NS, Varma A (2000) Biological approach towards increasing the survival rates of<br />

the micropropagated <strong>plant</strong>s. Curr Sci 78:126–129


15 Interaction of Piriformospora indica with Diverse Microorganisms and Plants 265<br />

Singh A, Varma A (2000) Orchidaceous mycorrhizal fungi. In: Mukerji KG, Chamola BP,<br />

Singh J (eds) Mycorrhizal biology. Kluwer Academic/Plenum Publishers, New York,<br />

pp 265–288<br />

Singh A, Sharma J, Rexer K-H, Varma A (2000) Plant productivity determinants beyond<br />

minerals, water and light: Piriformospora indica – A revolutionary <strong>plant</strong>s promoting<br />

fungus. Curr Sci 79:101–106<br />

Singh Ar, Singh An,Varma A (2002a) Piriformospora indica – in vitro raised leguminous<br />

<strong>plant</strong>s: a new dimension in establishment and phytopromotion. Ind J Biotechnol<br />

1:371–376<br />

Singh An, Singh Ar, Rexer K-H, Kost G,Varma A (2002b) Root endosymbiont: Piriformospora<br />

indica – a boon for orchids. J Orchid Soc India 15:89–102<br />

Singh An, Singh Ar, Kumari M, Rai MK,Varma A (2003a) Biotechnological importance of<br />

Piriformospora indica Verma et al. – a novel symbiotic mycorrhiza-like fungus: an<br />

overview. Indian J Biotechnol 2:65–75<br />

Singh An, Singh Ar, Kumari M, Kumar S, Rai MK, Sharma AP,Varma A (2003b) Unmassing<br />

the accessible treasures of the hidden unexplored microbial world. In: Prasad BN<br />

(ed) Biotechnology in sustainable biodiversity and food security. Science Publishers,<br />

Enfield, NH, pp 101–124<br />

Tester M, Smith SE, Smith FA (1987) The phenomenon of “nonmycorrhizal” <strong>plant</strong>s. Can<br />

J Bot 65:419–431<br />

Varma A (1998) Mycorrhizae, the friendly fungi: what we know and how do we know? In:<br />

Varma A (ed) Mycorrhiza manual. Springer, Berlin Heidelberg New York, pp 1–24<br />

Varma A (1999) Functions and applications of arbuscular mycorrhizal fungi in arid and<br />

semi-arid soils. In: Varma A, Hock B (eds) Mycorrhiza. Springer, Berlin Heidelberg<br />

New York, pp 521–556<br />

Varma A,Verma S, Sudha, Sahay NS, Franken P (1999) Piriformospora indica,a cultivable<br />

<strong>plant</strong> growth promoting root endophyte with similarities to arbuscular mycorrhizal<br />

fungi. Appl Environ Microbiol 65:2741–2744<br />

Varma A, Singh A, Sudha, Sahay N, Sharma J, Roy A, Kumari M, Rana D, Thakran S, Deka<br />

D, Bharati K, Franken P, Hurek T, Blechert O, Rexer K-H, Kost G., Hahn A, Hock B,<br />

Maier W, Walter M, Strack D, Kranner I (2001) Piriformospora indica: A cultivable<br />

mycorrhiza-like endosymbiotic fungus. In: Hock B (ed) Mycota IX. Springer, Berlin<br />

Heidelberg New York, pp 123–150<br />

Verma S,Varma A, Rexer K-H, Hassel A, Kost G, Sarbhoy A, Bisen P, Buetehorn B, Franken<br />

P (1998) Piriformospora indica gen. nov; a new root-colonizing fungus. Mycologia<br />

90:895–909


16 Cellular Basidiomycete–Fungus Interactions<br />

Robert Bauer and Franz Oberwinkler<br />

1 Introduction<br />

While basidiomycetes are well known as saprobes, ectomycorrhizal symbionts<br />

or parasites of <strong>plant</strong>s (e.g., Bauer et al. 2001; Hibbett and Thorn 2001),<br />

their role as parasites of other fungi has received scant attention. Thus, the<br />

ultrastructure of the host–parasite interaction in basidiomycetous mycoparasites<br />

has been studied only in a few species (Bauer and Oberwinkler 1990a, b,<br />

1991; Oberwinkler and Bauer 1990; Oberwinkler et al. 1990a, c, 1999; Zugmaier<br />

et al. 1994; Kirschner et al. 2001a).<br />

In this chapter, our data concerning the interfungal cellular interaction of<br />

basidiomycetes are summarized.<br />

2 Occurrence of Mycoparasites Within the Basidiomycota<br />

The division Basidiomycota comprises the classes Urediniomycetes, Ustilaginomycetes<br />

and Hymenomycetes (Swann and Taylor 1993; Begerow et al.<br />

1997). Mycoparasites occur in two of these groups: while scattered throughout<br />

the Urediniomycetes mycoparasites form one of the basal lineages of the<br />

Hymenomycetes (Swann et al. 2001; Weiß and Oberwinkler 2001). Urediniomycetous<br />

mycoparasites include the genera Colacogloea, Colacosiphon,<br />

Cryptomycocolax, Cystobasidium, Heterogastridium, Mycogloea, Naohidea,<br />

Occultifur, Spiculogloea, Zygogloea, and some species of Platygloea (Bandoni<br />

1956, 1984; Oberwinkler 1990; Oberwinkler and Bauer 1990; Oberwinkler et<br />

al. 1990a, b; Roberts 1994, 1996, 1997; Kirschner et al. 2001a). However, the<br />

phenomenon of mycoparasitism may be more widespread among Urediniomycetes<br />

than is currently suspected. Many species of Urediniomycetes (e.g.,<br />

members of Agaricostilbum, Atractogloea, Camptobasidium, Chionosphaera,<br />

Leucosporidium, Naiadella, Rhodosporidium or Sporidiobolus), currently<br />

thought to be saprobes may be capable of parasitizing fungi (Oberwinkler<br />

and Bandoni 1982, 1989; Marvanová and Bandoni 1987; Marvanová and<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


268<br />

Suberkropp 1990; Bauer et al. 1997; Roberts 1997; Kirschner et al. 2001b).<br />

Within the Hymenomycetes, mycoparasitism is common among the Tremellales<br />

sensu Bandoni (1984). In addition, some lichen parasites (Diederich<br />

1996) also may belong in the Tremellales.<br />

3 Hosts<br />

Robert Bauer and Franz Oberwinkler<br />

Hosts of the basidiomycetous mycoparasites are either ascomycetes or basidiomycetes.<br />

Mycoparasitism on chytrids or zygomycetes is unknown. There<br />

appears to be no phylogenetic correlation between the basidiomycetous<br />

mycoparasites and their respective host fungi. In other words, a distinct<br />

basidiomycetous group of mycoparasites usually occurs on both ascomycetes<br />

and basidiomycetes.<br />

4 Cellular Interactions<br />

The basidiomycetous interfungal cellular interactions can be divided into two<br />

main types at the structural level.<br />

4.1 Colacosome-Interactions<br />

Mycoparasites in the Microbotryomycetidae (Bauer et al. 1997; Swann et al.<br />

1999) are generally characterized by their interaction via a unique organelle,<br />

the colacosome, formed at the interface between the parasite and its fungal<br />

host. This mycoparasitic organelle was firstly described in detail from the<br />

interaction of the parasite Colacogloea peniophorae and its host Hyphoderma<br />

praetermissum (Oberwinkler et al. 1990a; Bauer and Oberwinkler 1991). Colacosomes<br />

develop in the contact area between the parasite and its host as illustrated<br />

in Fig. 1. They are positioned at the inner <strong>surface</strong> of the parasite cell<br />

outside the cytoplasm, but inside the cell wall. Their shape is globular, subglobular<br />

or beaked. The central part of the colacosome is electron-opaque,<br />

0.3–0.4 µm diameter, enclosed by a membrane and surrounded by an electron-transparent,<br />

unstructured sheath of approximately 0.05 µm diameter.<br />

The colacosome is covered by the plasmalemma. A thin secondary cell wall<br />

layer is often present along the plasmalemma covering the colacosome.<br />

During formation (Fig. 2), the plasma membrane of the parasite is folded<br />

into the cytoplasm, then recurves, and finally fuses with itself at a distance of<br />

0.2–0.3 µm from the original outgrowth. Consequently, it is surrounded by a<br />

membrane as a derivative from the plasma membrane. The globose compartment<br />

is now separated from the cytoplasm by an electron-transparent, intermembranaceous<br />

space. After separation from the cytoplasm, the vesicular


Fig. 1. Hypha of Colacogloea<br />

peniophorae with colacosomes<br />

(arrows) contacting<br />

host hypha. Bar 1µm<br />

16 Cellular Basidiomycete–Fungus Interactions 269<br />

core becomes homogeneous and finally more and more electron-opaque.<br />

Simultaneously, the intermembranaceous space between the central part of<br />

the colacosome and the cytoplasm increases slightly in thickness. Interaction<br />

starts with intrusion of electron-opaque core material of the colacosome into<br />

the cell wall of the parasite (Fig. 3). The cell wall close to the intrusion peg<br />

becomes electron-transparent and indistinct in substructure. Intrusion then<br />

continues through the closely attached cell wall of the host into an electrontransparent<br />

protuberance formed between the cell wall and the plasmalemma<br />

of the host (Fig. 4).<br />

In Colacogloea peniophorae colacosomes develop in great numbers close<br />

together (Fig. 1). It is evident that in most cases hyphae possessing colacosomes<br />

and their host hyphae lie for a relatively long distance side by side<br />

closely attached to one another (Fig. 1). Furthermore, the host hyphae often<br />

form one or two spirals around the colacosome-possessing hyphae (Bauer<br />

and Oberwinkler 1991). As discussed by Bauer and Oberwinkler (1991), this<br />

situation may be explained as follows: in the beginning, the parasite hypha<br />

grows loosely in the host fructifications. After a first, probably accidental,<br />

contact of the hypha with a host hypha, colacosomes develop rapidly and in<br />

great number. The electron-opaque content of the colacosomes penetrates the<br />

host cell wall. Thus, the colacosomes combine both cells and the first contact


270<br />

Robert Bauer and Franz Oberwinkler<br />

Fig. 2. Diagram of colacosome development, modified from Bauer and Oberwinkler<br />

(1991).Abbreviations and symbols: CH cell wall of the host Hyphoderma praetermissum,<br />

CP cell wall of the parasite Colacogloea peniophorae, CS secondary cell wall layer, H cell<br />

of the host Hyphoderma praetermissum, P cell of the parasite Platygloea peniophorae, PH<br />

plasma membrane of the host Hyphoderma praetermissum, PP plasma membrane of the<br />

parasite Colacogloea peniophorae. (top left) Initial stage of invagination of the plasma<br />

membrane of the parasite. (top right) The plasmalemma of the parasite recurves. (middle<br />

left) Delimitation of the young colacosome from the cytoplasm. (middle right) The<br />

central part of the colacosome becomes homogeneous and more and more electronopaque.<br />

The electron-transparent sheath of the colacosome increases in thickness.<br />

(lower left) The electron-opaque core material penetrates the cell wall of the parasite and<br />

begins to intrude the cell wall of the host. (lower left) Final developmental stage with<br />

colacosome penetration through host cell wall<br />

remains stable. Furthermore, if the parasite and/or the host hypha continue to<br />

grow, additional colacosomes are rapidly developed. Consequently, the number<br />

of connections between both organisms is continually increased and both<br />

are forced to grow in close contact to each other. The development of colacosomes<br />

is, therefore accompanied by an increase of the host – parasite interface.<br />

In this sense, the colacosomes could serve as connecting agents. It is<br />

unclear from the present data, however, whether or not the colacosomes are<br />

involved in host–parasite metabolism functions as no specific attempts to


16 Cellular Basidiomycete–Fungus Interactions 271<br />

Fig. 3. Hypha of Colacogloea peniophorae (lower cell) in contact with a host hypha (upper<br />

cell). The electron-opaque core (c) of the colacosome intrudes into the cell wall of the<br />

parasite. Note the tripartite membrane (arrow) around the core of the colacosome (c).<br />

The colacosome is covered by a thin secondary cell wall layer (arrowhead) and the<br />

plasma membrane (double arrowhead) of the parasite. Bar 0.1 µm<br />

Fig. 4. Hypha of<br />

Colacogloea peniophorae<br />

(lower cell) in contact with<br />

a host hypha (upper cell).<br />

Final stage of host – parasite<br />

interaction with the<br />

content of the electronopaque<br />

core of the colacosome<br />

(c) penetrating the<br />

host cell wall. Bar 0.2 µm


272<br />

Robert Bauer and Franz Oberwinkler<br />

Fig. 5. Longitudinally sectioned hypha of “Mycospira”(m) surrounded by a hyphal spiral<br />

of the host fungus (three-dimensional configuration reconstructed from serial sections).<br />

Note the colacosomes (arrows) at the contact area. Bar 1 µm<br />

Fig. 6. Host cell (H) intruding<br />

into a hyphal cell of<br />

Colacogloea sp. Colacosomes<br />

(arrows) surround the intracellular<br />

part of the host cell<br />

which lacks a cell wall. Bar<br />

1µm


16 Cellular Basidiomycete–Fungus Interactions 273<br />

identify them have been made. However, the change in the electron density of<br />

the core material of the colacosome suggests that an alteration of the chemical<br />

composition occurs after separation of the colacosome from the cytoplasm.<br />

Furthermore, the penetration of the parasite and host cell wall appears<br />

to be enzymatic since both cell walls are not distorted at the site of penetration.<br />

This interpretation is reinforced by the mycoparasitic behavior of a currently<br />

undescribed basidiomycete, called here “Mycospira”. In contact with its<br />

host, a member of Tulasnella, this fungus develops colacosomes in exact spirals.<br />

As a consequence, the host fungus grows in spirals around the colacosome-possessing<br />

hyphae (Fig. 5). Thus, the formation of colacosomes results<br />

Fig. 7. Host cell (H) intruding into a hyphal cell of Cryptomycocolax abnorme.Colacosomes<br />

(arrows) surround the intracellular part of the host cell which lacks a cell wall.<br />

Two fusion pores are visible at arrowheads. Note that the colacosomes are more electrontransparent<br />

than in Fig. 6. Bar 1 µm


274<br />

Robert Bauer and Franz Oberwinkler<br />

in a greatly increased contact zone between the parasite and its host. This is<br />

also the case in a third type of colacosome-arrangement. In Colacogloea bispora<br />

(Oberwinkler et al. 1999), Colacogloea sp. (Fig. 6), Colacosiphon (Kirschner<br />

et al. 2001a), Cryptomycocolax (Oberwinkler and Bauer 1990), Krieglsteinera,<br />

and Heterogastridium, the appearance of colacosomes is associated with<br />

curious interaction structures: filamentous outgrowths of the host cells are<br />

intimately enclosed by galloid parasite cells. Numerous colacosomes are present<br />

along the contact area between the host intrusion and the parasite cell<br />

(Fig. 6). These host intrusions always terminate in the parasite cell. They are<br />

unseptate, often branched and, astonishingly, lack cell walls, thus giving the<br />

impression of haustoria (of the host into the parasite!!!). In Cryptomycocolax<br />

a second type of colacosome was found along the cytoplasmic intrusions of<br />

the host formed into the hyphae of the parasite (Fig. 7; Oberwinkler and<br />

Bauer 1990). These colacosomes have a more electron-transparent core and<br />

they fuse with the host cell via a pore of approximately 7–14 nm in diameter<br />

(Figs. 7, 8). It is clear that the cellular interaction of Cryptomycocolax is complex<br />

and currently misunderstood.<br />

Colacosomes have also been found in Atractocolax, Leucosporidium,<br />

Mastigobasidium, Rhodosporidium and Sporidiobolus, indicating a potential<br />

for mycoparasitism in these genera that have been assumed to be saprobic<br />

(Kreger van Rij and Veenhuis 1971; Bauer et al. 1997; Kirschner et al. 1999).<br />

Fig. 8. Colacosome of Cryptomycocolax<br />

abnorme in<br />

contact with the host cytoplasm<br />

(H) showing the<br />

fusion pore (arrowhead) in<br />

detail. Note that the pore<br />

membranes are continuous<br />

with both the host plasma<br />

membrane and the membrane<br />

surrounding the core<br />

(c) of the colacosome<br />

(arrow). Bar 0.1 µm


4.2 Fusion-Interaction<br />

16 Cellular Basidiomycete–Fungus Interactions 275<br />

Typical fusion mycoparasites (Bauer and Oberwinkler 1990a, b) are the<br />

Tremellales (including the Filobasidiales) of the Hymenomycetes (Bandoni<br />

1984, 1995). Astonishingly, however, fusion mycoparasites are also scattered<br />

throughout the Urediniomycetes. For example, the members of Cystobasidium,<br />

Mycogloea, Naohidea, Occultifur, Spiculogloea and Zygogloea are fusion<br />

mycoparasites (unpubl. data). Usually, basidiomycetous fusion mycoparasites<br />

interact with their respective hosts by specialized interactive cells, designated<br />

often as “tremelloid haustorial cells”. These cells were first described and designated<br />

as “haustoria” by Olive (1947). Each tremelloid haustorial cell is subtended<br />

by a clamp and consists of a subglobose basal part with one or more<br />

thread-like filaments (e.g., see Oberwinkler et al. 1984) that are capable of fusing<br />

with host cells via a pore of approximately 14–19 nm (Figs. 9, 10; Bauer<br />

and Oberwinkler 1990a, b). Thus, a direct cytoplasm – cytoplasm connection<br />

between the parasites and their respective hosts occurs.As discussed by Bauer<br />

and Oberwinkler (1990a), the following stages in the development of the cel-<br />

Fig. 9. Haustorial filament of Tetragoniomyces uliginosus (lower cell) in contact with a<br />

host hypha (upper cell) demonstrating the fusion pore (arrowhead). Bar 0.5 µm


276<br />

Robert Bauer and Franz Oberwinkler<br />

Fig. 10. Haustorial filament of Tetragoniomyces uliginosus (lower cell) in contact with a<br />

host hypha (upper cell) illustrated to show the fusion pore (arrowhead) in detail. Note<br />

that the pore membranes are continuous with the plasma membranes of both cells. Bar<br />

0.1 µm<br />

Fig. 11. Haustorial filament<br />

of Christiansenia pallida<br />

(lower cell) penetrating a<br />

host cell (upper cell). One<br />

fusion pore is visible at<br />

arrowhead. Bar 0.2 µm


Fig. 12. Transverse section<br />

through a penetrating haustorial<br />

filament of Christiansenia<br />

pallida. One fusion pore medianly<br />

sectioned (arrow) and four<br />

pores nonmedianly sectioned<br />

(arrowheads). Bar 0.2 µm<br />

16 Cellular Basidiomycete–Fungus Interactions 277<br />

lular interaction can be recognized: (1) contact of the haustorial filament with<br />

the host cell, (2) nesting of the haustorial filament into the host cell wall, and<br />

(3) fusion of the haustorial and host cell protoplasts via a pore.<br />

In the interaction between Christiansenia pallida and its host (Bauer and<br />

Oberwinkler 1990b), the haustorial filament forms one or more protrusions<br />

into the host cells where a lot of fusion pores develop (Figs. 11, 12).<br />

Direct cytoplasm – cytoplasm connections between mycoparasites and<br />

their respective hosts represent an unusual type of cellular interaction.As discussed<br />

by Bauer and Oberwinkler (1990a), this type of interaction may be<br />

considered as most effective. Substances required by the parasite do not need<br />

to cross membranes or cell walls. Thus, the fusion pores could serve as direct<br />

avenues for nutrients (Hoch 1977).<br />

5 Basidiomycetous Mycoparasitism, a Result of Convergent<br />

Evolution?<br />

The different mode of mycoparasitism occurring in the basidiomycetes, as<br />

discussed above, suggests that mycoparasitism may have evolved at least twice<br />

(or more) in the basidiomycetous history. Thus, it appears that the Microbotryomycetidae<br />

(for the subclass, see Swann et al. 1999) arose independently


278<br />

Robert Bauer and Franz Oberwinkler<br />

from the fusion mycoparasites as colacosome-mycoparasites (Bauer et al.<br />

1997). For the occurrence of fusion mycoparasites in the Urediniomycetes on<br />

the one hand, and in the Hymenomycetes on the other, two explanations are<br />

possible: (1) the fusion mycoparasitism occurring in these two groups is a<br />

result of convergent evolution, and, alternatively (2), the common ancestor of<br />

both groups or of the basidiomycetes in general was a fusion mycoparasite.<br />

Additional phylogenetic studies are necessary to clarify this situation.<br />

6 Conclusions<br />

The phenomenon of mycoparasitism may be more widespread among basidiomycetes<br />

than is currently suspected. Our data illustrate that mycoparasitic<br />

basidiomycetes evolved a fascinating array of different strategies at the cellular<br />

level to benefit from host metabolites.<br />

Acknowledgements. We thank Uwe Simon for critically reading the manuscript, and the<br />

Deutsche Forschungsgemeinschaft for financial support.<br />

References and Selected Reading<br />

Bandoni RJ (1956) A preliminary survey of the genus Platygloea. Mycologia 48:821–840<br />

Bandoni RJ (1984) The Tremellales and Auriculariales: an alternative classification.<br />

Trans Mycol Soc Jpn 25:489–530<br />

Bandoni RJ (1995) Dimorphic heterobasidiomycetes: taxonomy and parasitism. Stud<br />

Mycologia 38:13–27<br />

Bauer R, Oberwinkler F (1990a) Direct cytoplasm-cytoplasm connection: an unusual<br />

host-parasite interaction of the tremelloid mycoparasite Tetragoniomyces uliginosus.<br />

Protoplasma 154:157–160<br />

Bauer R, Oberwinkler F (1990b) Haustoria of the mycoparasitic heterobasidiomycete<br />

Christiansenia pallida. Cytologia 55:419–424<br />

Bauer R, Oberwinkler F (1991) The colacosomes: new structures at the host-parasite<br />

interface of a mycoparasitic basidiomycete. Bot Acta 104:53–57<br />

Bauer R, Oberwinkler F, Vánky K (1997) Ultrastructural markers and systematics in<br />

smut fungi and allied taxa. Can J Bot 75:1273–1314<br />

Bauer R, Begerow D, Oberwinkler F, Piepenbring M, Berbee ML (2001) Ustilaginomycetes.<br />

In: McLaughlin DJ, McLaughlin EG, Lemke PA (eds) Mycota VII Part B, Systematics<br />

and evolution. Springer, Berlin Heidelberg New York, pp 57–83<br />

Begerow D, Bauer R, Oberwinkler F (1997) Phylogenetic studies on large subunit ribosomal<br />

DNA sequences of smut fungi and related taxa. Can J Bot 75:2045–2056<br />

Diederich P (1996) The lichenicolous heterobasidiomycetes. Bibliotheca Lichenologica<br />

61:1–198<br />

Hibbett DS, Thorn RG (2001) Basidiomycota: Homobasidiomycetes. In: McLaughlin DJ,<br />

McLaughlin EG, Lemke PA (eds) Mycota VII. Part B, Systematics and evolution.<br />

Springer, Berlin Heidelberg New York, pp 122–168


16 Cellular Basidiomycete–Fungus Interactions 279<br />

Hoch HC (1977) Mycoparasitic relationships: Gonatobotrys simplex parasitic on Alternaria<br />

tenuis. Phytopathology 67:309–314<br />

Kirschner R, Bauer R, Oberwinkler F (1999) Atractocolax, a new heterobasidiomycetous<br />

genus based on a species vectored by conifericolous bark beetles. Mycologia 91:538–<br />

543<br />

Kirschner R, Bauer R, Oberwinkler F (2001a) Colacosiphon: a new genus described for a<br />

mycoparasitic fungus. Mycologia 93:634–644<br />

Kirschner R, Begerow D, Oberwinkler F (2001b) A new Chionosphaera associated with<br />

conifer inhabiting bark beetles. Mycol Res 105:1403–1408<br />

Kreger-van Rij NJW, Veenhuis M (1971) Some features of the genus Sporidiobolus<br />

observed by electron microscopy. Antonie van Leeuwenhoek 37:253–255<br />

Marvanová L, Bandoni RJ (1987) Naiadella fluitans gen. et sp. nov.: a conidial basidiomycete.<br />

Mycologia 79:578–586<br />

Marvanová L, Suberkropp K (1990) Camptobasidium hydrophilum and its anamorph,<br />

Crucella subtilis: a new heterobasidiomycete from streams. Mycologia 82:208–217<br />

Oberwinkler F (1990) New genera of auricularioid heterobasidiomycetes. Rep Tottori<br />

Mycol Inst 28:113–127<br />

Oberwinkler F, Bandoni RJ (1982) Atractogloea: a new genus in the Hoehnelomycetaceae<br />

(Heterobasidiomycetes). Mycologia 74:634–639<br />

Oberwinkler F, Bauer R (1989) The systematics of gastroid, auricularioid heterobasidiomycetes.<br />

Sydowia 41:224–256<br />

Oberwinkler F, Bauer R (1990) Cryptomycocolax: a new mycoparasitic heterobasidiomycete.<br />

Mycologia 82:671–692<br />

Oberwinkler F, Bandoni RJ, Bauer R, Deml G, Kisimova-Horovitz L (1984) The life history<br />

of Christiansenia pallida, a dimorphic, mycoparasitic heterobasidiomycete.<br />

Mycologia 76:9–22<br />

Oberwinker F, Bauer R, Bandoni RJ (1990a) Colacogloea: a new genus in the auricularioid<br />

heterobasidiomycetes. Can J Bot 68:2531–2536<br />

Oberwinkler F, Bauer R, Bandoni RJ (1990b) Heterogastridiales: a new order of basidiomycetes.<br />

Mycologia 82:48–58<br />

Oberwinkler F, Bauer R, Schneller J (1990 c) Phragmoxenidium mycophilum sp. nov., an<br />

unusual mycoparasitic heterobasidiomycete. Syst Appl Microbiol 13:186–191<br />

Oberwinkler F, Bauer R, Tschen J (1999) The mycoparasitism of Platygloea bispora.Kew<br />

Bull 54:763–769<br />

Olive LS (1947) Notes on the Tremellales on Georgia. Mycologia 39:90–108<br />

Roberts P (1994) Zygogloea gemellipara: an auricularioid parasite of Myxarium nucleatum.<br />

Mycotaxon 52:241–246<br />

Roberts P (1996) Heterobasidiomycetes from Majorca and Cabrera (Balearic Islands).<br />

Mycotaxon 60:111–123<br />

Roberts P (1997) New heterobasidiomycetes from Great Britain. Mycotaxon 63:195–216<br />

Swann EC, Taylor JW (1993) Higher taxa of basidiomycetes: an 18S rRNA gene perspective.<br />

Mycologia 85:923–936<br />

Swann EC, Frieders EM, McLaughlin DJ (1999) Microbotryum, Kriegeria and the changing<br />

paradigm in basidiomycete classification. Mycologia 91:51–66<br />

Swann EC, Frieders EM, McLaughlin DJ (2001) Urediniomycetes. In: McLaughlin DJ,<br />

McLaughlin EG, Lemke PA (eds) Mycota VII Part B, Systematics and evolution.<br />

Springer, Berlin Heidelberg New York, pp 37–56<br />

Weiß M, Oberwinkler F (2001) Phylogenetic relationships in Auriculariales and related<br />

groups – hypotheses derived from nuclear ribosomal DNA sequences. Mycol Res<br />

105:403–415<br />

Zugmaier W, Bauer R, Oberwinkler F (1994) Mycoparasitism of some Tremella species.<br />

Mycologia 86:49–56


17 Fungal Endophytes<br />

Sita R. Ghimire and Kevin D. Hyde<br />

1 Introduction<br />

Fungal endophytes have been isolated from almost every vascular <strong>plant</strong> studied<br />

and much has been written about their role and ecology. In this paper we<br />

review these aspects, but also review the role of molecular techniques in endophyte<br />

identification, the possible relationship with host specificity of fungal<br />

saprobes and suggest future areas for study.<br />

2 Definition of a Fungal Endophyte<br />

The term endophyte was introduced by De Bary (1866) and was initially<br />

applied to any organism found within a <strong>plant</strong> (Wilson 1995). The meaning of<br />

the term endophyte has been refined over time with the addition of new<br />

information (Siegel et al. 1984; Carroll 1986; Petrini 1986). Petrini (1991) used<br />

the term endophyte to mean all organisms inhabiting <strong>plant</strong> organs that at<br />

some time in their life can colonize internal <strong>plant</strong> tissues without causing<br />

apparent harm to the host. This has been the most widely used definition of<br />

endophytes and also includes the organisms that have a more or less lengthy<br />

epiphytic phase and also latent pathogens (Petrini 1991; Schulz et al. 1998).<br />

There has however, been a certain level of disagreement expressed by some<br />

mycologists over the inclusion of <strong>plant</strong> pathogens as endophytes, since endophytes<br />

are nonaggressive, nonpathogenic and have developed a mutualistic<br />

role with their hosts (Freeman and Rodriguez 1993; Tyler 1993; Stone et al.<br />

1994; Sinclair and Cerkauskas 1996).<br />

Studies on the endophyte composition in different hosts have identified<br />

organisms with varying roles within their hosts. Organisms having weak parasitic<br />

associations, localized infection, quiescent infection, latent infection<br />

and aggressive parasitic relationships with their hosts have often been recovered<br />

(Jersch et al. 1989; Kehr 1992; Gotz et al. 1993; Kehr and Wulf 1993;<br />

Williamson 1994; Agrios 1997). Wilson (1995) provided a working definition<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


282<br />

Sita R. Ghimire and Kevin D. Hyde<br />

of the term by analyzing the different levels of endophytic association and<br />

stated that “endophytes are fungi or bacteria which, for all or part of their life<br />

cycle, invade the tissues of living <strong>plant</strong>s and cause unapparent and asymptomatic<br />

infections entirely within <strong>plant</strong> tissues but causes no symptoms of the<br />

disease”. The same organism may also be described as a saprobe or pathogen<br />

at other times (Boddy and Griffith 1989).<br />

3 Role of Endophytes<br />

Endophytes have previously been defined as mutualists and are closely<br />

related to virulent pathogens, but have limited pathogenicity, and have probably<br />

evolved directly from <strong>plant</strong> pathogenic fungi (Carroll 1988). The mutualistic<br />

symbiosis includes the lack of destruction of most cells or tissues,<br />

nutrient and chemical cycling between the fungus and hosts, enhanced<br />

longevity and photosynthetic capacity of cell and tissue under the influence<br />

of infection, enhanced survival of fungus, and a tendency towards greater<br />

host specificity than is seen in necrotrophic infections (Lewis 1973). It is<br />

often difficult to differentiate between an endophyte and pathogen as many<br />

<strong>plant</strong> pathogens undergo an extensive phase of asymptomatic latent infection<br />

before the appearance of disease symptoms and the mutation in a single<br />

genetic locus can change a pathogen to nonpathogenic endophytic<br />

organism with no effect on its host specificity (Freeman and Rodriguez<br />

1993). Latent infection is the state in which a host is infected with a<br />

pathogen, but does not show any symptoms and persists until signs or<br />

symptoms are prompted to appear by environmental or nutritional conditions<br />

or by the state of maturity of the host or pathogen (Agrios 1997). The<br />

latent infection is considered as the highest level of parasitism because the<br />

host and parasite coexist for a period of time with minimal damage to the<br />

host. Hence, the relationship between <strong>plant</strong> pathogenic fungi and host is<br />

considered as parasitic.<br />

Wilson (1995) argued that the term endophyte bears much affinity to the<br />

term pathogen and stated that it is often difficult to be able to classify a particular<br />

species. Sinclair and Cerkauskas (1996) compared endophyte colonization<br />

and latent infections by fungi and stated that they are distinctly different.<br />

Endophytic fungi are asymptomatic and considered mutualistic,<br />

whereas latent infecting fungi are parasitic and cannot be considered mutualistic.<br />

Rather, they are considered to be one of the most advanced stages of parasitism<br />

as the host and parasite co-exist for a period of time with minimal<br />

effect on the host (Sinclair and Cerkauskas 1996). Hammon and Faeth (1992)<br />

suggested that the disproportionate amount of attention that has been paid to<br />

the study of grass endophytes has lead to the impression that all endophytes<br />

must be mutualists. There seems to be a greater probability of mutualism in<br />

the fungal species that are transmitted through seeds, as transmission will


17 Fungal Endophytes 283<br />

increase directly as a result of host survival. The association where only one<br />

fungus is associated within the host <strong>plant</strong> is more likely to be mutualistic<br />

(Hammon and Faeth 1992).<br />

The endophytes associated with grasses have received much attention, and<br />

many of these have been found to produce physiologically active alkaloids<br />

that cause their hosts to be toxic to mammals and increase their resistance to<br />

insect herbivores (Funk et al. 1983; Clay 1988; Cheplick and Clay 1988;<br />

Prestidge and Gallagher 1988). In the grasses and other <strong>plant</strong> hosts, endophytes<br />

have also been shown to enhance <strong>plant</strong> growth, reduce infection by<br />

nematodes, increase stress tolerance and increase nitrogen uptake in nitrogen<br />

deficit-soils (Latch et al. 1985; Clay 1987, 1990; Kimmons 1990; Bacon 1993;<br />

Gasoni and Stegman De Gurfinkel 1997; Rommert et al. 1998; Verma et al.<br />

1999; Bultman and Murphy 2000;). Several reviews are available on secondary<br />

metabolite production by endophytes (Miller 1986; Clay 1991; Petrini et al.<br />

1992). Endophytes in culture can produce biologically active compounds<br />

(Brunner and Petrini 1992) including several alkaloids, paxilline, lolitrems<br />

and tertraenone steroids (Dahlman et al. 1991), antibiotics (Fisher et al. 1984a,<br />

b) and <strong>plant</strong> growth promoting factors (Petrini et al. 1992). Endophytes are<br />

increasingly being identified as a group of organisms capable of providing a<br />

source of secondary metabolites for use in biotechnology and agriculture<br />

(Bills and Polishook 1992).<br />

4 Modes of Endophytic Infection and Colonization<br />

The colonization of <strong>plant</strong> tissues by endophytes, <strong>plant</strong> pathogens and mycorrhizae<br />

involves several steps involving host recognition, spore germination,<br />

penetration of the epidermis and tissue colonization (Petrini 1991, 1996). The<br />

inoculum source of fungal endophytes is widely considered to be the airborne<br />

spores, and also seed transmission and transmission of propagules by insect<br />

vectors (Petrini 1991). A high level of genetic diversity of endophyte isolates<br />

suggests that infection foci arise from different strains of fungi derived from<br />

constant new inoculum (Hammerli et al. 1992; Rodrigues et al. 1993). In terms<br />

of mechanical and enzymatic elements of penetration by endophytic fungi, it<br />

can be assumed that endophytes adopt the same strategy for penetration of<br />

host tissue as pathogens (Petrini et al. 1992). Fungi can invade <strong>plant</strong> tissues by<br />

direct cuticular penetration, via appressoria formed on the cuticle, after<br />

which penetration occurs through the cuticle and epidermal cell wall or via<br />

natural openings like stomata (O’Donnell and Dickinson 1980; Muirhead and<br />

Deverall 1981; Kulik 1988; Cabral et al. 1993; Viret et al. 1993; Viret and Petrini,<br />

1994). Following penetration the infection may be inter-cellular or intra-cellular<br />

and may be limited to one cell or in a limited area around the penetration<br />

site. Limited cytological work on nonclavicipitaceous endophytes have<br />

shown that the infection of these endophytes in host <strong>plant</strong>s may be inter- or


284<br />

Sita R. Ghimire and Kevin D. Hyde<br />

intra-cellular and often localized in single cells (Stone 1988; Suske and Acker<br />

1989, Cabral et al. 1993).<br />

Some endophytic fungi (including those which are latent pathogens) are<br />

host-specific, whereas others seem to invade any available hosts (Carroll 1988;<br />

Petrini et al. 1992). When studying the infection of Juncus spp. with the endophytes<br />

Stagnospora innumerosa and Drechslera spp., Cabral et al. (1993)<br />

observed callose formation in the individual cells as a host defense response.<br />

Sieber et al. (1991) found that the synthesis of highly specialized enzymes<br />

associated the penetration of cuticular layers of the host by the endophyte<br />

Melanconium spp. Several other investigations have also reported a growth<br />

response of host calli to endophytes (Hendry et al. 1993; Peters et al. 1998).<br />

Schulz et al. (1999) studied the secondary metabolites produced by endophytes<br />

and their host interactions in order to understand why endophytic<br />

infections are symptomless. The production of herbicidally active substances<br />

was three times that of soil isolates and twice that of phytopathogenic fungi,<br />

whereas the phenolic metabolites in the host were higher in the roots of <strong>plant</strong>s<br />

infected with an endophyte than in those infected with pathogens. Their<br />

study hypothesized that both the pathogen–host and endophyte–host interaction<br />

involved constant mutual antagonisms, at least in part based on the<br />

secondary metabolites the partners produce. The pathogen – host interaction<br />

was thought to be imbalanced and resulted in disease while that of the endophytes<br />

and its host is a balanced antagonism.<br />

5 Isolation of Endophytes<br />

Techniques for endophyte isolation and culture have been developed gradually<br />

over time. Bacon and White (1994) have written an excellent review on<br />

staining, media and procedure for analyzing endophytes. Endophytes can be<br />

isolated from various <strong>plant</strong> parts such as seeds, leaf and stem and direct isolation<br />

of ascospores is also in practice. The <strong>plant</strong> and <strong>plant</strong> parts collected<br />

for studying endophytic communities should look apparently healthy, in<br />

order to minimize the compounding effect because of <strong>plant</strong> pathogenic and<br />

saprobic species. Young tissue is appropriate for isolation as older tissues<br />

often contain many additional fungi that make isolation of slow growing<br />

fungi difficult (Bacon and White 1994). The samples should be processed in<br />

the shortest time possible after collection. Plant parts for investigation<br />

should be cut into small pieces to facilitate sterilization and isolation<br />

processes. Bills (1996) discussed various <strong>surface</strong> sterilization techniques in<br />

detail. Any method can be used for <strong>surface</strong> sterilization provided that it can<br />

eliminate most of the epiphytic fungi from the exterior tissues and encourage<br />

the growth of the internal mycobiota. The method used by Petrini et al.<br />

(1992) has been used extensively and found very successful in studying<br />

endophytes (Rodrigues and Samuels 1990; Schulz et al. 1993). This method


comprises dipping samples in 96 % ethanol for 1 min, then in 65 % commercial<br />

Chlorox (final concentration 3.25 % aqueous sodium hypochlorite)<br />

for 10 min and finally in 96 % ethanol for 30 s. Malt extract agar is considered<br />

the most suitable media for the growth and sporulation of endophytic<br />

fungi (Bills and Polishook 1992; Bills 1996). Amendment of medium with<br />

streptomycin sulfate is practised to prevent bacterial contamination. To prevent<br />

the fast growing fungi overgrowing the plate, a growth inhibitor, Ross<br />

Bengal is added to the agar.<br />

Surface-sterilized <strong>plant</strong> tissues are plated in an appropriate medium<br />

amended with antibiotics and Rose Bengal and incubated at room temperature<br />

with periodic light and darkness. Incubated plates are checked after<br />

1 week of incubation at regular intervals for fungal development. If the colony<br />

is very small and there is a risk of engulfment by other colonies, it needs to be<br />

subcultured. Subcultured isolates are generally maintained at room temperature<br />

for many weeks to study morphological and other characteristics. Some<br />

isolates may fail to produce reproductive structures even after several<br />

months. Subculture of these isolates onto medium with autoclaved host tissue<br />

strips can promote sporulation (Matsushima 1971). In general, sterile isolates<br />

should be checked regularly for fruiting bodies over a period of 3–4 months<br />

and the isolates failing to produce fruiting body are referred to as sterile<br />

“morphotypes” depending on the characteristics of culture.<br />

Other methods to promote sporulation of morphospecies should also be<br />

tried. Guo et al. (1998) used twigs in conical flasks over a 3-month period to<br />

promote sporulation of endophytes. Other methods can be designed, but<br />

should try to mimic the situation in nature as closely as possible.<br />

6 Molecular Characterization of Endophytes<br />

17 Fungal Endophytes 285<br />

Molecular approaches have been used to resolve the problems in fungal taxonomy<br />

and in the identification of fungi (Rollo et al. 1995; Ma et al. 1997;<br />

Zhang et al. 1997; Ranghoo et al. 1999). The use of molecular techniques for<br />

the direct detection and identification of fungi within natural habitats has<br />

been reviewed by Liew et al. (1998). Molecular techniques have mainly been<br />

used in the detection and identification of mycorrhizal fungi and phytopathogenic<br />

fungi directly from within <strong>plant</strong> tissues (Mills et al. 1992; Johanson and<br />

Jeger 1993; Beck and Ligon 1995; Bonito et al. 1995; Abbas et al. 1996; Chambers<br />

et al. 1998). Similarly, molecular techniques have been employed to detect<br />

and identify fungi from the grass clothing of Iceman, from bamboo leaves and<br />

glacial ice strata (Rollo et al. 1995; Ma et al. 1997; Zhang et al. 1997).<br />

A most frequently encountered problem in endophyte study is the presence<br />

of mycelia sterilia, making their identification difficult (Guo et al. 2000).Variable<br />

proportions of mycelia sterilia have been reported ranging from 11 % of<br />

isolates from palm (Trachycarpus fortunei) in China, 13 % of endophytes


286<br />

Sita R. Ghimire and Kevin D. Hyde<br />

obtained from two Licuala species in Brunei and Australia, 15 % of isolates<br />

from evergreen shrubs in western Oregon, 16.5 % of isolates from fronds of<br />

Livistona chinensis in Hong Kong, 27 % of isolates from leaves of Sequoia sempervirens<br />

in central California and 54 % of isolates obtained from twigs of<br />

Quercus ilex in Switzerland (Petrini et al. 1982; Espinosa-Garcia and Langenheim<br />

1990; Fisher et al. 1994; Taylor et al. 1999; Frohlich et al. 2000; Gou et al.<br />

2000). A high proportion of unidentified endophytic isolates resulting from<br />

traditional methodology has prompted various workers to develop methodology<br />

to improve sporulation in mycelia sterilia (Matsushima 1971; Guo et al.<br />

1998; Taylor et al. 1999; Frohlich et al. 2000). The problem of having many<br />

nonidentifiable mycelia sterilia, however still remains. Hence, molecular techniques<br />

could be the best alternative to identify this taxa.<br />

There have been only a small number of studies using molecular techniques<br />

to investigate endophytic fungal communities. Random amplified<br />

polymorphic DNA (RAPD) markers were used to study the genotypic diversity<br />

in the populations of Rabdocline parkeri from Douglas fir (McCutcheon<br />

and Carroll 1993). Specific PCR primers were used to amplify rDNA fragments<br />

of the endophyte Acremonium coenophialum from infected tall fescue<br />

tissues (Doss and Welty 1995). The genetic diversity of Epichloe typhina, an<br />

endophyte in Bromus erectus, was studied using a microsatellite-containing<br />

locus as a molecular marker (Groppe et al. 1995). Guo et al. (2000) performed<br />

phylogenetic analysis based on rDNA of 19 morphospecies from frond tissues<br />

of Livistona chinensis and found that they were filamentous Ascomycota<br />

belonging to the different taxonomic levels in the Loculoascomycetes and<br />

Pyrenomycetes. The 5.8S gene and flanking internal transcribed spacer of<br />

rDNA were used in detection and taxonomic placement of endophytic fungi<br />

within frond tissues of Livistona chinensis (Guo et al. 2001). Ribosomal DNA<br />

sequence analysis was used to validate the morphospecies concept used in<br />

endophyte study to group mycelia sterilia (Hyde et al. 2001). Therefore, rDNA<br />

sequence analysis is in frequent use to resolve the identification problem<br />

associated with endophytic fungi. These studies show increasing an use of<br />

molecular techniques in detection, identification, and population and ecological<br />

studies of endophytes.<br />

7 Are Endophytes Responsible for Host<br />

Exclusivity/Recurrence in Saprobic Fungi?<br />

There has been much debate as to whether saprobic fungi are host-specific as<br />

this has important implications for estimates of fungal numbers (Hawksworth<br />

1991, 2001; Fröhlich and Hyde 1999; Hyde 2001). Zhou and Hyde (2001)<br />

explored the literature on host-specific saprobes and came to the conclusion<br />

that it was hard to prove that saprobic fungi were host-specific. They introduced<br />

the terms host exclusivity and host recurrence as more suitable for use


with saprobic fungi. Host exclusivity is the exclusive occurrence of a strictly<br />

saprobic fungus on a particular host, while host recurrence is the frequent or<br />

predominant occurrence of a fungus on a particular host.<br />

The basis for host recurrence in saprobic fungi is interesting and several<br />

factors may be responsible.Wong and Hyde (2001) thought that host exclusive<br />

saprobes may be responding to differences in physical structure or nutrient<br />

levels of the potential hosts. There is also the possibility that enzyme production<br />

capabilities may influence whether a certain fungus can decay a certain<br />

host. However, recent studies have shown that most fungi can produce a wide<br />

range of enzymes capable of degrading simple sugars and cellulose (Lumyong<br />

et al. 2002). Fewer fungi can produce enzymes capable of digesting lignins<br />

(Leung and Pointing 2002). This would however, restrict fungi to lignified versus<br />

nonlignified <strong>plant</strong> tissues and is unlikely to be responsible for host recurrence<br />

(restricting fungi to certain <strong>plant</strong> species) as a large range of host tissues<br />

incorporate lignins into their tissues.<br />

Wong and Hyde (2001) studied the saprobes on six grass and one sedge<br />

species in Hong Kong and found that certain fungi showed host exclusivity or<br />

specificity. They hypothesized that these fungi may be host-specific endophytes<br />

that later become saprobes. There is much circumstantial evidence<br />

supporting this hypothesis and this has been discussed by Zhou and Hyde<br />

(2001) and Hyde (2001).<br />

8 Conclusions<br />

17 Fungal Endophytes 287<br />

There have now been many studies on the diversity and ecology of endophytes<br />

of grass and nongrass hosts in both tropical and temperate regions<br />

(Viret and Petrini 1994; Bussaban et al. 2001; Photita et al. 2001). The problem<br />

in most of these studies is that two uninformative groups of fungi are generally<br />

isolated; the first major group being typical endophytic genera such as<br />

Colletotrichum, Phomopsis and Phyllosticta while the second are mycelia sterilia.<br />

The first group are rarely recorded as saprobes on the host, although<br />

some may be pathogens. Therefore, the role of these fungi is puzzling and they<br />

may actually have no function. It is possible that the spores have landed on the<br />

<strong>plant</strong> <strong>surface</strong> and produced a germ tube which has penetrated the <strong>plant</strong><br />

stoma, but then cannot progress further due to <strong>plant</strong> defense. Future studies<br />

should, therefore concentrate on the role of these common endophytes, rather<br />

than provide uninformative lists with ecological data that have little consequence.<br />

The mycelia sterilia may be a more important group, but until we can find<br />

some way to identify more of them, it is impossible to elucidate their function.<br />

Methods need to be developed to stimulate these fungi to sporulate, or at least<br />

molecular techniques need to be refined in order to make identification simpler.<br />

Future studies should, therefore concentrate on developing these meth-


288<br />

Sita R. Ghimire and Kevin D. Hyde<br />

ods, rather than providing uninformative data on mycelia sterilia with ecological<br />

data that again have little consequence.<br />

Studies on endophytes have shown the beneficial roles of endophytic associations<br />

to the host as protection against mammals, resistance to insect herbivores<br />

and other pathogenic fungi, increased growth and development, nutrient<br />

uptake and stress tolerance in <strong>plant</strong>s including agriculturally important<br />

crops. The actual mechanisms involved for such phenomenon, however are<br />

poorly understood. The current understanding of secondary metabolites of<br />

host and endophyte origin and their interactions is limited. This area<br />

demands further studies which could lead to the discovery of novel compounds<br />

of biotechnological and agricultural importance. The mechanism by<br />

which a latent form of a pathogen turns pathogenic and vice-versa could be<br />

an interesting area of research of the highest <strong>plant</strong> pathological significance.<br />

References and Selected Reading<br />

Abbas JD, Hertrick BAD, Jurgenson JE (1996) Isolate specific detection of mycorrhizal<br />

fungi using genome specific primer pairs. Mycologia 88:939–946<br />

Agrios GN (1997) Plant pathology. Academic Press, London<br />

Bacon CW (1993) Abiotic stress tolerances (moisture and nutrients) and photosynthesis<br />

in endophyte-infected tall fescue. Agric Ecosyst Environ 44:123–141<br />

Bacon CW, White JF (1994) Stain, media and procedure for analyzing endophytes. In:<br />

Bacon CW, White JF (eds) Biotechnology of endophytic fungi of grasses, CRC Press,<br />

Boca Raton, pp 47–56<br />

Beck JJ, Ligon JM (1995) Polymerase chain reaction assays for the detection of<br />

Stagonospora nodorum and Septoria tritici in wheat. Phytopathol 85:319–324<br />

Bills GF (1996) Isolation and analysis of endophytic fungal communities from woody<br />

<strong>plant</strong>s. In: Redlin SC, Carris LM (eds) Endophytic fungi in grasses and woody <strong>plant</strong>s:<br />

systematic, ecology and evolution, APS Press, St. Paul, MN, pp 31–65<br />

Bills GF, Polishook JD (1992) Recovery of endophytic fungi from Chamaecyparis thyroides.<br />

Sydowia 44:1–12<br />

Boddy L, Griffith GS (1989) Role of endophytes and latent invasion in the development<br />

of decay communities in sapwood of angiospermous trees. Sydowia 41:41–73<br />

Bonito RD, Elliott ML, Jardin EAD (1995) Detection of the arbuscular mycorrhizal fungus<br />

in roots of different <strong>plant</strong>s species with the PCR.Appl Environ Microbiol 61:2809–<br />

2810<br />

Brunner F, Petrini O (1992) Taxonomic studies of Xylaria species and xylariaceous endophytes<br />

by isozymeelectrophoresis. Mycol Res 96:723–733<br />

Bultman TL, Murphy JC (2000) Do fungal endophytes mediate wound-induced resistance?<br />

In: Bacon CW, White JF (eds) Microbial endophytes, Marcel Dekker, New York<br />

Bussaban B, Lumyong, S, Lumyong P, McKenzie EHC, Hyde KD (2001) Endophytic fungi<br />

from Amomum siamense. Can J Microbiol 47:943–948<br />

Cabral D, Stone JK, Carroll G (1993) The internal mycobiota of Juncas sp.: microscopic<br />

and cultural observations of infection pattern. Mycol Res 97:367–376<br />

Carroll G (1986) The biology of endophytism in <strong>plant</strong>s with particular references to<br />

woody perennials. In: Fokkema NJ, Van den Heuvel J (eds) Microbiology of phyllosphere,<br />

Cambridge University Press, Cambridge, pp 205–222


17 Fungal Endophytes 289<br />

Carroll G (1988) Fungal endophytes in stems and leaves: from latent pathogen to mutualistic<br />

symbiont. Ecology 69:2–9<br />

Chambers SM, Sharples JM, Cairney JWG (1998) Towards a molecular identification of<br />

the Pisonia mycobiont. Mycorrhiza 7:319–321<br />

Cheplick GP, Clay K (1988) Acquired chemical defenses in grasses: The role of fungal<br />

endophytes. Oikos 52:309–318<br />

Clay K (1987) Effect of fungal endophytes on the seed and seedling biology of Lolium<br />

perenne and Festuca arundinaceae. Oecologia (Berlin) 73:358–362<br />

Clay K (1988) Clavicipitaceous fungal endophytes of grasses: co-evolution and change<br />

from parasitism to mutualism. In: Pyrozynski KA, Hawksworth DL (eds) Co-evolution<br />

of fungi with <strong>plant</strong>s and animals. Academic Press, New York, pp 79–105<br />

Clay K (1990) Fungal endophytes of grasses. Annu Rev Ecol Syst 21:275–297<br />

Clay K (1991) Fungal endophytes, grasses and herbivores. In: Barbosa P, Krischik VA,<br />

Jones CG (eds) Microbial mediation of <strong>plant</strong> herbivore interactions. Wiley, New York,<br />

pp 199–226<br />

Dahlman DL, Eichenseer H, Siegel MR (1991) Chemical perspective on endophyte grass<br />

interactions and their implications to insect herbivory. In: Barbosa P, Krischik VA,<br />

Jones CG (eds) Microbial mediation of <strong>plant</strong> herbivore interactions. Wiley, New York,<br />

pp 227–252<br />

De Bary A (1866) Morphologie und Physiologie der Pilze, Flechten, und Myxomyceten.<br />

Hofmeister’s Handbook of Physiological Botany. vol 2. Leipzig<br />

Doss PR, Welty RE (1995) A polymerase chain reaction based procedure for detection of<br />

Acremonium coenophialum in tall fescue. Phytopathol 85:913–917<br />

Espinosa-Garcia FJ, Langenheim JH (1990) The leaf fungal endophyte community of a<br />

coastal red wood population diversity and spatial patterns. New Phytol 116:89–97<br />

Fisher PJ,Anson AE, Pertini O (1984a) Antibiotic activity of some endophytic fungi from<br />

ericaceous <strong>plant</strong>s. Bot Helv 94:249–253<br />

Fisher PJ, Anson AE, Pertini O (1984b) Novel antibiotic activity of an endophyte Cryptosporiopsis<br />

sp. isolated from Vaccinium myrtillus. Trans Br Mycol Soc 83:145–148<br />

Fisher PJ, Pertini O, Petrini LE, Sutton, BC (1994) Fungal endophytes from leaves and<br />

twigs of Quercus ilex L. from England, Majorca and Switzerland. New Phytol 127:<br />

133–137<br />

Freeman S, Rodriguez RJ (1993) Genetic conversion of a fungal <strong>plant</strong> pathogen to a nonpathogenic,<br />

endophytic mutualist. Science 260:75<br />

Fröhlich J, Hyde KD (1999). Biodiversity of palm fungi in the tropics: are global fungal<br />

diversity estimates realistic? Biodivers Conserv 8:977–1004<br />

Fröhlich J, Hyde KD, Petrini O (2000) Endophytic fungi associated with palm. Mycol Res<br />

104:1202–1212<br />

Funk CR, Halisky PM, Johnson MC, Siegel MR, Stewart AV, Ahamad S, Hurley RH, Harvey<br />

IC (1983) An endophytic fungi and resistance to Sod Webworms: association of<br />

Lolium perenne L. Bio/technology April:189–191<br />

Gasoni L, Stegman De Gurfinkel B (1997) The endophyte Cladorrhinum foecundissimum<br />

in cotton roots: phosphorus uptake and host growth. Mycol Res 101:867–870<br />

Gotz M, Zornabach W, Boyle C (1993) Life cycle of Mycosphaerelle brassicicola (Duby)<br />

Lindau and ascospore production in vitro. J Phytopathol 139:298–308<br />

Guo LD, Hyde KD Liew ECY (1998) A method to promote sporulation in palm endophytic<br />

fungi. Fungal Divers 1:109–113<br />

Guo LD, Hyde KD, Liew ECY (2000) Identification of endophytic fungi from Livistona<br />

chinensis based on morphology and rDNA sequences. New Phytol 147:617–630<br />

Guo LD, Hyde KD, Liew ECY (2001) Detection and taxonomic placement of endophytic<br />

fungi within frond tissues of Livistina chinensis based on rDNA sequences. Mol Phylogenet<br />

Evol 20:1–13


290<br />

Sita R. Ghimire and Kevin D. Hyde<br />

Groppe K, Sanders I, Wiemken A, Boller T (1995) A microsatellite marker for studying<br />

the ecology and diversity of fungal endophytes (Epichloe spp.) in grasses. Appl Environ<br />

Microbiol 61:3943–3949<br />

Hammerli UA, Brandle UE, Petrini O, McDermott JM (1992) Differentiation of isolates of<br />

Discula umbrinella (teleomorph: Apiognomonia errabunda) from beech, chestnut<br />

and oak using RAPD markers. Mol Plant-Microbe Interact 5:479–483<br />

Hammon K.E, Faeth SH (1992) Ecology of <strong>plant</strong>-herbivore communities: a fungal component.<br />

Nat Toxins 1:197–208<br />

Hawksworth DL (1991) The fungal dimension of biodiversity: magnitude, significance,<br />

and conservation. Mycolog Res 95:641–655<br />

Hawksworth DL (2001) The magnitude of fungal diversity: the 1.5 million species estimate<br />

revisited. Mycol Res 105:1422–1432<br />

Hendry SJ, Boddy L, Lonsdale D (1993) Interaction between callus culture of European<br />

beech, indigenous ascomycetes and derived fungal extracts. New Phytol 123:421–428<br />

Hyde KD (2001) Where are the missing fungi? Does Hong Kong have the answers? Mycol<br />

Res 105:1514–1518<br />

Hyde KD, Lacap DC, Liew ECY (2001) An evaluation of the fungal ‘morphospecies’ concept<br />

based on ribosomal DNA sequences (Abstract). Phytopathology 91:S113<br />

Jersch S, Scherer C, Hutz G, Schlosser E (1989) Proanthocianidins as a basis for quiescence<br />

of Botrytis cinerea in immature strawberry fruits. Z Pflanzenkrankheiten<br />

Pflanzenschutz 96:365–378<br />

Johanson A, Jeger MJ (1993) Use of PCR for detection of Mycosphaerella fijiensis and M.<br />

musicola, the causal agent of Sigayoka leaf spot in banana and <strong>plant</strong>ain. Mycol Res<br />

97:670–674<br />

Kehr RD (1992) Pezicula cancer of Quercus rubra L., caused by Pezicula cinnamomea<br />

(DC) Sacc. II. Morphology and biology of the causal agent. Eur J For Pathol 22:29–40<br />

Kehr RD, Wulf A (1993) Fungi associated with above ground portion of declined oaks<br />

(Quercus rubra) in Germany. Eur J For Pathol 23:18–27<br />

Kimmons CA (1990) Nematode reproduction on endophyte infected and endophyte free<br />

tall fescue. Plant Dis 74:757–761<br />

Kulik MM (1988) Observations by scanning electron and brightfield microscopy on the<br />

mode of penetration of soybean seedlings by Phomopsis phaseoli. Plant Dis 72:115–<br />

118<br />

Latch GCM, Hunt WF, Musgrave DR (1985) Endophytic fungi affect growth of perennial<br />

rye grass. New Zealand J Agric Res 28:165–168<br />

Leung PC, Pointing SB (2002) Effect of different carbon and nitrogen regimes on Poly R<br />

decolorization by white rot fungi. Mycolog Res 106:86–92<br />

Lewis DH (1973) Concept in fungal nutrition and the origin of biotrophy. Biol Rev<br />

48:261–278<br />

Liew ECY, Guo LD, Ranghoo VM, Goh TK, Hyde KD (1998) Molecular approaches to<br />

assessing fungal diversity in the natural environment. Fungal Divers 1:1–17<br />

Lumyong S, Lumyong P, McKenzie EH, Hyde KD (2002) Enzymatic activity of endophytic<br />

fungi of six native seedling species from Doi Suthep-Pui National Park, Thailand. Can<br />

J Microbiol 48(12):1109–1112<br />

Ma LJ, Catramis CM, Rogers SO, Starmer WT (1997) Isolation and characterization fungi<br />

entrapped in glacial ice. Inoculum 48:23–24<br />

Matsushima T (ed) (1971) Microfungi of the Solomon Islands and Papua New Guinea.<br />

Matsushima, Kobe, Japan, pp 15–20<br />

McCutcheon TL, Carroll GC (1993) Genotypic diversity in populations of a fungal endophytes<br />

from Douglas fir. Mycologia 85:180–186<br />

Miller JD (1986) Toxic metabolites of epiphytic and endophytic fungi of conifers needles.<br />

In: Fokkema, NJ, Heuvel JVD (eds) Microbiology of phyllosphere. Cambridge<br />

University Press, Cambridge, pp 223–231


17 Fungal Endophytes 291<br />

Mills PR, Sreenivasaprasad S, Brown AE (1992) Detection and differentiation of Colletotrichum<br />

gloeosporioides isolates using PCR. FEMS Microbiol Lett 98:137–144<br />

Muirhead IF Deverall BJ (1981) Role of appressoria in latent infection of banana fruits by<br />

Colletotrichum musae. Physiol Plant Pathol 19:77–84<br />

O’Donnell J and Dickinson CH (1980) Pathogenicity of Alternaria and Cladosporium<br />

isolated on Phaseolus. Trans Br Mycol Soc 74:335–342<br />

Peters S, Aust AJ, Draeger S, Schulz B (1998) Interaction in dual cultures of endophytic<br />

fungi with host and non-host <strong>plant</strong> calli. Mycologia 90:360–367<br />

Petrini O (1986) Taxonomy of endophytic fungi of aerial <strong>plant</strong> tissue. In: Fokkema NJ,<br />

Van den Heuvel J (eds) Microbiology of phyllosphere. Cambridge University Press,<br />

Cambridge, pp 175–187<br />

Petrini O (1991) Fungal endophytes of tree leaves. In: Andrews J, Hirano S (eds) Microbila<br />

ecology of leaves, Springer, Berlin Heidelberg New York, pp 179–197<br />

Petrini O (1996) Ecological and physiological aspect of host specificity in endophytic<br />

fungi. In: Redlin SC, Carris LM (eds) Endophytic fungi in grasses and woody <strong>plant</strong>s.<br />

APS Press, St. Paul, MN<br />

Petrini O, Stone J, Carroll FE (1982) Endophytic fungi in evergreen shrubs in western<br />

Oregon: a preliminary study. Can J Botany 60:789–796<br />

Petrini O, Fisher PJ, Petrini LE (1992) Fungal endophytes of bracken (Pteridium aquilinum)<br />

with some reflections on their use in biological control. Sydowia 44:282–293<br />

Photita W, Lumyong S, Lumyong P, Hyde KD (2001) Endophytic fungi of wild banana<br />

(Musa acuminata) at Doi Suthep Pui National Park, Thailand. Mycol Res 105:1508–<br />

1514<br />

Prestidge RA, Gallagher RT (1988) Endophyte fungus confers resistance to rye grass:<br />

Argentine stem weevil larval studies. Ecol Entomol 13:429–435<br />

Ranghoo VM, Hyde KD, Liew ECY, Spatafora JW (1999) Family placement of Ascotaiwanian<br />

and Ascolacicola based on DNA sequences from the large subunit rRNA gene.<br />

Fungal Divers 2:159–168<br />

Rodrigues KF, Samuels GJ (1990) Preliminary study of endophytic fungi in tropical<br />

palm. Mycol Res 94:827–830<br />

Rodrigues KF, Leucthmann A, Petrini O (1993) Endophytic species of Xylaria: Cultural<br />

and isozymic studies. Sydowia 45:116–138<br />

Rollo F, Sassaroli S, Ubaldi M (1995) Molecular phylogeny of the fungi of the Iceman’s<br />

grass clothing. Curr Genet 28:289–297<br />

Rommert AK, Strack D, Aust HJ, Schulz B (1998) Verhalten sich Endophyten unter Nstress<br />

als Schwachenparasiten? Bielefelder Okol Beitr 14:307–311<br />

Schulz B, Wanke U, Draeger S and Aust HJ (1993) Endophytes from herbaceous <strong>plant</strong>s<br />

and shrubs: effectiveness of <strong>surface</strong> sterilization methods. Mycol Res 97:1447–1450<br />

Schulz B, Guske S, Dammann U, Boyle C (1998) Endophyte-host interactions. II. Defining<br />

symbiosis of the endophyte-host interaction. Symbiosis 25:213–227<br />

Schulz B, Rommert AK, Dammann U, Aust HJ, Strack D (1999) The endophyte-host<br />

interaction: a balanced antagonism? Mycol Res 103:1275–1283<br />

Sieber TN, Sieber-Canavesi F, Dorworth CE (1991) Endophytic fungi of red alder (Alnus<br />

rubra) leaves and twigs in British Colombia. Can J Bot 69:407–411<br />

Siegel MR, Johnson M.,Varney DR, Nesmith WC, Buckner RC, Bush LP, Burrus PB (1984)<br />

A fungal endophyte in tall fescue: incidence and dissemination. Phytopathology<br />

74:932–937<br />

Sinclair JB, Cerkauskas RF (1996) Latent infection vs. endophytic colonization by fungi.<br />

In: Redlin SC, Carris LM (eds) Endophytic fungi in grasses and woody <strong>plant</strong>s: systematics,<br />

ecology and evolution. APS Press, St. Paul, pp 3–29<br />

Stone JK (1988) Fine structure of latent infection by Rhabdocline parkeri on Douglas fir,<br />

with observation on uninfected epidermal cells. Can J Bot 66:45–54


292<br />

Sita R. Ghimire and Kevin D. Hyde<br />

Stone JK, Viret O, Petrini O, Chapela IH (1994) Histological studies of host penetration<br />

and colonization by endophytic fungi. In: Pertini O, Ouellette G (eds) Host wall alterations<br />

by parasitic fungi. APS Press, St. Paul, pp 115–126<br />

Suske, J, Acker G (1989) Identification of endophytic hyphae of Lophodermium piceae in<br />

tissues of green, symptomless Norway spruce needles by immunoelectron microscopy.<br />

Can J Bot 67:1768–1774<br />

Taylor JE, Hyde KD, Jones EBJ (1999) Endophytic fungi associated with the temperate<br />

palm Trachycarpus fortunei within and outside its natural geographical range. New<br />

Phytol 142:335–346<br />

Tyler BM (1993) To kill or not to kill: the genetic relationship between a parasite and<br />

endophyte. Trends Microbiol 1:252–254<br />

Verma A,Verma S, Sudha, Sahay N, Butehorn B, Franken P (1999) Piriformospora indica,<br />

a cultivable plat growth promoting root endophyte. Appl Environ Microbiol 65:2741–<br />

2744<br />

Viret O, Petrini O (1994) Colonization of beech leaves (Fagus sylcatica) by the endophyte<br />

Discula umbrinella (teleomorph, Apiognomonia errabunda). Mycol Res 98:423–432<br />

Viret O, Scheidegger C, Pertini O (1993) Infection of beech leaves (Fagus sylcatica) by the<br />

endophyte Discula umbrinella (teleomorph, Apiognomonia errabunda) – low temperature<br />

scanning electron microscopy studies. Can J Bot 71:1520–1527<br />

Williamson B (1994) Latency and quiescence in survival and success of fungal <strong>plant</strong><br />

pathogen. In: Blackman JP, Williamson B (eds) Ecology of <strong>plant</strong> pathogens. CAB<br />

International, London, pp 187–207<br />

Wilson D (1995) Endophyte – the evolution of term, a classification of its use and definition.<br />

Oikos 73:274–276<br />

Wong MKM, Hyde KD (2001) Diversity of fungi on six species of Gramineae and one<br />

species of Cyperaceae in Hong Kong. Mycol Res 105:1485–1491<br />

Zhang W, Wildel JF, Clark LG (1997) Bamboozled again! Inadvertent isolation of fungal<br />

rDNA sequences from bamboos (Poaceae: Bambusoideae). Mol Phylogenet Evol<br />

8:205–217<br />

Zhou DQ, Hyde KD (2001) Host-specificity, host-exclusivity and host-recurrence in<br />

saprobic fungi. Mycol Res 105:1449–1457


18 Mycorrhizal Development and Cytoskeleton<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

1 Introduction<br />

The formation of mycorrhiza requires morphological changes, both in the<br />

<strong>plant</strong> cells and fungal hyphae, necessary for the development and maintenance<br />

of the signal and nutrient exchange at <strong>plant</strong> fungal interfaces. It is well<br />

known that cytoskeletal elements play a central role both in the morphogenesis<br />

of <strong>plant</strong> root cells (Barlow and Baluška 2000) and of fungal hyphae (Raudaskoski<br />

et al. 2001). In the present review, the <strong>plant</strong> and fungal genes encoding<br />

the structural proteins of main cytoskeletal elements, microtubules (MTs)<br />

and microfilaments (MFs), are described. Some speculations of the functional<br />

significance of cytoskeletal rearrangements observed in <strong>plant</strong> cells and fungal<br />

hyphae at the formation of endo- and ectomycorrhiza are presented. The<br />

reorganization of the cytoskeleton results from interactions with proteins that<br />

serve by themselves as targets for intra- and extracellular signal mediating<br />

pathways (Johnson 1999; Kost et al. 1999b). The presence of such pathways in<br />

mycorrhiza is discussed. The different phases in the cell cycle also requires<br />

rearrangements in the cytoskeleton (Mews et al. 1997; John et al. 2001). This<br />

aspect is shortly discussed in association with known effects of mycorrhiza<br />

on the <strong>plant</strong> cell cycle. Finally, the general methods used in visualization of<br />

cytoskeletal components are shortly introduced.<br />

2 Cytoskeletal Components<br />

The cytoskeleton is composed of filamentous structures whose arrangements<br />

are continuously changing in living cells in response to different developmental<br />

and environmental cues. In <strong>plant</strong> and fungal cells there are two main<br />

cytoskeletal proteins: actin and tubulin. Actin monomers polymerize to thin<br />

filaments known as MFs or actin filaments. Tubulin polymerizes to MTs. Both<br />

MFs and MTs are polarized structures with minus and plus ends. The minus<br />

end is often attached to some subcellular structure while the plus end is<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


294<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

mainly untouched and is able to polymerize and depolymerize more freely<br />

than the minus end. The polarity of the cytoskeletal elements is well studied<br />

in animal cells while in <strong>plant</strong> cells and fungal hyphae, their polarity is known<br />

only in a few cases (Euteneuer and McIntosh 1980).<br />

In eukaryotic cells, MTs participate in cell division, cell shape changes, cell<br />

motility and intracellular organelle trafficking as well as in cell wall synthesis<br />

(Wasteneys 2000; Lloyd and Hussey 2001). Actin cytoskeleton is involved in<br />

many different developmental processes including the establishment of cell<br />

polarity and tip growth (Hepler et al. 2001; Raudaskoski et al. 2001), division<br />

plane determination, cell elongation, positioning of proteins on membranes<br />

and cytoplasmic streaming (Meagher et al. 1999). Recently, connections have<br />

been revealed between MT and actin cytoskeleton mediated by interaction<br />

between MF- and MT-associated motor molecules or scaffold proteins<br />

(Goode et al. 2000). Thus, the reorganization in one cytoskeletal component<br />

may also lead to the reorganization of the other.<br />

2.1 Expression of Tubulin-Encoding Genes<br />

MTs in eukaryotic cells are composed of polymerized a- and b-tubulin heterodimers.<br />

A less abundant form, g-tubulin occurs also in eukaryotic cells<br />

(Liu et al. 1993). g-Tubulin functions in MT organizing centers in centrosomes<br />

in animal cells (Joshi et al. 1992) and in spindle pole bodies in fungi (Oakley<br />

et al. 1990). Higher <strong>plant</strong>s have no discrete MT organizing centers and g-tubulin<br />

appears to be dispersed around the cells (Joshi and Palevitz 1996). Plant aand<br />

b-tubulin gene families consist of five to ten genes while in fungi the<br />

number is much lower with only one or two family members (Joyce et al. 1992;<br />

Kopczak et al. 1992; Snustad et al. 1992; Villemur et al. 1992, public databases).<br />

Analysis of tubulin gene expression, mainly done in Arabidopsis and maize,<br />

has shown that transcripts of tubulin genes occur in all <strong>plant</strong> tissues, but their<br />

accumulation can be specific for different <strong>plant</strong> organs or developmental<br />

stages (Montoliu et al. 1989; Hussey et al. 1990; Joyce et al. 1992; Villemur et al.<br />

1994) or induced by environmental factors (Kerr and Carter 1990a). Tubulin<br />

expression studies in mycorrhizal <strong>plant</strong>s are of special interest, since the<br />

invading fungus alters the expression pattern of tubulins known from uninfected<br />

root (Bonfante et al. 1996).<br />

In maize it has been shown that the accumulation of the transcripts from<br />

all six a-tubulin genes is relatively high in the root tip, but low in the root cortex<br />

(Joyce et al. 1992). More detailed analyses at cellular level have shown that<br />

in the maize root the preferential expression of tua1 and tua3 genes occurs in<br />

root meristem cells differentiating into cortex and vascular tissue, respectively.<br />

The transcripts of tua2 gene accumulate in maize root epidermis<br />

(Uribe et al. 1998), while tua4 transcripts are mainly expressed in root vascular<br />

tissue (Joyce et al. 1992). In agreement with the idea that a mycorrhizal


18 Mycorrhizal Development and Cytoskeleton 295<br />

fungus might affect the expression pattern of tubulin genes is the observation<br />

that tua3 transcripts increased in the differentiated cortical cells at the invasion<br />

of an endomycorrhizal fungus. Similarly, in the transgenic tobacco, in<br />

which the Gus expression took place under the promoter of maize tubulin<br />

gene tua3, Gus activity occurred in differentiated cortical cells infected by<br />

endomycorrhizal fungus (Bonfante et al. 1996).As with a-tubulins the expression<br />

of Arabidopsis b-tubulins is relatively high in the root tip and vascular<br />

tissue, but low in root cortical cells (Villemur et al. 1994). The tub6 and tub8<br />

genes are preferentially expressed in the root tip and vascular cylinder while<br />

the high expression of tub4 gene seems to be a unique feature for vascular tissue<br />

(Villemur et al. 1994). It has not been investigated whether the formation<br />

of endomycorrhiza is affecting the expression pattern of b-tubulin genes.<br />

Until now, Eucalyptus globulus is the only ectomycorrhiza forming <strong>plant</strong> in<br />

which tubulin expression during ectomycorrhiza formation has been studied<br />

at the RNA level. An a-tubulin gene was shown to be upregulated during formation<br />

of symbiosis, and the upregulation of a-tubulin expression paralleled<br />

the increased formation of lateral roots in Eucalyptus seedlings that were in<br />

contact with the fungal mycelium (Diaz et al. 1996). In Pinus sylvestris- Suillus<br />

bovinus and P. contorta–S. variegatus ectomycorrhiza the expression of tubulins<br />

has been analyzed at the protein level (Timonen et al. 1993, 1996; Niini et<br />

al. 1996). Different mobility of <strong>plant</strong> and fungal a-tubulin allowed their comparison<br />

in one-dimensional (1-D) immunoblots, which suggested that in<br />

mature ectomycorrhiza the fungal a-tubulin dominated (Timonen et al.<br />

1996). The comparison of the amount of <strong>plant</strong> and fungal a-tubulin during<br />

the development of P. contorta–S. variegates ectomycorrhiza for 60 days also<br />

indicated that the amount of <strong>plant</strong> a-tubulin decreased gradually, probably<br />

due to the development of fungal sheath around the root (Timonen et al.<br />

1996). No such comparisons could be made between <strong>plant</strong> and fungal b-tubulin<br />

or actin due to their similar mobility during the electrophoretic separation.<br />

The immunoblots of two-dimensional gels from three root types of P.<br />

sylvestris radicles, main root and first order laterals and short roots as well as<br />

from different developmental stages of P. sylvestris–S. bovinus ectomycorrhiza<br />

revealed more differences in the tubulin protein patterns than 1-D<br />

immunoblots (Niini et al. 1996). Three <strong>plant</strong> a-tubulins were detected in all<br />

root types, but the pattern in the short roots differed from that in radicles and<br />

first-order laterals. This is an interesting observation, since the formation of<br />

ectomycorrhiza occurs in Pinus short roots probably due to their reduced<br />

growth rate that could be associated with the occurrence of the short-rootspecific<br />

a-tubulin pattern. During the development of ectomycorrhiza the initial<br />

short root-specific a-tubulin pattern gradually changed and two new<br />

a-tubulins were distinguished in mature ectomycorrhiza. Whether the atubulin<br />

protein patterns in P. sylvestris short roots and ectomycorrhiza results<br />

from alterations in the expression of a-tubulin genes or post-transcriptional


296<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

modifications of a-tubulin proteins, will be resolved when the number and<br />

the degree of transcript accumulation of a-tubulin encoding genes in P.<br />

sylvestris short roots have been clarified. In the immunoblots from the different<br />

root types and ectomycorrhiza, three different b-tubulins were identified.<br />

For the expression of P. sylvestris b-tubulins no alterations comparable to<br />

those in a-tubulin were observed.<br />

The expression of the tubulin genes in endo- and ectomycorrhizal fungi<br />

has also obtained some attention. Fragments encoding 267 amino acids from<br />

the central part of the b-tubulin gene have been cloned from several endomycorrhizal<br />

fungi including Glomus mossae, G. geosporum, G. coronatum, G.<br />

clarum, Gigaspora rosea, Acaulospora laevis, andScutellospora castanea (M.<br />

Stommel and P. Franken, public databases). The deduced amino acid<br />

sequences of the fragments suggest clearly that there are at least two b-tubulin<br />

encoding genes in the endomycorrhizal fungi. Comparison of the deduced<br />

amino acid sequences of the b-tubulin genes in and between different species<br />

indicates that in each species the b-tubulin amino acid sequences are more<br />

similar to b-tubulins of other species than to those within the same species.<br />

The G. rosea b-tubulin transcripts accumulate in dormant and germinated<br />

spores, in extraradical hyphae and in pea mycorrhiza (Franken et al. 1997;<br />

Bütehorn et al. 1999), while a- and b-tubulin protein was detected in the<br />

hyphae of G. mossae elicited by the host <strong>plant</strong> (Åström et al. 1994). None of<br />

these experiments have yet shown whether the expression of either the btubulin<br />

gene is associated with the formation of endomycorrhiza or some<br />

other specific stage in the life cycle of the endomycorrhizal fungi.<br />

The immunoblots of a- and b-tubulins from the ectomycorrhizal fungus S.<br />

bovinus and from its ectomycorrhiza with P. sylvestris indicated the presence<br />

of three a- and two b-tubulin polypeptides (Niini et al. 1996) in nonsymbiotic<br />

hyphae and ectomycorrhiza. From the filamentous homobasidiomycete<br />

Schizophyllum commune that is closely related to S. bovinus,three a- and two<br />

b-tubulins have also been identified by 2-D gel electrophoresis. Until now,<br />

only two a- and one b-tubulin encoding genes have been isolated from S.<br />

commune in spite of several attempts (Russo et al. 1992; Raudaskoski unpublished<br />

data). The higher number of polypeptides than tubulin encoding genes<br />

suggests that fungal a- and b-tubulins are targets for posttranslational modifications.<br />

Recently, a b-tubulin encoding cDNA highly similar to that of S.<br />

commune has been isolated from S. bovinus. By using the encoding region of<br />

the S. bovinus b-tubulin gene as a probe, a high, but similar amount of b-tubulin<br />

mRNAs were detected both in nonsymbiotic and symbiotic hyphae (Lahdensalo<br />

et al., unpublished). Both in vegetative and ectomycorrhizal hyphae of<br />

S. bovinus the tubulin polypeptides occurred in doublet patterns (Niini et al.<br />

1996), which are thought to be due to allelic differences between tubulins of<br />

the haploid genomes present in the dikaryotic hyphae of S. bovinus. This<br />

needs to be certified by cloning and further analysis of S. bovinus tubulin<br />

genes.


2.2 Expression of Actin-Encoding Genes<br />

18 Mycorrhizal Development and Cytoskeleton 297<br />

Arabidopsis is known to have ten actin genes, two of which are pseudogenes<br />

(Meagher et al. 1999). Out of the eight expressed actin genes ACT2, ACT8,and<br />

ACT7 are named Arabidopsis vegetative actin genes due to the accumulation<br />

pattern of transcripts. ACT2 is expressed in young and old vegetative tissue,<br />

but ACT8 only in a subset of the organs and tissues expressing ACT2 (An et al.<br />

1996a). ACT7 is expressed in young expanding vegetative tissues and is also<br />

involved in phytohormone responses (Kandasamy et al. 2001). The rest of the<br />

Arabidopsis actin genes appear to be associated with reproductive processes<br />

(An et al. 1996b; Huang et al. 1997; Meagher et al. 1999). In spite of the high<br />

number of actin genes in most <strong>plant</strong> species and the important cell biological<br />

functions of the actin cytoskeleton, only a few analyses about the expression<br />

of different actin genes in root tissue have been performed (McLean et al.<br />

1990) and the effect of mycorrhiza on the expression of actin genes at mRNA<br />

level has not been investigated.<br />

One-dimensional analyses of actin expression at the protein level have<br />

been performed in Pinus ectomycorrhiza. The similar mobility of <strong>plant</strong> and<br />

fungal actins in the immunoblots of 1-D gels from P. sylvestris- S. bovinus<br />

ectomycorrhiza made it difficult to record the contribution of each symbiotic<br />

partner to the actin signal (Timonen et al. 1993). The presence of the actin signal<br />

throughout the different developmental stages of ectomycorrhiza was suggested<br />

to be a reliable marker for still metabolically active symbiosis (Timonen<br />

et al. 1996).<br />

In the immunoblots of 2-D gels four actin polypeptides were detected in P.<br />

sylvestris radicles, main roots and first order laterals and in short roots. The<br />

polypeptides were also detected in P. sylvestris- S. bovinus young and dichotomous<br />

mycorrhizal short roots. In fully mature coralloid mycorrhiza only two<br />

actin polypeptides occurred. Whether they represented <strong>plant</strong> or fungal actin<br />

or both was not possible to conclude. The presence of four actin polypeptides<br />

in Pinus root tissues is in agreement with the occurrence of a similar number<br />

of actin polypeptides in Vicia faba roots (Janssen et al. 1996), while in the<br />

roots of Phaseolus vulgaris one and two actin polypeptides were detected in<br />

symbiotic root nodules and in uninfected roots, respectively (Pérez et al.<br />

1994).<br />

In the immunoblots of 2-D gels of the vegetative hyphae of S. bovinus two<br />

actin polypeptides occurred that were also detected in ectomycorrhiza (Niini<br />

et al. 1996). Recently, two actin-encoding genes, Sbact1 and Sbact2,were isolated<br />

from S. bovinus nonsymbiotic hyphae (Tarkka et al. 2000). Northern<br />

hybridization with specific probes for each actin gene of S. bovinus indicated<br />

that both actins are expressed in vegetative hyphae and ectomycorrhiza. In<br />

vegetative hyphae, the expression rate and protein level of Sbact1 was tenfold<br />

higher than that of Sbact2. A ten times higher accumulation of Sbact1 than<br />

Sbact2 mRNA was also observed in ectomycorrhizal hyphae, although the


298<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

analyzed sample represented a pool of young and mature ectomycorrhizal<br />

roots. In the future, the analysis of different developmental stages of ectomycorrhiza<br />

might improve our understanding of the expression pattern of the S.<br />

bovinus actin genes in symbiosis. From S. commune, a nonectomycorrhizal<br />

homobasidiomycete closely related to S. bovinus, two actin-encoding genes<br />

were also isolated (Tarkka et al. 2000), which indicates that filamentous<br />

homobasidiomycetes differ from filamentous deutero- and ascomycetes, having<br />

only a single actin gene. Recently, two actin-encoding genes have also<br />

been identified in the genome sequence of Schizosaccharomyces pombe<br />

(Wood et al. 2002).<br />

3 Organization of Cytoskeleton in Endomycorrhiza<br />

3.1 Root Cells<br />

Indirect immunofluorescence (IIF) microscopy and related methods have been<br />

used to study the structure of the cytoskeleton in nonmycorrhizal and<br />

endomycorrhizal root cells of tobacco (Genre and Bonfante 1997, 1998),<br />

Asparagus (Matsubara et al. 1999), Medicago truncatula (Blancaflor et al.<br />

2001), and in protocorms of orchid seeds (Uetake et al. 1997; Uetake and<br />

Peterson 1997,1998). Protocorms develop at the base of germinating orchid<br />

seeds and invasion of the parenchyma cells by a symbiotic fungus is necessary<br />

for the further development of the embryo. Several common features were<br />

observed in the reorganization of MT cytoskeleton in roots and protocorms<br />

after invasion of the symbiotic fungus into the <strong>plant</strong> cells. In all three cases the<br />

fungus invades differentiated parenchyma cells containing mainly transversely<br />

orientated cortical (below the plasma membrane) MTs connected with<br />

a few cytoplasmic MTs to the nucleus. After hyphal penetration, the plasma<br />

membrane separating the fungal hyphae from the <strong>plant</strong> cell cytoplasm, the<br />

perifungal membrane (Uetake and Peterson 1998), expands to follow the<br />

branching of the fungal hyphae. The growth of the hyphae in the intracellular<br />

space leads to formation of an arbuscule in endomycorrhiza and a peloton of<br />

hyphal coils in orchid mycorrhiza. The hyphal growth is associated with profound<br />

reorganization of MT cytoskeleton in the <strong>plant</strong> cell (Uetake et al. 1997;<br />

Uetake and Peterson 1997; Genre and Bonfante 1997, 1998; Matsubara et al.<br />

1999; Blancaflor et al. 2001). During fungal invasion the cortical MTs of the<br />

<strong>plant</strong> cell disappear probably through depolymerization, and new MTs, less<br />

well orientated, reappear at the plasma membrane surrounding the intracellular<br />

hyphae.<br />

The signals and mechanisms behind the observed reorganization of MT<br />

cytoskeleton in <strong>plant</strong> cells colonized by endomycorrhizal fungi are not yet<br />

known. However, it can be speculated that the invasion and proliferation of<br />

fungal hyphae in the space between cell wall and plasma membrane of a dif-


18 Mycorrhizal Development and Cytoskeleton 299<br />

ferentiated <strong>plant</strong> parenchyma cell with full turgor pressure could cause<br />

mechanical stress (Ko and McCulloch 2000; Stamatas and McIntire 2001) at<br />

the <strong>plant</strong> cell membrane, perhaps associated with chemical signals from the<br />

fungus to the <strong>plant</strong> cell. These signals could cause the depolymerization of<br />

MTs at the plasma membrane surrounding the cell and direct MT repolymerization<br />

at the expanding plasma membrane surrounding the growing hyphae,<br />

together with altered gene expression of the invaded parenchyma cells. In line<br />

with this idea is the observation that in transgenic tobacco the GUS expression<br />

under the promoter of the maize tubulin gene Tuba3 increased in differentiated<br />

cortical cells infected by endomycorrhizal fungus (Bonfante et al.<br />

1996). The accumulation of Tuba3 transcripts was similarly observed to<br />

increase in maize cortical cells during the invasion of the cells by an endomycorrhizal<br />

fungus. Cell wall material is deposited into the extracellular space<br />

between the <strong>plant</strong> cell membrane and fungal hyphae (Peterson et al. 1996),<br />

which could require the presence of MTs as these are required for cell plate<br />

formation in meristems (Lloyd and Hussey 2001), primary wall formation in<br />

elongating cells (Wasteneys 2000), and secondary wall thickenings in differentiating<br />

cells (Chaffey et al. 2000). In addition, proteins necessary for the<br />

nutrient exchange between the symbionts probably are synthesized and<br />

transported to the perifungal membrane (Rosewarne et al.1999; Hahn and<br />

Mendgen 2001), which could also require transport along the MTs.<br />

The distribution of MFs has also been studied in endomycorrhiza formed<br />

in tobacco roots (Genre and Bonfante 1998) and in protocorm cells (Uetake<br />

and Peterson 1997). In noninfected cells, MFs appeared to have the distribution<br />

reported for parenchyma cells in a large number of <strong>plant</strong>s investigated<br />

(Staiger 2000), with thin and thick MF cables crossing the cell cytoplasm. At<br />

fungal invasion, reorganization of MFs were observed in cortical cells of<br />

tobacco, in which the cables disappeared and MFs seemed to become tightly<br />

associated with the plasma membrane surrounding the arbuscular branches<br />

(Genre and Bonfante 1998). In protocorm cells, no clear reorganization of<br />

MFs was observed, but the distribution of MFs was comparable to that in<br />

uninfected cells (Uetake and Peterson 1997), which was a result quite different<br />

from the reorganization of MTs in the protocorm cells at fungal invasion. The<br />

different behavior of MFs in tobacco and protocorm cells at fungal invasion is<br />

suggested to be due to the physiological difference between the endomycorrhizal<br />

and orchid endosymbiotic fungus (Genre and Bonfante 1997), or it<br />

could result from differences in the processing of the cells for confocal microscopic<br />

investigation (Uetake and Peterson 1997). In tobacco root cells, the<br />

accumulation of MFs in close association with the plasma membrane surrounding<br />

the fungus is suggested to be due to the involvement of the actin<br />

cytoskeleton in localization of proteins necessary for membrane transport<br />

and signal transduction between symbionts (Genre and Bonfante 1997). Noteworthy<br />

is that at the invasion of the <strong>plant</strong> cell by the endomycorrhizal fungus,<br />

the <strong>plant</strong> cell nucleus moves from the periphery of the cell to the center and


300<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

the central vacuole becomes fragmented (Bonfante and Perotto 1995). These<br />

processes could also be controlled by rearrangements of MTs or MFs, or by<br />

both cytoskeletal elements.<br />

3.2 Fungal Hyphae<br />

In the research of cytoskeleton in endomycorrhiza, the structure and function<br />

of fungal MTs and MFs have gained less attention than those of <strong>plant</strong> cells. In<br />

the IIF microscopical investigation of nonsymbiotic hyphae of Glomus<br />

mossae, the MTs were visualized with tubulin and MFs with actin antibodies<br />

in the multinucleate hyphae originating from germinated spores (Åström et<br />

al. 1994). MTs extended in the cortical and central parts of the hyphae to the<br />

extreme hyphal tip, continued from the main hypha into a branch and the<br />

position of nuclei appeared to follow the MT tracks. Long MFs were also visualized<br />

in the hyphae (Åström et al. 1994). MTs and MFs could be involved in<br />

the intra- and intercellular nuclear movements and cytoplasmic streaming,<br />

respectively, recorded in living nonsymbiotic hyphae of different endomycorrhizal<br />

fungi (Bago et al. 1998; Giovannetti et al. 1999). The presence of both<br />

MTs and MFs in nonsymbiotic hyphae suggests that these structures could<br />

also play a significant role in hyphal morphogenesis associated with the formation<br />

of endomycorrhiza, such as the differentiation of the appressorium at<br />

the root <strong>surface</strong> at the beginning of the symbiosis and the formation of vesicular<br />

and arbuscular structures in the <strong>plant</strong> cell after the establishment of the<br />

symbiosis.<br />

4 Organization of Cytoskeleton in Ectomycorrhiza<br />

4.1 Root Cells<br />

The effect of ectomycorrhiza formation on the <strong>plant</strong> cell cytoskeleton is more<br />

difficult to investigate than that of endomycorrhiza or orchid mycorrhiza. In<br />

the ectomycorrhizal symbiosis, the fungal hyphae grow between the cortical<br />

cells of the host <strong>plant</strong>, forming a hyphal network for nutrient exchange called<br />

the Hartig net. The <strong>plant</strong> cells of the Hartig net have thick cell walls and accumulations<br />

of secondary metabolites such as phenols and starch. The thick cell<br />

walls inhibit rapid penetration of fixatives necessary for preservation of<br />

cytoskeletal elements, which is seen as lack of MTs or MFs from the published<br />

ultrastructural studies of ectomycorrhiza. Autofluorescence of secondary<br />

metabolites hampers the recording of cytoskeletal structures when they have<br />

been preserved during fixation. In spite of these difficulties, some knowledge<br />

of cytoskeletal structure has been obtained in Pinus sylvestris–Suillus bovinus<br />

ectomycorrhiza (Timonen et al. 1993; Niini and Raudaskoski 1998).


18 Mycorrhizal Development and Cytoskeleton 301<br />

P. sylvestris has a root system consisting of three morphologically and<br />

anatomically different root types, which is also found in other pines as well as<br />

in eucalypts and beeches (Smith and Read 1997). The primary root or the<br />

main root has an undetermined capacity for continuous growth, the lateral<br />

roots have a somewhat limited ability to elongate, and the so-called short<br />

roots have a very limited ability to grow. In order to survive, they have to be<br />

colonized by a symbiotic fungus (Robertson 1954; Wilcox 1968). In some<br />

pines, i.e., P. sylvestris and P. strobus, the mycorrhiza only forms in the short<br />

roots (Piché et al. 1983). In Scots pine seedlings, two types of short roots are<br />

distinguished: one type consists of long and slender roots with a high number<br />

of root hairs, the other of truncated, robust roots with a round apex and only<br />

a few root hairs. The majority of the short roots of pine seedlings belong to the<br />

latter type and mycorrhiza is only formed in this root type (Niini and Raudaskoski<br />

1998).<br />

The IIF microscopical studies on the MT cytoskeleton in nonmycorrhizal<br />

and mycorrhizal short roots have indicated some common features. The most<br />

prominent MT cytoskeleton is detected in meristematic and vascular tissue.<br />

In the meristems of truncated short roots the MTs are often vertical in interphase<br />

cells and mitotic spindles are horizontally oriented. These features suggest<br />

that in the meristem the cells elongate and divide horizontally which<br />

probably is the reason for the blunt form of the tip in the truncated short roots<br />

(Niini and Raudaskoski 1998). Almost next to the meristem in short roots<br />

occur xylem elements with cell wall thickenings and cortical cells with amyloplasts,<br />

which indicates that cell differentiation takes place very close to the<br />

short root tip. In the topmost cell layer of the meristem the direction of cell<br />

divisions is no longer horizontal, but oblique or vertical. From the central part<br />

of this layer originate vertically elongated cells with horizontally orientated<br />

MTs, and from the borders cells with amyloplasts representing differentiating<br />

vascular and cortical tissue, respectively. Thus, in short roots divisions in only<br />

one cell layer seem to provide initials for the elongation and differentiation<br />

zones while in lateral roots the transition zone between the meristem and<br />

elongation zone appears to consist of four to five cell layers with vertical cell<br />

divisions producing initials for cell elongation and differentiation. This must<br />

affect the growth rate of the roots and could explain why the elongation of the<br />

short roots is retarded and tissue maturation occurs closer to the meristem in<br />

the short roots than in the laterals.<br />

In nonmycorrhizal short roots, very few MTs are detected in the cortical<br />

cells for which a high number of amyloplasts is typical (Fig. 1A). In ectomycorrhizal<br />

roots, MT fluorescence is only observed in association with the<br />

nucleus in cortical cells (Fig. 1B). The low number or absence of MTs from<br />

cortical cells seems to be a specific feature of Pinus short roots. Although the<br />

tubulin expression in cortical cells of Arabidopsis (Villemur et al. 1994), and<br />

maize (Joyce et al. 1992), roots is low, MTs are always detected at the inner<br />

face of the plasma membrane in uninfected cortical cells of these <strong>plant</strong>s. It


302<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini


18 Mycorrhizal Development and Cytoskeleton 303<br />

is possible that the antibody used for IIF microscopical detection of MTs in<br />

Pinus is not able to recognize the MTs of cortical cells although it visualizes<br />

well MTs in meristem and vascular tissue. The MT cytoskeleton of cortical<br />

cells of Pinus could also be more sensitive to the processing of the samples<br />

for IIF microscopy than the MTs in meristem and vascular tissue. Interestingly,<br />

it has not been possible to visualize MFs in the cortical cells of short<br />

roots although plenty of MFs are seen in vascular tissue, where they were<br />

already visualized in the early days of the study of MFs in <strong>plant</strong> cells (Pesacreta<br />

et al. 1982).<br />

The structure of cell wall and cytoskeleton in differentiating cortical cells<br />

in short roots of pine is of special interest, since this is the region of the short<br />

root in which the ectomycorrhizal fungus invades and establishes the Hartig<br />

net. When root morphogenesis and ectomycorrhiza formation in Scots pine<br />

was studied (Niini et al. 1996; Tarkka et al. 1998), a group of polypeptides with<br />

molecular weight slightly above 43 kDa were observed to be short root-specific.<br />

By using peptide sequencing, it was shown that the polypeptides represented<br />

a group of peroxidases. By reverse genetics a full-length cDNA of one<br />

of the peroxidases, Psyp1, was cloned and sequenced (Tarkka et al. 2001). The<br />

signal sequence suggests that Psyp1 is secreted and could be involved in cell<br />

wall formation. In ectomycorrhiza Psyp1 expression is downregulated, which<br />

agrees with the idea that the growth of the fungal hyphae in the intercellular<br />

space might inhibit the cortical cell wall differentiation (Niini 1998). There<br />

may be signalling or linkages between adjacent <strong>plant</strong> cells that can regulate<br />

the organization of their cytoskeletal structures. This exchange of information<br />

might be mediated through plasmodesmata, or alternatively, through the<br />

intervening cell wall (Canut et al. 1998; Overall et al. 2001).<br />

Fig. 1. Microtubule cytoskeleton visualized with indirect immunofluorescence technique<br />

with a-tubulin antibody and viewed with laser scanning confocal microscopy in<br />

Pinus sylvestris short root (A) and ectomycorrhiza with Suillus bovinus (B). A In cortex<br />

only few microtubules are distinguished in the cortical cells with numerous round amyloplasts.<br />

In stele microtubules with mainly transverse orientation are abundant in elongating<br />

cells differentiating to vascular tissue. Strong vertical bands represent wall thickenings<br />

in a xylem cell. B Microtubules are hardly seen in pine cortical cells, but they are<br />

clearly distinguished as long tracks in hyphae forming the fungal sheath and penetrating<br />

into the root cortex. A,B Bars 20 mm. C–E Cytoskeletal elements in Suillus bovinus<br />

hyphae visualized with rhodamine-phalloidin staining of actin (C, D) and indirect<br />

immunofluorescence microscopy with a-tubulin antibody (E). C A strong actin signal at<br />

hyphal tip, D an actin ring at the site of the future septum. E Microtubule tracks in a<br />

hyphal branch. C–E Bars 10 mm


304<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

4.2 Fungal Hyphae<br />

An extensive MT cytoskeleton with strictly axial orientation is observed in the<br />

long polarized apical cells of ectomycorrhiza-forming and other filamentous<br />

fungi (Salo et al. 1989; Raudaskoski et al. 1991; Niini and Raudaskoski 1998;<br />

Raudaskoski et al. 2001). In the dikaryotic hyphae of these fungi the longitudinally<br />

running MTs (Fig. 1E) appear to keep the two nuclei with different<br />

mating type genes close to each other by forming a cage of crossing MTs<br />

around the nuclear pair (Runeberg et al. 1986; Salo et al. 1989).Actin is visualized<br />

as a cap in the hyphal tips (Fig. 1C), as small plaques along the apical cell<br />

and as a ring (Fig. 1D) at the site where the cross wall will be formed (Salo et<br />

al. 1989; Raudaskoski et al. 1991; Gorfer et al. 2001).<br />

Comparison of the structure/organization of actin cytoskeleton in the taxonomically<br />

closely related slow-growing ectomycorrhiza-forming S. bovinus<br />

and fast-growing wood-decaying S. commune reveals several differences. In<br />

the slow-growing hyphae of S. bovinus the actin cap is more extensive than in<br />

S. commune, and the cap can be easily visualized with fluorochrome-labeled<br />

phalloidin that binds only to filamentous actin (Barak et al. 1980). In contrast,<br />

the visualization of the actin cap at the hyphal tips of S. commune succeeds<br />

only with an actin antibody, which visualizes actin monomers in addition to<br />

actin filaments. These differences suggest that the structure of MFs is more<br />

stable and their number is higher at the hyphal tip in the slow-growing ectomycorrhizal<br />

than in the fast-growing wood-decay fungus. In the hyphae of S.<br />

bovinus, it is also possible to distinguish occasionally actin filaments (Gorfer<br />

et al. 2001) comparable to those seen at a specific growth phase in budding<br />

yeast cells of S. cerevisiae (Kilmartin and Adams 1984).Actin filaments are not<br />

observed in the hyphae of fast-growing filamentous fungi, such as S. commune<br />

(Runeberg et al. 1986; Raudaskoski et al. 1991) or Neurospora crassa<br />

(Heath et al. 2000). These observations suggest that there probably are some<br />

basic differences in the structure of the actin cytoskeleton between slowgrowing<br />

and fast-growing filamentous fungi. The question whether these differences<br />

are associated with the ability of S. bovinus to form ectomycorrhiza<br />

with P. sylvestris root cells and whether these specific features occur in all<br />

ectomycorrhiza-forming fungi has to be answered the in the future.<br />

The use of drugs against polymerized tubulin and actin has given insights<br />

into their roles in hyphal growth. The depolymerization of the MT cytoskeleton<br />

with an anti-MT drug leads to strong branching of the hyphae in ectomycorrhizal<br />

fungi such as Amanita muscaria, Hebeloma cylindrosporum, Paxillus<br />

involutus, and S. bovinus (Niini and Raudaskoski 1993). In contrast, the<br />

depolymerization of actin filaments from the hyphae of S. bovinus with<br />

cytochalasin D leads to swelling of the hyphal tip cells and loss of the polarized<br />

growth pattern (Niini 1998; Raudaskoski et al. 2001). Nonpolarized<br />

growth and strong branching of hyphae are also observed when an ectomycorrhizal<br />

fungus is associated with the <strong>plant</strong> root cells (Kottke and Oberwin-


18 Mycorrhizal Development and Cytoskeleton 305<br />

kler 1987; Timonen et al. 1993; Niini 1998; Raudaskoski et al. 2001). The similar<br />

morphology of nonsymbiotic hyphae treated with inhibitors and of the<br />

hyphae grown in association with the root cells have led us to the hypothesis<br />

that the change in the hyphal morphology is due to a signal from root cells,<br />

the recognition and transduction of which leads to a reorganization of the<br />

actin cytoskeleton.<br />

5 Regulation of Actin Cytoskeleton Organization in Fungal<br />

Hyphae and Plant Cells<br />

Recently, several GTPases known to play an important role in linking extracellular<br />

signals to reorganization of actin cytoskeleton in yeasts and mammalian<br />

cells (Johnson 1999) have been isolated from S. bovinus and S. commune.<br />

The GTPases are conserved molecular switches that are normally<br />

anchored to the plasma membrane by a C-terminally attached farnesyl tail. In<br />

the active form the protein is bound to GTP, in the inactive one to GDP (Nuoffer<br />

and Balch 1994).<br />

Until now, a Ga,aCdc42 and a rac cDNA and two ras cDNAs have been isolated<br />

and characterized from S. bovinus and three Ga cDNAs, a Cdc42, a ras<br />

and a rho3 cDNA from S. commune (Gorfer et al. 2001; Raudaskoski et al.<br />

2001). Out of these genes only ras had been isolated before from an ectomycorrhizal<br />

fungus Laccaria laccata (Sundaram et al. 2001). The GTPases cloned<br />

from S. bovinus are expressed in vegetative hyphae, but also during ectomycorrhiza<br />

formation (Gorfer et al. 2001; Raudaskoski et al. 2001). SbCdc42 cDNA<br />

is able to complement the temperature-sensitive S. cerevisiae cdc42 mutation<br />

causing disruption of actin cytoskeleton (Johnson and Pringle 1990), which<br />

suggests that SbCdc42 is also involved in regulating the organization of actin<br />

cytoskeleton as it is in yeast and animal cells (Gorfer and Raudaskoski,<br />

unpubl. data).<br />

The small GTPases were chosen as the target for research in the ectomycorrhizal<br />

fungus S. bovinus on the basis of the following previous results: (1)<br />

IIF microscope analyses show rearrangement of cytoskeletal elements in S.<br />

bovinus hyphae at the formation of ectomycorrhiza (Timonen et al. 1993;<br />

Niini 1998; Raudaskoski et al. 2001). (2) The reorganization of the cytoskeleton<br />

in fungal hyphae occurs without differential expression of fungal tubulin<br />

or actin genes (Niini et al. 1996; Tarkka et al. 2000). (3) In S. commune, closely<br />

related to S. bovinus, the mating interaction necessary for sexual reproduction<br />

is regulated by the signal transduction pathway starting from the pheromone-<br />

G-protein-coupled receptor (GPCR) interaction (Wendland et al. 1995; Vaillancourt<br />

et al. 1997; Raudaskoski 1998; Raudaskoski et al. 1998; Fowler et al.<br />

1999). In animal cells (Schmidt and Hall 1998), and in the yeast S. cerevisiae<br />

(Johnson 1999), the effect of small GTPases (e.g., Cdc42, Rac and Rho) on the<br />

organization of the actin cytoskeleton is initiated by the binding of a signal


306<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

molecule to a G-protein coupled receptor, which is the receptor-type shown to<br />

mediate the signalling between haploid hyphae at the mating interaction in S.<br />

commune.<br />

A high number of different types of ligands/signals are recognized by the<br />

members of the GPCR superfamily in animal cells (Neer 1995). Recently, it has<br />

been shown that a set of GPCRs may function in fungi as well, since a distinct<br />

GPCR system from the one recognizing the pheromones senses glucose in<br />

yeasts (Versele et al. 2001). It could thus be speculated that in ectomycorrhizaforming<br />

fungi a GPCR originally involved in sexual reproduction is able to<br />

interact with signal molecules produced by <strong>plant</strong> roots, due to a mutational<br />

change in its structure. The contact with the <strong>plant</strong> would be signalled into the<br />

hyphal cells where it might lead to a change in the organization of actin<br />

cytoskeleton from highly polarized to a more relaxed form via a pathway<br />

involving the small GTPases Cdc42 and Rac1 (Gorfer et al. 2001). This could<br />

then lead to the development of a hyphal morphology suitable for the symbiotic<br />

growth. Perhaps it is meaningful that the majority of ectomycorrhizaforming<br />

fungi belong to homobasidiomycetes. This group of filamentous<br />

fungi includes species, such as S. commune and Coprinus cinereus, in which<br />

sexual reproduction is regulated by the signal transduction pathway starting<br />

from pheromone-GPCR interaction (Fowler et al. 1999; Olesnicky et al. 1999).<br />

In endomycorrhizal fungi, the characteristic changes in hyphal morphology,<br />

the formation of the appressorium on the root <strong>surface</strong> at the beginning of<br />

colonization and the strong branching of hyphae at arbuscule formation, may<br />

require the reorganization of actin cytoskeleton. It may be speculated that the<br />

different hyphal morphologies are due to the perception of <strong>plant</strong> signals,<br />

which could be mediated to the actin cytoskeleton through the small GTPases<br />

belonging to the Rho subfamily.<br />

Small GTPases of the Rho subfamily exist also in <strong>plant</strong>s, where they are<br />

addressed as Rac or Rop proteins. In Arabidopsis 11 Rac/Rop genes have been<br />

identified (Kost et al. 1999a; Li et al. 2001). The expression analysis of the constitutively<br />

active mutant form of Rac/Rop proteins unable to hydrolyse GTP or<br />

the dominant negative mutant form unable to exchange GDP to GTP has<br />

shown that the GTPases are involved in the regulation of apical growth and<br />

organization of MFs in pollen tubes (Kost et al. 1999a; Zheng and Yang 2000;<br />

Fu et al. 2001), and in root hairs (Molendijk et al. 2001). Rac/Rop proteins<br />

appear also to be involved in the regulation of organization of the actin<br />

cytoskeleton during stomatal closure in response to abscisic acid (Lemichez et<br />

al. 2001). The expression of constitutively active and dominant negative forms<br />

of Rac/Rop under the 35S universal promoter indicated that these proteins<br />

participate in multiple distinct signalling pathways that control <strong>plant</strong> growth,<br />

development and responses to the environment (Li et al. 2001).


6 Actin Binding Proteins<br />

18 Mycorrhizal Development and Cytoskeleton 307<br />

In eukaryotic cells, a high number of actin binding proteins (ABPs) regulate<br />

polymerization and depolymerization, bundling and cross-linking of MFs,<br />

and movement of cargo along the MFs. In animal cells, many of the ABP<br />

encoding genes have been isolated and the function of the corresponding<br />

proteins has been characterized. In <strong>plant</strong>s and filamentous fungi, the<br />

ABP research is just beginning. In Arabidopsis, the members of the gene<br />

families encoding ADF proteins (actin depolymerizing factor/cofilin; Dong<br />

et al. 2001), profilins (Ramachandran et al. 2000), Arp2 (Klahre and Chua<br />

1999), villins (Klahre et al. 2000), and myosins (Reddy and Day 2001) have<br />

been cloned and their expression patterns in different <strong>plant</strong> organs characterized.<br />

ADF protein family members interact with actin monomers and filaments<br />

in a pH-sensitive manner. When ADF/cofilin binds to filamentous (F) actin it<br />

accelerates the dissociation of subunits from the pointed ends of actin filaments<br />

(Bamburg 1999; Cooper and Schafer 2000). The properties of maize<br />

cofilins have been analysed by using recombinant ZmADF1 and ZmADF3<br />

proteins (Hussey et al. 1998). ZmADF3 has the ability to bind monomeric (G)<br />

actin and F-actin and to decrease the viscosity of polymerized actin solutions,<br />

indicating an ability to depolymerize actin filaments (Lopez et al. 1996).<br />

ZmADF3 is phosphorylated on Ser6 by a calcium-stimulated protein kinase in<br />

<strong>plant</strong> extracts (Smertenko et al. 1998), which suggests that phosphorylation<br />

regulates cofilin’s actin binding activity and affects the stability of the actin<br />

cytoskeleton in a manner shown in animal cells (Daniels and Bokoch 1999).<br />

Profilin is a G-actin binding protein known to interact in animal and yeast<br />

cells with proline-rich motifs of other proteins and with polyphosphoinositides.<br />

The interaction of profilin with G-actin provides a mechanism to<br />

sequester actin monomers and promote actin depolymerization. It appears<br />

that profilin may also be involved in promoting actin polymerization. This<br />

might take place by binding to proline-rich motifs in proteins that convey<br />

intra- or extracellular signals to reorganization of actin cytoskeleton (Mullins<br />

2000). In Arabidopsis,thePFN-1 gene, from profilin gene family with eight to<br />

ten members, is expressed in root and root hairs, and in a ring of cells in the<br />

elongating zone of the root (Ramachandran et al. 2000), in which the profilin<br />

levels could be involved in the regulation of cell elongation as a rate-limiting<br />

factor.<br />

Plants have also several genes with high homology to animal villin (Klahre<br />

et al. 2000). The first <strong>plant</strong> villin was isolated from pollen tubes as a 135-kDa<br />

actin-bundling protein (Yokota et al. 1998; Yokota and Shimmen 1999). Its<br />

identity as a villin-gelsolin family member (Vidali et al. 1999) was confirmed<br />

by partial amino acid sequencing and by isolating the corresponding cDNA<br />

from a pollen grain cDNA expression library. Immunodetection of villin<br />

revealed its co-localization with actin bundles in pollen tubes. Due to the gel-


308<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

solin-like headpiece, villin may also act as an actin-severing protein, although<br />

this activity has not yet been demonstrated for <strong>plant</strong> villins.<br />

In Arabidopsis, 17 myosin encoding genes have been identified, which on<br />

the basis of phylogenetic analysis, fall into <strong>plant</strong>-specific myosin classes VIII<br />

(4 genes) and XI (13 genes). The three cloned myosins from maize and four<br />

from Helianthus annuus also fall into these classes (Reddy and Day 2001). The<br />

true structure, enzymatic properties, intracellular localization and physiology<br />

of <strong>plant</strong> myosin are not yet known.<br />

The structure and function of actin binding proteins in association with<br />

the reorganization of the actin cytoskeleton in <strong>plant</strong> cells at mycorrhiza formation<br />

has not yet been studied, but seems to require attention. In endomycorrhiza,<br />

rearrangement of actin cytoskeleton was observed at the colonization<br />

of tobacco cortical cells by endomycorrhizal fungus (Genre and Bonfante<br />

1997). Actin cables, typical for noncolonized cortical root cells, disappeared<br />

and MF polymerized to a network at the plasma membrane surrounding the<br />

arbuscule branches. The observed changes may be hypothesized to require<br />

both the activation and function of proteins involved in the reorganization of<br />

the actin cytoskeleton as a consequence of the perception of signals from the<br />

intruding fungus.<br />

The research on actin binding proteins in filamentous fungi seems to be<br />

restricted to myosin (McGoldrick et al. 1995), in Aspergillus nidulans and to<br />

actin-related proteins Arp1 (Plamann et al. 1994) in Neurospora crassa,<br />

although in yeast, Saccharomyces cerevisiae, most actin binding proteins previously<br />

described in <strong>plant</strong>s are present and have been studied in detail<br />

(Ayscough 1998). In A. nidulans, the myosin I encoding gene myoA has been<br />

cloned and different mutational studies have indicated that MYOA is necessary<br />

for the maintenance of polarized growth, secretion and endocytosis<br />

(McGoldrick et al. 1995; Osherov et al. 1998; Yamashita and May 1998). The<br />

yeast S. cerevisiae and Schizosaccharomyces pombe, similar to animal cells,<br />

contain myosins from classes II and V in addition to those belonging to class<br />

I which predicts that additional myosins will be detected in future from filamentous<br />

fungi.<br />

7 Microtubule-Associated Proteins<br />

7.1 Plant Cells<br />

Microtubule-associated proteins (MAPs) are divided into structural and<br />

motor proteins. Several genes encoding structural <strong>plant</strong> MAPs have been isolated<br />

in the last few years. One of the MAP encoding genes was isolated from<br />

Arabidopsis, where its heat-sensitive mutation resulted in the disintegration of<br />

cortical microtubules in leaf, hypocotyl and root cells. The gene was named


18 Mycorrhizal Development and Cytoskeleton 309<br />

MOR1 (Whittington et al. 2001), its product is homologous to animal MAPs<br />

belonging to class TOGp-XMAP215, and in <strong>plant</strong> cells, the MOR1 protein is<br />

essential for cortical microtubule organization (Wasteneys 2002). Proteins<br />

with a MW of 60–68 kDa which associate in vitro with MTs, cause their polymerization<br />

and bundling and induce cross-bridges between them, have been<br />

biochemically purified from tobacco BY-2 cells (Jiang and Sonobe 1993) and<br />

carrot tissue culture cells (Chan et al. 1996, 1999). These proteins are called<br />

MAP-65 proteins and screening of a tobacco BY-2 cell cDNA library with an<br />

antiserum raised against them led to the isolation of a cDNA clone named<br />

NtMAP65–1a. The clone encodes a 580 amino acid polypeptide with no<br />

homology with any known animal MAPs. Further screening of the same<br />

library led to the isolation of two other similar cDNA clones, NtMAP65–1b<br />

and NtMAP65–1c (Smertenko et al. 2000). NtMAP65–1a protein induces polymerization,<br />

but not bundling of the MTs in vitro, and it is localized to areas of<br />

overlapping microtubules in spindle and phragmoplast and on a certain subpopulation<br />

of stable cytoplasmic MTs (Smertenko et al. 2000; Lloyd and<br />

Hussey 2001). The isolation of genes encoding NtMAP65 proteins and characterization<br />

of the properties of the proteins in vitro and in vivo have clearly<br />

shown that there are <strong>plant</strong>-specific MAPs whose ability to interact and bind to<br />

microtubules is different from that of animal MAPs.<br />

The group of motor MAPs consists of kinesins and dyneins. Kinesins are<br />

mainly plus end-directed MT-dependent motor molecules, but minus enddirected<br />

kinesins are also known (Miki et al. 2001). The heavy chains of<br />

kinesins are formed of a motor domain with ATP and MT binding regions and<br />

ATP hydrolyzing activity; stalk domain with coiled-coil structure and a cargobinding<br />

domain. Recently, it has become obvious that kinesin and myosin<br />

share a common core structure in the ATP-binding region that converts<br />

energy from ATP into protein motion, using a similar conformational strategy<br />

(Vale and Milligan 2000). Kinesins are monomeric, homodimeric or homotetrameric<br />

(Kim and Endow 2000). Several small polypeptides can be associated<br />

with kinesins and they regulate the activity and function of these motor<br />

proteins. The position of the ATP binding domain, whether on the N- or C-terminus<br />

of the polypeptide, makes the protein either a plus or a minus enddirected<br />

motor.<br />

The kinesin superfamily is divided into various subfamilies including conventional<br />

kinesins and several kinesin-related proteins (KRPs; Kim and<br />

Endow 2000). Conventional kinesins are mainly involved in the (inter) intracellular<br />

transport of membranous organelles, whereas most KRPs function in<br />

nuclear division. Genes encoding KRPs have been isolated from tobacco (Mitsui<br />

et al. 1996; Asada et al. 1997), potato (Reddy et al. 1996b), and Arabidopsis<br />

(Mitsui et al. 1993; Liu et al. 1996; Reddy et al. 1996a). Of these, the best studied<br />

is TKRP125 (Asada et al. 1997), which is a plus end-directed motor molecule<br />

with an ATP hydrolyzing domain at the N-terminus. Structurally, it is a<br />

BimC-type kinesin that functions as a homotetramer (Kim and Endow 2000).


310<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

TKRP125 is located in anaphase spindle and phragmoplast.A similar location<br />

has been recently shown for a TKRP125 homologue and a KRP slightly deviating<br />

from TKRP125 isolated from carrot tissue culture (Barroso et al. 2000).<br />

In <strong>plant</strong>s, a subfamily of kinesin-like genes encodes proteins with a calmodulin-binding<br />

domain. Genes encoding members of this subfamily have been<br />

isolated from Arabidopsis (AKCBP; Reddy et al. 1996a), potato (PKCBP; Reddy<br />

et al. 1996b), and tobacco (TCK1; Wang et al. 1996). The genes encode minus<br />

end-directed motor proteins with the calmodulin-binding region in the<br />

motor domain at the very C-terminus of the polypeptide, suggesting that calcium/calmodulin<br />

may be involved in the regulation of the microtubule-based<br />

movements in <strong>plant</strong>s. Genes encoding other types of minus end-directed<br />

motor molecules are also present in Arabidopsis and they have been named<br />

katA to katE (Mitsui et al. 1993, 1994). The production of minus end-directed<br />

KatB and KatC kinesins increases during M-phase of the cell cycle in tobacco<br />

BY-2 cell cultures, suggesting that mitotic spindle and phragmoplast may also<br />

be their site of action (Mitsui et al. 1996). Dyneins are large minus enddirected<br />

motors. Until now, no dynein heavy chain encoding gene has been<br />

detected in expressed sequence tag (EST) databases from flowering <strong>plant</strong>s or<br />

in the recently sequenced Arabidopsis genome (Lawrence et al. 2002).<br />

In endomycorrhiza, the organization of MT cytoskeleton changes when the<br />

fungus colonizes the <strong>plant</strong> cell (Genre and Bonfante 1997). In addition, in P.<br />

sylvestris ectomycorrhiza cortical cells surrounded by the Hartig net seem to<br />

contain less cortical MTs than the noninfected cortical cells (Niini 1998). The<br />

reorganization of the MT cytoskeleton takes place not only in the colonized<br />

endomycorrhizal <strong>plant</strong> cells, but also in the cells adjacent to the colonized<br />

ones, and when the arbuscules have completely collapsed, the transversely<br />

oriented cortical MTs next to the plasma membrane reappear (Blancaflor et<br />

al. 2001). The reorganization of the MT cytoskeleton observed in connection<br />

with symbiosis requires destabilization (depolymerization) of existing cortical<br />

MTs, polymerization of new MT areas at the plasma membrane next to the<br />

colonizing fungus, and repolymerization of cortical MTs after the senescence<br />

of the arbuscular structure. The observed changes present a very dynamic<br />

behavior of MT cytoskeleton in endomycorrhiza, which probably involves the<br />

function of both structural and motor MAPs in <strong>plant</strong> cells.<br />

7.2 Fungal Hyphae<br />

In filamentous fungi, the genes encoding MT-associated motor proteins<br />

kinesins and dyneins have caught more attention than structural MAPs. The<br />

first kinesin-related protein was cloned from ascomycete Aspergillus nidulans<br />

(Enos and Morris 1990) as a temperature-sensitive mutation that blocked<br />

mitosis in germinating conidia. Later studies have shown that this bimC<br />

(blocked in mitosis) gene encodes a protein with amino-terminal motor and


18 Mycorrhizal Development and Cytoskeleton 311<br />

coiled-coil tail domains. The protein forms homotetramers and it is required<br />

for nuclear division. A motor protein has been biochemically characterized<br />

from Neurospora crassa (Steinberg and Schliwa 1996) and the zygomycete<br />

Syncephalastrum racemosum (Steinberg 1997), and the production of an antibody<br />

against the N. crassa protein helped to isolate the first fungal conventional<br />

kinesin encoding gene, Nkin (Steinberg and Schliwa 1995). Using<br />

probes derived from conserved regions of kinesins and screening existing<br />

genomic databases, has led to the isolation of conventional kinesins from<br />

Ustilago maydis (Lehmler et al. 1997), S. racemosum (Grummt et al. 1998),<br />

Nectria haematococca (Wu et al. 1998), and A. nidulans (Requena et al. 2001).<br />

Conventional kinesins belong to the fastest kinesins known in eukaryotic<br />

cells, moving as dimers on MTs with a velocity of 2.5 µm/s in in vitro gliding<br />

experiments, which is three- to fivefold faster than that of their animal counterparts<br />

(Steinberg 1997; Grummt et al. 1998). The structural features responsible<br />

for the high gliding velocity of fungal kinesins are not yet well understood,<br />

but under active investigation (Kallipolitou et al. 2001).<br />

The deletion of the conventional kinesin-encoding gene from any of the filamentous<br />

fungi investigated leads to a reduction of polarized growth and the<br />

size of Spitzenkörper, the secretory vesicle aggregation at the hyphal tip<br />

(Lehmler et al. 1997; Seiler et al. 1997; Wu et al. 1998; Seiler et al. 1999; Requena<br />

et al. 2001). These observations support the idea that conventional kinesin is<br />

involved in the transportation of components necessary for hyphal growth<br />

along the MTs towards the hyphal tip. It is noteworthy that in no case the<br />

mutation of the conventional kinesin gene has led to a complete cessation of<br />

growth, which implies the existence of other hyphal transportation systems,<br />

either based on other less efficient kinesins, or on the actin – myosin system.<br />

In A. nidulans, the deletion of kinesin also caused disturbance in nuclear distribution<br />

in the germ tube and hyphae suggesting that conventional kinesin<br />

plays a role in nuclear migration (Requena et al. 2001). In addition, the functional<br />

analysis suggested that the conventional kinesin of A. nidulans is<br />

involved in destabilization of MTs (Requena et al. 2001).<br />

The MT-associated motor cytoplasmic dynein is generally proposed to<br />

provide the motive force for nuclear movement in filamentous fungi (Morris<br />

et al. 1995; Fischer 1999; Suelmann and Fischer 2000). The heavy chain of<br />

cytoplasmic dynein is a large polypeptide with a region for dimerization at<br />

the N-terminal and globular motor domain with four ATP binding and MTbinding<br />

sites at the C-terminal portion of the polypeptide. In association with<br />

the N-terminus of the dynein heavy chain, smaller polypeptides called intermediate<br />

and light chains occur, which are involved in the regulation of dynein<br />

heavy chain activity and function (Steinberg 1998, 2000). A multi-subunit<br />

complex dynactin is also required for efficient MT-associated transport by<br />

cytoplasmic dynein. In the dynactin complex actin-related protein ARP1 is<br />

the most abundant and p150 Glued the largest subunit. The dynactin complex<br />

also includes several other polypeptides that play an important role both in


312<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

the activation of the dynein motor molecule and in the binding of dynein to<br />

membranes (King and Schroer 2000).<br />

Dynein heavy chain encoding genes have been characterized from A. nidulans<br />

with the help of a temperature-sensitive mutation nudA that affects<br />

nuclear distribution, and from N. crassa as a morphological mutation with<br />

curly hyphae named ro-1. In the temperature-sensitive mutation of the nudA<br />

gene the nuclear movement from conidia into the germ tube failed and small<br />

hyphal colonies were formed at restrictive temperature (Xiang et al. 1994). In<br />

the ro-1 mutant of N. crassa, nuclear aggregates formed in the hyphae at some<br />

distance from the hyphal tip (Plamann et al. 1994). The cloning and sequencing<br />

of the nudA and ro-1 genes revealed that they both encode the cytoplasmic<br />

dynein heavy chains with ATP-binding motor domain at the C-terminus.<br />

Later, the dynein heavy chains have been identified from N. haematococca<br />

(Inoue et al. 1998), and U. maydis (Straube et al. 2001), by using degenerate<br />

oligonucleotide primers. Interestingly, this approach has led to the identification<br />

of a dynein with split motor domain in U. maydis, in which the N-terminus<br />

of the motor domain including the four ATP binding regions is encoded<br />

by dyn1 and the MT-binding part by dyn2 gene. Some of the ro-1-like phenotypic<br />

N. crassa mutants carry mutations in genes encoding subunits of the<br />

dynactin complex (Bruno et al. 1996). This has facilitated the isolation of ro-4<br />

and ro-3 genes encoding the actin-related protein,Arp1 (Plamann et al. 1994),<br />

and p150 Glued (Tinsley et al. 1996). Several other ro genes, such as ro-2, ro-7 and<br />

ro-12, encoding different subunits of the dynactin complex have been identified<br />

(Lee et al. 2001).<br />

By comparing the effects of the deletion of kinesin (Nkin), or dynein (ro-1),<br />

and both proteins on nuclear distribution, vesicle transport, secretion and<br />

vacuole formation in N. crassa hyphae, it was concluded that conventional<br />

kinesin indeed is responsible for the apical transport of vesicles destined for<br />

secretion, whereas dynein is responsible for nuclear movements and transport<br />

of vacuole precursors in the opposite direction. The latter phenomenon<br />

is suggested to support the formation of vacuoles in the basal part of N. crassa<br />

hyphae (Seiler et al. 1999), while conventional kinesin encoded by kin2 was<br />

shown to be responsible for the accumulation of vacuoles to the basal part of<br />

in U. maydis dikaryotic hyphae (Steinberg et al. 1998). These results indicate<br />

that mutations in different motor molecules may result in the same phenotype<br />

in fungi belonging to different taxa, the phenotype in this case being the<br />

vacuolation of the basal part of a hypha. Interestingly, in a dynein-deficient<br />

mutant of N. haematococca, astral-like arrays of cytoplasmic MTs radiating<br />

from nuclear spindle pole bodies were missing, which probably causes the<br />

clustered nuclear distribution in the mutant hyphae. In filamentous fungi, the<br />

astral MTs are suggested to be responsible for post-mitotic nuclear migration<br />

and anchoring of the interphase nuclei to membrane structures (Aist and<br />

Bayles 1988; Salo et al. 1989; Raudaskoski et al. 1991; Morris et al. 1995; Inoue<br />

et al. 1998; Raudaskoski 1998).


18 Mycorrhizal Development and Cytoskeleton 313<br />

Recent observations on living nonsymbiotic hyphae of endomycorrhizal<br />

fungi (Bago et al. 1998) have revealed active nuclear movements. This phenomenon<br />

has been observed in anastomosing hyphae before the establishment<br />

of symbiosis (Giovannetti et al. 1999), and between hyphae growing out<br />

from infected roots (Giovannetti et al. 2001). The nuclear movements was<br />

shown to be accompanied by cytoplasmic flow. These observations have<br />

awoken interest in the mechanism responsible for the nuclear movements,<br />

e.g., whether it is MT- or actin cytoskeleton-dependent. In ectomycorrhizal<br />

hyphae mobility of vacuoles has been observed in and between adjacent cells<br />

of young dikaryotic hyphae of Pisolithus tinctorius (Shepherd et al. 1993), as<br />

well as in hyphae growing out from the Eucalyptus pilularis–P. tinctorius ectomycorrhiza<br />

(Allaway and Ashford 2001). The vacuole motility includes tubule<br />

extensions and retractions, undulating movements, projections of tubules<br />

from spherical vacuoles and fusions of tubules with spherical vacuoles and<br />

other tubules (Cole et al. 1998). The motile vacuolar system has a high phosphorus,<br />

nitrogen and potassium content. A comparable content of ions also<br />

occurs in the hyphal vacuoles of the mycorrhizal sheath and the Hartig net<br />

(Ashford et al. 1999). This suggests that the tubular vacuolar system is perhaps<br />

involved in the transfer of phosphorus and nitrogen, both in short distance<br />

transport from cell to cell and in long distance transport from hyphal tips to<br />

the <strong>plant</strong>/fungal interface (Cole et al. 1998). Interestingly, the motility of tubular<br />

vacuolar system is dependent on an intact MT cytoskeleton (Hyde et al.<br />

1999).<br />

8 Cell Cycle and Cytoskeleton in Mycorrhiza<br />

In P. sylvestris ectomycorrhiza, an increase in the number of short roots was<br />

observed in the root region that was in contact with the fungus (Niini and<br />

Raudaskoski 1998). Similarly, in Eucalyptus grandis- Pisolithus ectomycorrhiza,<br />

the number of root tips increased in seedlings inoculated with the symbiotic<br />

fungus (Burgess et al. 1995). In the colonized short roots of P. sylvestris<br />

the fungus also activates the cell divisions in the root tip leading to the formation<br />

of dichotomous and coralloid mycorrhiza (Niini 1998), unique for<br />

pines. The increase in short root number and the formation of dichotomous<br />

and coralloid short roots suggests that the presence of fungal mycelium activates<br />

the cell cycle in the pericycle for short root production and in the root<br />

tip meristem at the formation of dichotomous and coralloid short roots. In<br />

contrast, in endomycorrhiza the <strong>plant</strong> cell cycle appears not to be activated,<br />

but the chromatin of the <strong>plant</strong> nucleus in the root cortical cells decondensates<br />

which leads to nuclear hypertrophy (Berta et al. 1990).<br />

Activation of the cell cycle in the root cortex occurs when root nodules are<br />

formed in the legume <strong>plant</strong>s associated with Rhizobium bacteria (Mylona et<br />

al. 1995), or in the roots of woody <strong>plant</strong>s at actinorhizal nodule formation


314<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

(Berg 1999). The integration of T-DNA from an unmodified Ti-plasmid of<br />

Agrobacterium tumefaciens into the <strong>plant</strong> genome induces tumor formation<br />

in <strong>plant</strong>s by activating the <strong>plant</strong> cell cycle. Agrobacterium T-DNA is known to<br />

encode enzymes necessary for cytokinin and auxin synthesis, which in turn<br />

promote tumor formation via reactivation of the cell cycle in Agrobacteriuminfected<br />

<strong>plant</strong>s (Sheng and Citovsky 1996). In Rhizobium it has been shown<br />

that the Nod-factors, the signal molecules produced by Rhizobium,are necessary<br />

for reactivation of the cell cycle. Nod factors are shown to elicit local<br />

reduction in <strong>plant</strong> auxin transport and auxin accumulation, which probably<br />

stimulates root cortical cell division (Mathesius et al. 1998; Boot et al. 1999).<br />

Formation of a nodule-like structure can also be induced by treatment with<br />

auxin transport inhibitors (Hirsch and Fang 1994).<br />

Early experiments by Slankis (1949) using isolated P. sylvestris roots and<br />

exogenous auxin (IAA) treatment showed that it was possible to obtain mycorrhiza-like<br />

dichotomous branching without the fungus. Some ectomycorrhizal<br />

fungi including S. bovinus have been shown to be able to produce IAA<br />

or cytokinin (Beyrle 1995). The effect of auxin on root branching and on the<br />

formation of dichotomous and coralloid short roots has been recently reinvestigated<br />

in several pine species by using auxin and auxin transport<br />

inhibitors (Kaska et al. 1999). This research indicated that auxin induces a<br />

marked increase in the formation of lateral roots while the treatment with<br />

auxin transport inhibitors induced dichotomous and coralloid short roots.<br />

The relationship between the hormone treatments or the effect of the fungal<br />

hyphae on the expression of cell cycle regulating genes in Pinus has not<br />

been investigated. In <strong>plant</strong>s as in other eukaryotic organisms, the cyclins and<br />

cyclin-dependent kinases (CDKs) are key regulators of cell cycle (Mironov et<br />

al. 1999). Both A- and B-type cyclins are expressed during mitosis, the A-type<br />

cyclins being also active during S-phase progression. D-type cyclins have an<br />

important role in the G1 to S phase transition. Transcription of D-type cyclins<br />

can be induced by the phytohormone cytokinin or by sucrose, which means<br />

that mitogenic signals stimulate transcription of D-cyclins and modulate cell<br />

cycle activity. Recently, a new D-type cyclin has been identified in Arabidopsis<br />

that is expressed during lateral root formation and the expression of which is<br />

stimulated by sucrose (De Veylder et al. 1999).<br />

In higher-<strong>plant</strong> cells, the MT organization is regulated during the cell cycle<br />

(Vantard et al. 2000). Especially G2-phase and mitosis are accompanied by<br />

changes in the distribution of MTs. In early G2 the cortical interphase MTs<br />

accumulate to form the preprophase band (PPB) which precisely marks the<br />

future site for the cell plate formation. The breakdown of the nuclear envelope<br />

leads to the PPB disassembly and polymerization of spindle MTs. After<br />

karyokinesis the phragmoplast is formed at the site marked at G2 phase by the<br />

PPB. The phragmoplast consists of a ring of anti-parallel, inter-digitating<br />

microtubules and actin filaments, which are thought to transport the vesicles<br />

containing the material for cell plate construction. A close relationship


etween the function of cell cycle and MT cytoskeleton is suggested by the<br />

association of cell cycle-regulating components with specific MT arrays in<br />

dividing <strong>plant</strong> cells. B-cyclins and cyclin-dependent kinase Cdc2a are<br />

immunolocalized to preprophase band, mitotic spindle and chromosomes,<br />

while A1 cyclin is detected at cytokinesis in association with phragmoplast<br />

MTs. At interphase, Cdc2a kinase and A cyclin are localized in the nucleus<br />

(John et al. 2001).<br />

In ectomycorrhiza, the association of <strong>plant</strong> root cells with the fungus leads<br />

probably to mitogenic stimuli that activate the cell cycle in pericycle and root<br />

meristematic tissue, seen in Pinus as production of short roots with dichotomous<br />

and coralloid tips. Cell cycle activation involves the regulation of MT<br />

dynamics so that the transition of one MT array to another is achieved during<br />

different cell cycle phases and also at cellular differentiation.<br />

The Nobel Prize in Physiology or Medicine 2001 was awarded to L.H.<br />

Hartwell, R.T. Hunt and Sir Paul M. Nurse for their discoveries of key regulators<br />

of the cell cycle. The yeasts Schizosaccharomyces pombe and Saccharomyces<br />

cerevisiae were important model organisms for the prize winning<br />

research. In spite of this, very little is known about cell cycle regulation in filamentous<br />

fungi. The central genes have only been isolated and their function<br />

studied in the filamentous fungus, A. nidulans (Ye et al. 1999). In future, the<br />

isolation and characterization of the genes involved in regulation of the cell<br />

cycle (Tarkka 2001) and the cytoskeleton from both symbiotic partners, tree<br />

roots and fungal hyphae, will provide us with new insight about the development<br />

of the ectomycorrhizal association.<br />

9 Cytoskeletal Research Methods<br />

18 Mycorrhizal Development and Cytoskeleton 315<br />

In the previous sections the use of in situ hybridization (Uribe et al. 1998), and<br />

GUS-reporter gene constructs (An et al. 1996a, b; Chu et al. 1998; Kandasamy<br />

et al. 2001), were described in localization transcripts of actin and tubulin<br />

gene products or for visualization of their activity in different <strong>plant</strong> tissues,<br />

even in endomycorrhiza (Bonfante et al. 1996). The application of these methods<br />

requires that the genes encoding tubulin or actin, and a transformation<br />

system are available. When actins or tubulins are detected at the protein level<br />

by Western blotting (Raudaskoski et al. 1987), commercially produced antibodies<br />

are applied. When commercial antibodies are used, the possibility<br />

always exists that some of the tubulin or actin proteins are not recognized.<br />

Therefore, efforts have been made to produce antibodies specific for the different<br />

actin or tubulin proteins (McLean et al. 1990). The availability of such<br />

antibodies would also be helpful for investigating the distribution of the different<br />

cytoskeletal proteins in <strong>plant</strong> and fungal cells by immunocytochemical<br />

methods.


316<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

9.1 Indirect Immunofluorescence Microscopy<br />

Most antibodies that lead to signal detection in immunoblots can also be used<br />

to visualize the intracellular structure of actin filaments or MTs by indirect<br />

immunofluorescence (IIF) microscopy (Fig. 1A, B, E) as discussed in previous<br />

sections. IIF is based on the use of two antibodies, primary and secondary.<br />

The primary antibody is raised against a cytoskeletal protein or its peptide<br />

while the fluorochrome-labeled secondary antibody is able to recognize the<br />

primary antibody. If the primary antibody is monoclonal, it binds to a single<br />

epitope of the target protein.A polyclonal primary antibody binds several epitopes<br />

of the target protein. The immunocytochemical methods can also be<br />

applied at the electron microscopic level, although then the secondary antibody<br />

has to be labeled with gold particles. MFs are often visualized by fluorochrome-labeled<br />

phallotoxins which bind specifically to filamentous actin<br />

(Barak et al. 1980).<br />

The successful visualization of MFs and MTs in root cells and fungal<br />

hyphae requires a fixative that penetrates quickly into the <strong>plant</strong> tissue and stabilizes<br />

the cytoskeletal elements in the polymerized form. A method that<br />

stops the metabolism of the cell even more quickly than chemical fixative and<br />

preserves the structure of MTs and MFs in fungal and <strong>plant</strong> cells is cryo-fixation<br />

(Raudaskoski et al. 1987, 1991, 1994; Åström et al. 1994; Lancelle et al.<br />

1997; Bourett et al. 1998). Pretreatment of <strong>plant</strong> cells with an MF-stabilizing<br />

agent MBS (3-maleimidobenzoiz acid N-hydroxysuccinimide) ester has also<br />

been used (Sonobe and Shibaoka 1989; Miller et al. 1999; de Ruijter et al.<br />

2001). Although the method leads to well-preserved MFs, the MBS pretreatment<br />

may also cause redistribution of the target molecules and the pretreatment<br />

may lead to less flexible images of actin cytoskeleton.<br />

The penetration of the antibody into the <strong>plant</strong> or fungal cell requires enzymatic<br />

digestion of the cell wall and permeabilization of the plasma membrane<br />

with a detergent after fixation. During the enzymatic digestion, the protease<br />

activity in the enzyme preparation has to be reduced by protease inhibitors<br />

and/or by adding 1 % bovine serum albumin in the digestion buffer. For enzymatic<br />

treatment and for labeling with the antibodies, cells or cell rows can be<br />

isolated or cut manually from the fixed material (Uetake et al. 1997; Uetake<br />

and Peterson 1997, 1998), which can also be embedded in 15 % agar (Genre<br />

and Bonfante 1997, 1998) or cyanoacrylate (Blancaflor et al. 2001) for sectioning.<br />

The visualization of cytoskeletal elements also succeeds well when the<br />

fixed <strong>plant</strong> roots are embedded in Steedsman’s wax and then sectioned<br />

(Baluška et al. 1992, 1995, 1997, 2001; Olinevich et al. 2001). Before labeling<br />

with the primary antibody the sections are dewaxed in ethanol, passed<br />

through a graded ethanol series diluted with PBS and either treated with a cell<br />

wall-degrading enzyme (Baluška et al. 1992), or not (Balu_ka et al. 2001).With<br />

these methods good preservation and visualization of cytoskeletal components<br />

is achieved in the roots of herbaceous <strong>plant</strong>s, even with endomycor-


hiza. Cells with well-preserved cytoskeletal structures can then be examined<br />

either by a regular fluorescence microscope or by a laser scanning confocal<br />

microscope.<br />

In ectomycorrhiza, the thick hydrophobic sheath around the root formed<br />

by fungal hyphae inhibits the penetration of the fixative or rapid freezing of<br />

the fungal and <strong>plant</strong> cells. The fixation of ectomycorrhizal root in situ under<br />

vacuum facilitates and speeds up the penetration of the fixative into the ectomycorrhizal<br />

root (Timonen et al. 1993; Niini and Raudaskoski 1998). In ectomycorrhiza<br />

it is necessary to prepare sections from the fixed roots and until<br />

now, the best results in immunolocalization of cytoskeletal elements have<br />

been achieved by using cryosections (Fig. 1A, B; Niini and Raudaskoski 1998).<br />

No success in the visualization of MTs or MFs has yet been achieved by<br />

embedding the Pinus short roots or ectomycorrhizal roots in wax.<br />

9.2 Microinjection Method<br />

Microinjection of fluorescent-labeled phalloidin or tubulin has been used to<br />

visualize MFs (Schmit and Lambert 1990; Zhang et al. 1993; Cleary 1995; Kim<br />

et al. 1995; Miller et al.1996) and MTs (Zhang et al. 1990; Yuan et al. 1994) in<br />

living <strong>plant</strong> cells. However, the visualization of cytoskeletal elements succeeds<br />

with microinjection only in a few <strong>plant</strong> cell types, such as stamen hair cells of<br />

Tradescantia (Zhang et al. 1993), or epidermal cells of leaves (Yuan et al. 1994),<br />

while cytoskeletal elements in narrow fungal hyphae are not easily studied<br />

with this method.<br />

9.3 Green Fluorescence Protein Technique<br />

18 Mycorrhizal Development and Cytoskeleton 317<br />

The MFs and MTs in <strong>plant</strong> and fungal cells have been successfully visualized<br />

with the help of green fluorescence protein (GFP) fused to actin, tubulin or to<br />

a cytoskeleton-associated protein. In <strong>plant</strong> cells, tubulin-GFP fusion protein<br />

has been used to visualize MTs in different Arabidopsis tissues (Ueda et al.<br />

1999; Whittington et al. 2001). Transient expression of the MT binding<br />

domain of mammalian MAP4 labeled with GFP and transgenic <strong>plant</strong>s with<br />

the same construct have been used for visualization of the MT organization in<br />

epidermal cells of fava bean (Marc et al.1998) and in Arabidopsis root cells<br />

(Bao et al. 2001). Fusion of GFP to the actin-binding domain of talin has led to<br />

visualization of actin cytoskeleton transiently in tobacco BY-2 suspension<br />

cells (Kost et al. 1998) and pollen tubes (Kost et al. 1998, 1999a, b; Fu et al.<br />

2001). Constitutive expression of the same construct visualized actin<br />

cytoskeleton in different tissues of Arabidopsis including root hairs (Kost et<br />

al. 1998; Baluška et al. 2000; Baluška and Volkmann 2002). Recently, both MTs<br />

and MFs were visualized in living onion epidermal cells by using the MT


318<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

binding domain of MAP4 and actin binding domain of talin fused with different<br />

spectral variants of GFP (Blancaflor 2002).<br />

MTs (Straight et al. 1997) and actin (Doyle and Botstein 1996) of yeast have<br />

also been successfully visualized by GFP-tubulin and -actin fusions as well as<br />

the MTs in the hyphae of Aspergillus nidulans (Han et al. 2001). Recently, it has<br />

been shown that GFP can be used as a reporter of a gene function in the<br />

hyphae of filamentous basidiomycetes (Lugones et al. 1999; Ma et al. 2001),<br />

although no fungal protein has yet been localized by fusion to GFP in these<br />

fungi. In future it seems possible, at least in endomycorrhiza forming <strong>plant</strong>s,<br />

to visualize the effect of fungal growth on the <strong>plant</strong> cytoskeleton in living root<br />

cells by GFP-fusion proteins. The application of GFP for visualization of<br />

cytoskeletal elements in endo- and ectomycorrhizal fungi during vegetative<br />

or symbiotic growth requires the development of an efficient transformation<br />

system for these fungi (Pardo et al. 2002).<br />

Acknowledgements. The authors thank Erja Laitiainen (M.Sc.) for technical help in<br />

preparing the manuscript. The work was supported by a grant from the Academy of Finland<br />

to M.R.<br />

References and Selected Reading<br />

Aist JR, Bayles CJ (1988) Video motion analysis of mitotic events in living cells of the<br />

fungus Fusarium solani. Cell Motil Cytoskel 9:325–336<br />

Allaway WG, Ashford AE (2001) Motile tubular vacuoles in extramatrical mycelium and<br />

sheath hyphae of ectomycorrhizal systems. Protoplasma 215:218–225<br />

An Y-Q, McDowell JM, Huang S, McKinney EC, Chambliss S, Meagher RB (1996a) Strong,<br />

constitutive expression of the Arabidopsis ACT2/ACT8 actin subclass in vegetative tissues.<br />

Plant J 10:107–121<br />

An Y-Q, Huang S, McDowell JM, McKinney EC, Meagher RB (1996b) Conserved expression<br />

of the Arabidopsis ACT1 and ACT3 actin subclass in organ primordia and mature<br />

pollen. Plant Cell 8:15–30<br />

Asada T, Kuriyama R, Shibaoka H (1997) TKRP125, a kinesin-related protein involved in<br />

the centrosome-independent organization of the cytokinetic apparatus in tobacco<br />

BY-2 cells. J Cell Sci 110:179–189<br />

Ashford AE, Vesk PA, Orlovich DA, Markovina A-L, Allaway WG (1999) Dispersed<br />

polyphosphate in fungal vacuoles in Eucalyptus pilularis/Pisolithus tinctorius ectomycorrhizas.<br />

Fungal Genet Biol 28:21–33<br />

Åström H, Giovannetti M, Raudaskoski M (1994) Cytoskeletal components in the arbuscular<br />

mycorrhizal fungus Glomus mosseae. Mol Plant-Microb Interact 7:309–312<br />

Ayscough KR (1998) In vivo functions of actin-binding proteins. Curr Opin Cell Biol<br />

10:102–111<br />

Bago B, Zipfel W, Williams RM, Chamberland H, Lafontaine JG, Webb WW, Piché Y<br />

(1998) In vivo studies on the nuclear behavior of the arbuscular mycorrhizal fungus<br />

Gigaspora rosea grown under axenic conditions. Protoplasma 203:1–15<br />

Baluška F,Volkmann D (2002) Pictures in cell biology.Actin-driven polar growth of <strong>plant</strong><br />

cells. Trends Cell Biol 12:14


18 Mycorrhizal Development and Cytoskeleton 319<br />

Baluška F, Parker JS, Barlow PW (1992) Specific patterns of cortical and endoplasmic<br />

microtubules associated with cell growth and tissue differentiation in roots of maize<br />

(Zea mays L.). J Cell Biol 103:191–200<br />

Baluška F, Barlow PW, Hauskrecht M, Kubica Š, Parker JS, Volkmann D (1995) Microtubule<br />

arrays in maize root cells. Interplay between the cytoskeleton, nuclear organization<br />

and post-mitotic cellular growth patterns. New Phytol 130:177–192<br />

Baluška F, Kreibaum A, Vitha S, Parker JS, Barlow PW, Sievers A (1997) Central root cap<br />

cells are depleted of endoplasmic microtubules and actin microfilament bundles:<br />

implications for their role as gravity-sensing statocytes. Protoplasma 196:212–223<br />

Baluška F, Salaj J, Mathur J, Braun M, Jasper F, Samaj J, Chua N-H, Barlow PW,Volkmann<br />

D (2000) Root hair formation: F-actin-dependent tip growth is initiated by local<br />

assembly of profilin-supported F-actin meshworks accumulated within expansinenriched<br />

bulges. Dev Biol 227:618–632<br />

Baluška F, Jasik J, Edelmann HG, Salajová T, Volkmann D (2001) Latrunculin B-induced<br />

<strong>plant</strong> dwarfism: <strong>plant</strong> cell elongation is F-actin-dependent. Dev Biol 231:113–124<br />

Bamburg JR (1999) Proteins of the ADF/cofilin family: essential regulators of actin<br />

dynamics. Annu Rev Cell Dev Biol 15:185–230<br />

Bao Y, Kost, B, Chua N-H (2001) Reduced expression of a-tubulin genes in Arabidopsis<br />

thaliana specifically affects root growth and morphology, root hair development and<br />

root gravitropism. Plant J 28:145–157<br />

Barak LS,Yocum RR, Nothnagel EA,Webb WW (1980) Fluorescence staining of the actin<br />

cytoskeleton in living cells with 7-nitrobenz-2-oxa-1,3-diazolephallacidin. Proc Natl<br />

Acad Sci USA 77:980–984<br />

Barlow PW, Baluška F (2000) Cytoskeletal perspectives on root growth and morphogenesis.<br />

Annu Rev Plant Physiol Plant Mol Biol 51:289–322<br />

Barroso C, Chan J, Allan V, Doonan J, Hussey P, Lloyd C (2000) Two kinesin-related proteins<br />

associated with the cold-stable cytoskeleton of carrot cells: characterization of a<br />

novel kinesin, DcKRP120–2. Plant J 24:859–868<br />

Berg HR (1999) Cytoplasmic bridge formation in the nodule apex of actinorhizal root<br />

nodules. Can J Bot 77:1351–1357<br />

Berta G, Sgorbati S, Soler V, Fusconi A, Trotta A, Citterio A, Bottone MG, Sparvoli E, Scannerini<br />

S (1990) Variations in chromatin structure in host nuclei of a vesicular arbuscular<br />

mycorrhiza. New Phytol 114:199–203<br />

Beyrle H (1995) The role of phytohormones in the function and biology of mycorrhizas.<br />

In: Varma A, Hock B (eds) Mycorrhiza. Structure, function, molecular biology and<br />

biotechnology. Springer, Berlin Heidelberg New York, pp 365–390<br />

Blancaflor EB (2002) The cytoskeleton and gravitropism in higher <strong>plant</strong>s. J Plant Growth<br />

Regul 21:120–135<br />

Blancaflor EB, Zhao L, Harrison MJ (2001) Microtubule organization in root cells of<br />

Medicago truncatula during development of an arbuscular mycorrhizal symbiosis<br />

with Glomus versiforme. Protoplasma 217:154–165<br />

Bonfante P, Perotto S (1995) Strategies of arbuscular mycorrhizal fungi when infecting<br />

host <strong>plant</strong>s. New Phytol 130:3–21<br />

Bonfante P, Bergero R, Uribe X, Romera C, Rigau J, Puigdomènech P (1996) Transcriptional<br />

activation of a maize a-tubulin gene in mycorrhizal maize and transgenic<br />

tobacco <strong>plant</strong>s. Plant J 9:737–743<br />

Boot KJM, Van Brussel AAN, Tak T, Spaink HP, Kijne JW (1999) Lipochitin oligosaccharides<br />

from Rhizobium leguminosarum bv.viciae reduce auxin transport capacity in<br />

Vicia sativa subsp. nigra roots. Mol Plant-Microb Interact 12:839–844<br />

Bourett TM, Czymmek KJ, Howard RJ (1998) An improved method for affinity probe<br />

localization in whole cells of filamentous fungi. Fungal Gen Biol 24:3–13


320<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

Bruno KS, Tinsley JH, Minke PF, Plamann M (1996) Genetic interactions among cytoplasmic<br />

dynein, dynactin, and nuclear distribution mutants of Neurospora crassa.<br />

Proc Natl Acad Sci USA 93:4775–4780<br />

Burgess T, Laurent P, Dell B, Malajczuk N, Martin F (1995) Effect of fungal-isolate aggressivity<br />

on the biosynthesis of symbiosis-related polypeptides in differentiating eucalypt<br />

ectomycorrhizas. Planta 195:408–417<br />

Bütehorn B, Gianinazzi-Pearson V, Franken P (1999) Quantification of b-tubulin RNA<br />

expression during asymbiotic and symbiotic development of the arbuscular mycorrhizal<br />

fungus Glomus mosseae. Mycol Res 103:360–364<br />

Canut H, Carrasco A, Galaud J-P, Cassan C, Bouyssou H, Vita N, Ferrara P, Pont-Lezica R<br />

(1998) High affinity RGD-binding sites at the plasma membrane of Arabidopsis<br />

thaliana links the cell wall. Plant J 16:63–71<br />

Chaffey N, Barlow P, Barnett J (2000) A cytoskeletal basis for wood formation in<br />

angiosperm trees: the involvement of microfilaments. Planta 210:890–896<br />

Chan J, Rutten T, Lloyd C (1996) Isolation of microtubule-associated proteins from carrot<br />

cytoskeletons: a 120 kDa map decorates all four microtubule arrays and the<br />

nucleus. Plant J 10:251–259<br />

Chan J, Jensen CG, Jensen LCW, Bush M, Lloyd CW (1999) The 65-kDa carrot microtubule-associated<br />

protein forms regularly arranged filamentous cross-bridges<br />

between microtubules. Pro Natl Acad Sci USA 96:14931–14936<br />

Chu B, Wilson TJ, McCune-Zierath C, Snustad DP, Carter JV (1998) Two b-tubulin genes,<br />

TUB1 and TUB8, ofArabidopsis exhibit largely nonoverlapping patterns of expression.<br />

Plant Mol Biol 37:785–790<br />

Cleary AL (1995) F-actin redistributions at the division site in living Tradescantia stomatal<br />

complexes as revealed by microinjection of rhodamine-phalloidin. Protoplasma<br />

185:152–165<br />

Cole L, Orlovich DA, Ashford AE (1998) Structure, function, and motility of vacuoles in<br />

filamentous fungi. Fungal Genet Biol 24:86–100<br />

Cooper JA, Schafer DA (2000) Control of actin assembly and disassembly at filament<br />

ends. Curr Opin Cell Biol 12:97–103<br />

Daniels RH, Bokoch GM (1999) p21-activated protein kinase: a crucial component of<br />

morphological signaling? TIBS 24:350–355<br />

de Ruijter NCA, Esseling JJ, Emons AMC (2001) The roles of calcium and the actin<br />

cytoskeleton in regulation of root hair tip growth by rhizobial signal molecules. In:<br />

Geitmann A, Cresti M, Heath IB (eds) Cell biology of <strong>plant</strong> and fungal tip growth. IOS<br />

Press, NATO Sci Ser 328:55–67<br />

De Veylder L, de Almeida Engler J, Burssens S, Manevski A, Lescure B, van Montagu M,<br />

Engler G, Inze D (1999) A new D-type cyclin of Arabidopsis thaliana expressed during<br />

lateral root primordial formation. Planta 208:453–462<br />

Diaz EC, Martin F, Tagu D (1996) Eucalypt a-tubulin: cDNA cloning and increased level<br />

of transcripts in ectomycorrhizal root system. Plant Mol Biol 31:905–910<br />

Dong C-H, Kost B, Xia G, Chua N-H (2001) Molecular identification and characterization<br />

of the Arabidopsis AtADF1, AtADF5 and AtADF6 genes. Plant Mol Biol 45:517–527<br />

Doyle T, Botstein D (1996) Movement of yeast cortical actin cytoskeleton visualized in<br />

vivo. Proc Natl Acad Sci USA 93:3886–3891<br />

Dutcher SK (2001) The tubulin fraternity: alpha to eta. Curr Opin Cell Biol 13:49–54<br />

Enos AP, Morris NR (1990) Mutation of a gene that encodes a kinesin-like protein blocks<br />

nuclear division in A. nidulans. Cell 60:1019–1027<br />

Euteneuer U, McIntosh JR (1980) Polarity of midbody and phragmoplast microtubules.<br />

J Cell Biol 87:509–515<br />

Fischer R (1999) Nuclear movement in filamentous fungi. FEMS Microbiol Rev<br />

23:38–69


18 Mycorrhizal Development and Cytoskeleton 321<br />

Fowler TJ, Desimone SM, Mitton MF, Kurjan J, Raper CA (1999) Multiple sex pheromones<br />

and receptors of a mushroom-producing fungus elicit mating in yeast. Mol Biol Cell<br />

10:2559–2572<br />

Franken P, Lapopin L, Meyer-Gauen G, Gianinazzi-Pearson V (1997) RNA accumulation<br />

and genes expressed in spores of the arbuscular mycorrhizal fungus, Gigaspora rosea.<br />

Mycol 89:293–297<br />

Fu Y,Wu G,Yang Z (2001) Rop GTPase-dependent dynamics of tip-localized F-actin controls<br />

tip growth in pollen tubes. J Cell Biol 152:1019–1032<br />

Genre A, Bonfante P (1997) A mycorrhizal fungus changes microtubule orientation in<br />

tobacco root cells. Protoplasma 199:30–38<br />

Genre A, Bonfante P (1998) Actin versus tubulin configuration in arbuscule-containing<br />

cells from mycorrhizal tobacco roots. New Phytol 140:745–752<br />

Giovannetti M, Azzolini D, Citernesi AS (1999) Anastomosis formation and nuclear and<br />

protoplasmic exchange in arbuscular mycorrhizal fungi. Appl Environ Microbiol<br />

65:5571–5575<br />

Giovannetti M, Fortuna P, Citernesi AS, Morini S, Nuti MP (2001) The occurrence of<br />

anastomosis formation and nuclear exchange in intact arbuscular mycorrhizal networks.<br />

New Phytol 151:717–724<br />

Goode BL, Drubin DG, Barnes G (2000) Functional cooperation between the microtubule<br />

and actin cytoskeletons. Curr Opin Cell Biol 12:63–71<br />

Gorfer M, Tarkka MT, Hanif M, Pardo AG, Laitiainen E, Raudaskoski M (2001) Characterization<br />

of small GTPases Cdc42 and Rac and the relationship between Cdc42 and<br />

actin cytoskeleton in vegetative and ectomycorrhizal hyphae of Suillus bovinus.<br />

MPMI 14:135–144<br />

Grummt M, Pistor S, Lottspeich F, Schliwa M (1998) Cloning and functional expression<br />

of a ‘fast’ fungal kinesin. FEBS Lett 427:79–84<br />

Hahn M, Mendgen K (2001) Signal and nutrient exchange at biotrophic <strong>plant</strong>-fungus<br />

interfaces. Curr Opin Plant Biol 4:322–327<br />

Han G, Liu B, Zhang J, Zuo W, Morris NR, Xiang X (2001) The Aspergillus cytoplasmic<br />

dynein heavy chain and NUDF localize to microtubule ends and affect microtubule<br />

dynamics. Curr Biol 11:719–724<br />

Heath IB, Gupta G, Bai S (2000) Plasma membrane-adjacent actin filaments, but not<br />

microtubules, are essential for both polarization and hyphal tip morphogenesis in<br />

Saprolegnia ferax and Neurospora crassa. Fungal Genet Biol 30:45–62<br />

Hepler PK, Vidali L, Cheung AY (2001) Polarized cell growth in higher <strong>plant</strong>s. Annu Rev<br />

Cell Dev Biol 17:159–187<br />

Hirsch AM, Fang Y (1994) Plant hormones and nodulation: what’s the connection? Plant<br />

Mol Biol 26:5–9<br />

Huang S, An Y-Q, McDowell JM, McKinney EC, Meagher RB (1997) The Arabidopsis<br />

ACT11 actin gene is strongly expressed in tissues of the emerging inflorescence,<br />

pollen, and developing ovules. Plant Mol Biol 33:125–139<br />

Hussey PJ, Haas N, Hunsperger J, Larkin J, Snustad DP, Silflow CD (1990) The b-tubulin<br />

gene family in Zea mays: two differentially expressed b-tubulin genes. Plant Mol Biol<br />

15:957–972<br />

Hussey PJ,Yuan M, Calder G, Khan S, Lloyd CW (1998) Microinjection of pollen-specific<br />

actin-depolymerizing factor, ZmADF1, reorientates F-actin strands in Tradescantia<br />

stamen hair cells. Plant J 14:353–357<br />

Hyde GJ, Davies D, Perasso L, Cole L, Ashford AE (1999) Microtubules, but not actin<br />

microfilaments, regulate vacuole motility and morphology in hyphae of Pisolithus<br />

tinctorius. Cell Motil Cytoskel 42:114–124<br />

Inoue S, Turgeon BG, Yoder OC, Aist JR (1998) Role of fungal dynein in hyphal growth,<br />

microtubule organization, spindle pole body motility and nuclear migration. J Cell<br />

Sci 111:1555–1566


322<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

Janssen M, Hunte C, Schulz M, Schnabl H (1996) Tissue specification and intracellular<br />

distribution of actin isoforms in Vicia faba L. Protoplasma 191:158–163<br />

Jiang C-J, Sonobe S (1993) Identification and preliminary characterization of a 65 kDa<br />

higher-<strong>plant</strong> microtubule-associated protein. J Cell Sci 105:891–901<br />

Jiang C-J,Weeds AG, Hussey PJ (1997) The maize actin-depolymerizing factor, ZmADF3,<br />

redistributes to the growing tip of elongating root hairs and can be induced to<br />

translocate into the nucleus with actin. Plant J 12:1035–1043<br />

John PCL, Mews M, Moore R (2001) Cyclin/Cdk complexes: their involvement in cell<br />

cycle progression and mitotic division. Protoplasma 216:119–142<br />

Johnson DJ (1999) Cdc42: an essential rho-type GTPase controlling eukaryotic cell<br />

polarity. Microbiol Mol Biol Rev 63:54–105<br />

Johnson DJ, Pringle JR (1990) Molecular characterization of CDC42, a Saccharomyces<br />

cerevisiae gene involved in the development of cell polarity. J Cell Biol 111:143–152<br />

Joshi HC, Palevitz, BA (1996) g-Tubulin and microtubule organization in <strong>plant</strong>s. Trends<br />

Cell Biol 6:41–44<br />

Joshi HC, Palacios MJ, McNamara L, Cleveland DW (1992) g-Tubulin is a centrosomal<br />

protein required for cell cycle-dependent microtubule nucleation. Nature 356:80–83<br />

Joyce CM, Villemur R, Snustad DP, Silflow CD (1992) Tubulin gene expression in maize<br />

(Zea mays L.). Change in isotype expression along the developmental axis of seedling<br />

root. J Mol Biol 227:97–107<br />

Kallipolitou A, Deluca D, Majdic U, Lakämper S, Cross R, Meyhöfer E, Moroder L, Schliwa<br />

M, Woehlke G (2001) Unusual properties of the fungal conventional kinesin neck<br />

domain from Neurospora crassa. EMBO J 20:6226–6235<br />

Kandasamy MK, Gilliland LU, McKinney EC, Meagher RB (2001) One <strong>plant</strong> actin isovariant,<br />

ACT7, is induced by auxin and required for normal callus formation. Plant<br />

Cell 13:1541–1554<br />

Kaska DD, Myllylä R, Cooper JB (1999) Auxin transport inhibitors act through ethylene<br />

to regulate dichotomous branching of lateral root meristems in pine. New Phytol<br />

142:49–58<br />

Kerr GP, Carter JV (1990a) Tubulin isotypes in rye roots are altered during cold acclimation.<br />

Plant Physiol 93:83–88<br />

Kilmartin JV,Adams AEM (1984) Structural rearrangements of tubulin and actin during<br />

the cell cycle of the yeast Saccharomyces. J Cell Biol 98:922–933<br />

Kim AJ, Endow SA (2000) A kinesin family tree. J Cell Sci 113:3681–3682<br />

Kim M, Hepler PK, Eun S-O, Ha KS, Lee Y (1995) Actin filaments in mature guard cells are<br />

radially distributed and involved in stomatal movement. Plant Physiol 109:1077–1084<br />

King SJ, Schroer TA (2000) Dynactin increases the processivity of the cytoplasmic<br />

dynein motor. Nat Cell Biol 2:20–24<br />

Klahre U, Chua N (1999) The Arabidopsis actin-related protein 2 (AtARP2) promoter<br />

directs expression in xylem precursor cells and pollen. Plant Mol Biol 41:65–73<br />

Klahre U, Friederich E, Kost B, Louvard D, Chua N-H (2000) Villin-like actin-binding<br />

proteins are expressed ubiquitously in Arabidopsis. Plant Physiol 122:35–47<br />

Ko KS, McCulloch CA (2000) Partners in protection: interdependence of cytoskeleton<br />

and plasma membrane in adaptations to applied forces. J Membr Biol 174:85–95<br />

Kopczak SD, Haas NA, Hussey PJ, Silflow CD, Snustad DP (1992) The small genome of<br />

Arabidopsis contains at least six expressed a-tubulin genes. Plant Cell 4:539–547<br />

Kost B, Spielhofer P, Chua N-H (1998) A GFP-mouse talin fusion protein labels <strong>plant</strong><br />

actin filaments in vivo and visualizes the actin cytoskeleton in growing pollen tubes.<br />

Plant J 16:393–401<br />

Kost B, Lemichez E, Spielhofer P, Hong Y, Tolias K, Carpenter C, Chua N-H (1999a) Rac<br />

homologues and compartmentalized phosphatidylinositol 4,5-bisphosphate act in a<br />

common pathway to regulate polar pollen tube growth. J Cell Biol 145:317–330


18 Mycorrhizal Development and Cytoskeleton 323<br />

Kost B, Mathur J, Chua N-H (1999b) Cytoskeleton in <strong>plant</strong> development. Curr Opin Cell<br />

Biol 2:462–470<br />

Kottke I, Oberwinkler F (1987) The cellular structure of the Hartig net: coenocytic and<br />

transfer cell-like organization. Nord J Bot 7:85–95<br />

Lancelle SA, Cresti M, Hepler PK (1997) Growth inhibition and recovery in freeze-substituted<br />

Lilium longiflorum pollen tubes: structural effects of caffeine. Protoplasma<br />

196:21–33<br />

Lawrence CJ, Malmberg RL, Muszynski MG, Dawe RK (2002) Maximum likelihood<br />

methods reveal conservation of function among closely related kinesin families. J Mol<br />

Evol 54:42–53<br />

Lee IH, Kumar S, Plamann M (2001) Null mutants of the Neurospora actin-related protein<br />

1 pointed-end complex show distinct phenotypes. Mol Biol Cell 12:2195–2206<br />

Lehmler C, Steinberg G, Snetselaar KM, Schliwa M, Kahmann R, Bölker M (1997) Identification<br />

of a motor protein required for filamentous growth in Ustilago maydis.<br />

EMBO J 16:3464–3473<br />

Lemichez E, Wu Y, Sanchez J-P, Mettouchi A, Mathur J, Chua N-H (2001) Inactivation of<br />

AtRac1 by abscisic acid is essential for stomatal closure. Genes Dev 15:1808–1816<br />

Li H, Shen J-J, Zheng Z-L, Lin Y, Yang Z (2001) The Rop GTPase switch controls multiple<br />

developmental processes in Arabidopsis. Plant Physiol 126:670–684<br />

Liu B, Marc J, Joshi HC, Palevitz BA (1993) A g-tubulin-related protein associated with<br />

the microtubule arrays of higher <strong>plant</strong>s in a cell cycle-dependent manner. J Cell Sci<br />

104:1217–1228<br />

Liu B, Cyr RJ, Palevitz BA (1996) A kinesin-like protein, KatAp, in the cells of Arabidopsis<br />

and other <strong>plant</strong>s. Plant Cell 8:119–132<br />

Lloyd C, Hussey P (2001) Microtubule-associated proteins in <strong>plant</strong>s – why we need a<br />

map. Nat Rev Mol Cell Biol 2:40–47<br />

Lopez I, Anthony RG, Maciver SK, Jiang C-J, Khan S, Weeds AG, Hussey PJ (1996) Pollen<br />

specific expression of maize genes encoding actin depolymerizing factor-like proteins.<br />

Proc Natl Acad Sci USA 93:7415–7420<br />

Lugones LG, Scholtmeijer K, Klootwijk R, Wessels JGH (1999) Introns are necessary for<br />

mRNA accumulation in Schizophyllum commune. Mol Microbiol 32:681–689<br />

Ma B, Mayfield MB, Gold MH (2001) The green fluorescent protein gene functions as a<br />

reporter of gene expression in Phanerochaete chrysosporium.Appl Environ Microbiol<br />

67:948–955<br />

Marc J, Granger CL, Brincat J, Fisher DD, Kao T-H, McCubbin AG, Cyr RJ (1998) A GFP-<br />

MAP4 reporter gene for visualizing cortical microtubule rearrangements in living<br />

epidermal cells. Plant Cell 10:1927–1939<br />

Mathesius U, Schlaman HRM, Spaink HP, Sautter C, Rolfe BG, Djordjevic MA (1998)<br />

Auxin transport inhibition precedes root nodule formation in white clover roots and<br />

is regulated by flavonoids and derivatives of chitin oligosaccharides. Plant J 14:23–34<br />

Matsubara Y, Uetake Y, Peterson RL (1999) Entry and colonization of Asparagus officinalis<br />

roots by arbuscular mycorrhizal fungi with emphasis on changes in host microtubules.<br />

Can J Bot 77:1159–1167<br />

McGoldrick CA, Gruver C, May GS (1995) myoA of Aspergillus nidulans encodes an<br />

essential myosin I required for secretion and polarized growth. J Cell Biol 128:577–<br />

587<br />

McLean BG, Eubanks S, Meagher RB (1990) Tissue-specific expression of divergent<br />

actins in soybean root. Plant Cell 2:335–344<br />

Meagher RB, McKinney EC,Vitale AV (1999) The evolution of new structures: clues from<br />

<strong>plant</strong> cytoskeletal genes. Trends Genet 15:278–284<br />

Mews M, Sek FJ, Moore R, Volkmann D, Gunning BES, John PCL (1997) Mitotic cyclin<br />

distribution during maize cell division: implications for the sequence diversity and<br />

function of cyclins in <strong>plant</strong>s. Protoplasma 200:128–145


324<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

Miki H, Setou M, Kaneshiro K, Hirokawa N (2001) All kinesin superfamily protein, KIF,<br />

genes in mouse and human. Pro Natl Acad Sci USA 98:7004–7011<br />

Miller DD, Lancelle SA, Hepler PK (1996) Actin microfilaments do not form a dense network<br />

in Lilium longiflorum pollen tube tips. Protoplasma 195:123–132<br />

Miller DD, de Ruijter NCA, Bisseling T, Emons AMC (1999) The role of actin in root hair<br />

morphogenesis: studies with lipochito-oligosaccharide as a growth stimulator and<br />

cytochalasin as an actin perturbing drug. Plant J 17:141–154<br />

Mironov V, de Veylder L, van Montagu M, Inze D (1999) Cyclin-dependent kinases and<br />

cell division in <strong>plant</strong>s – the nexus. Plant Cell 11:509–521<br />

Mitsui H, Yamaguchi-Shinozaki K, Shinozaki K, Nishikawa K, Takahashi H (1993) Identification<br />

of a gene family (kat) encoding kinesin-like proteins in Arabidopsis<br />

thaliana and the characterization of secondary structure of KatA. Mol Gen Genet<br />

238:362–368<br />

Mitsui H, Nakatani K, Yamaguchi-Shinozaki K, Shinozaki K, Nishikawa K, Takahashi H<br />

(1994) Sequencing and characterization of the kinesin-related genes katB and katC of<br />

Arabidopsis thaliana. Plant Mol Biol 25:865–876<br />

Mitsui H, Hasezawa S, Nagata T, Takahashi H (1996) Cell cycle-dependent accumulation<br />

of a kinesin-like protein, KatB/C, in synchronized tobacco BY-2 cells. Plant Mol Biol<br />

30:177–181<br />

Molendijk AJ, Bischoff F, Rajendrakumar CSV, Friml J, Braun M, Gilroy S, Palme K (2001)<br />

Arabidopsis thaliana Rop GTPases are localized to tips of root hairs and control polar<br />

growth. EMBO J 20:2779–2788<br />

Montoliu L, Rigau J, Puigdomènech P (1989) A tandem of a-tubulin genes preferentially<br />

expressed in radicular tissues from Zea mays. Plant Mol Biol 14:1–15<br />

Morris NR, Xiang X, Beckwith SM (1995) Nuclear migration advances in fungi. Trends<br />

Cell Biol 5:278–282<br />

Mullins RD (2000) How WASP-family proteins and the Arp2/3 complex convert intracellular<br />

signals into cytoskeletal structures. Curr Opin Cell Biol 12:91–96<br />

Mylona P, Pawlowski K, Bisseling T (1995) Symbiotic nitrogen fixation. Plant Cell<br />

7:869–885<br />

Neer EJ (1995) Heterotrimeric G proteins: organizers of transmembrane signals. Cell<br />

80:249–257<br />

Niini S (1998) Growth pattern and cytoskeleton in Suillus bovinus hyphae, Pinus<br />

sylvestris roots and in Pinus sylvestris–Suillus bovinus ectomycorrhiza. Dissertationes<br />

Biocentri Viikki Universitatis Helsingiensis 16<br />

Niini S, Raudaskoski M (1998) Growth patterns in non-mycorrhizal and mycorrhizal<br />

short roots of Pinus sylvestris. Symbiosis 25:101–114<br />

Niini SS, Tarkka MT, Raudaskoski M (1996) Tubulin and actin protein patterns in Scots<br />

pine (Pinus sylvestris) roots and developing ectomycorrhiza with Suillus bovinus.<br />

Physiol Plant 96:186–192<br />

Nuoffer C, Balch WE (1994) GTPases: multifunctional molecular switches regulating<br />

vesicular traffic. Annu Rev Biochem 63:949–990<br />

Oakley BR, Oakley CE, Yoon Y, Jung MK (1990) g-Tubulin is a component of the spindle<br />

pole body that is essential for microtubule function in Aspergillus nidulans. Cell<br />

61:1289–1301<br />

Olesnicky NS, Brown AJ, Dowell SJ, Casselton LA (1999) A constitutively active G-protein-coupled<br />

receptor causes mating self-compatibility in the mushroom Coprinus.<br />

EMBO J 18:2756–2763<br />

Olinevich OV, Khokhlova LP, Raudaskoski M (2001) Effect of abscisic acid and cold acclimation<br />

on the cytoskeletal and phosphorylated proteins in different cultivars of<br />

Triticum aestivum L. Cell Biol Int 24:365–373<br />

Osherov N, Yamashita RA, Chung YS, May GS (1998) Structural requirements for in vivo<br />

myosin I function in Aspergillus nidulans. J Biol Chem. 273:27017–27025


18 Mycorrhizal Development and Cytoskeleton 325<br />

Overall RL, Dibbayawan TP, Blackman LM (2001) Intercellular alignments of the <strong>plant</strong><br />

cytoskeleton. J Plant Growth Regul 20:162–169<br />

Pardo AG, Hanif M, Raudaskoski M, Gorfer M (2002) Genetic transformation of ectomycorrhizal<br />

fungi mediated by Agrobacterium tumefaciens. Mycol Res 106:132–137<br />

Pérez HE, Sánchez N, Vidali L, Hernández JM, Lara M, Sánchez F (1994) Actin isoforms<br />

in non-infected roots and symbiotic root nodules of Phaseolus vulgaris L. Planta<br />

193:51–56<br />

Pesacreta TC, Carley WW, Webb WW, Parthasarathy MV (1982) F-actin in conifer roots.<br />

Proc Natl Acad Sci USA 79:2898–2901<br />

Peterson RL, Bonfante P, Faccio A, Uetake Y (1996) The interface between fungal hyphae<br />

and orchid protocorm cells. Can J Bot 74:1861–1870<br />

Piché Y, Peterson RL, Ackerley CA (1983) Early development of ectomycorrhizal short<br />

roots of pine. Scan Electron Microsc III:1467–1474<br />

Plamann M, Minke PF, Tinsley JH, Bruno KS (1994) Cytoplasmic dynein and actinrelated<br />

protein Arp1 are required for normal nuclear distribution in filamentous<br />

fungi. J Cell Biol 127:139–149<br />

Ramachandran S, Christensen HEM, Ishimaru Y, Dong C-H, Chao-Ming W, Cleary AL,<br />

Chua N-H (2000) Profilin plays a role in cell elongation, cell shape maintenance, and<br />

flowering in Arabidopsis. Plant Physiol 124:1637–1647<br />

Raudaskoski M (1998) The relationship between B-mating-type genes and nuclear<br />

migration in Schizophyllum commune. Fungal Genet Biol 24:207–227<br />

Raudaskoski M, Åström H, Penttilä K, Virtanen I, Louhelainen J (1987) Role of microtubule<br />

cytoskeleton in pollen tubes: an immunocytochemical and ultrastructural<br />

approach. Biol Cell 61:177–188<br />

Raudaskoski M, Rupe_ I, Timonen S (1991) Immunofluorescence microscopy of the<br />

cytoskeleton in filamentous fungi after quick-freezing and low-temperature fixation.<br />

Exp Mycol 15:167–173<br />

Raudaskoski M, Mao W-Z, Yli-Mattila T (1994) Microtubule cytoskeleton in hyphal<br />

growth. Response to nocodazole in a sensitive and a tolerant strain of the homobasidiomycete<br />

Schizophyllum commune. Eur J Cell Biol 64:131–141<br />

Raudaskoski M, Färdig M, Uuskallio M (1998) The structure of pheromone and receptor<br />

gene transcripts in Ba1 and Bb1 mating-type loci of Schizophyllum commune.In:Van<br />

Griensven LJLD,Visser J (eds) Proceedings of the fourth meeting on the genetics and<br />

cellular biology of basidiomycetes. Mushroom Experimental Station, Horst, Netherlands,<br />

pp 119–124<br />

Raudaskoski M, Pardo AG, Tarkka MT, Gorfer M, Hanif M, Laitiainen E (2001) Small<br />

GTPases, cytoskeleton and signal transduction in filamentous homobasidiomycetes.<br />

In: Geitmann A, Cresti M, Heath IB (eds) Cell biology of <strong>plant</strong> and fungal tip growth.<br />

IOS Press, NATO Sci Ser 328:123–136<br />

Reddy ASN, Day IS (2001) Analysis of the myosins encoded in the recently completed<br />

Arabidopsis thaliana genome sequence. Genome Biol 2:research0024.1–0024.17<br />

Reddy ASN, Safadi F, Narasimhulu SB, Golovkin M, Hu X (1996a) A novel <strong>plant</strong> calmodulin-binding<br />

protein with a kinesin heavy chain motor domain. J Biol Chem 271:<br />

7052–7060<br />

Reddy ASN, Narasimhulu SB, Safadi F, Golovkin M (1996b) A <strong>plant</strong> kinesin heavy chainlike<br />

protein is a calmodulin-binding protein. Plant J 10:9–21<br />

Requena N,Alberti-Segui C,Winzenburg E, Horn C, Schliwa M, Philippsen P, Liese R, Fischer<br />

R (2001) Genetic evidence for a microtubule-destabilizing effect of conventional<br />

kinesin and analysis of its consequences for the control of nuclear distribution in<br />

Aspergillus nidulans. Mol Microbiol 42:121–132<br />

Robertson NF (1954) Studies on the mycorrhiza of Pinus sylvestris. I. The pattern of<br />

development of mycorrhizal roots and its significance for experimental studies. New<br />

Phytol 53:253–283


326<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

Rosewarne GM, Barker SJ, Smith SE, Smith FA, Schachtman DP (1999) A Lycopersicon<br />

esculentum phosphate transporter (LePT1) involved in phosphorus uptake from a<br />

vesicular-arbuscular mycorrhizal fungus. New Phytol 144:507–516<br />

Runeberg P,Virtanen I, Raudaskoski M (1986) Cytoskeletal elements in the hyphae of the<br />

homobasidiomycete Schizophyllum commune visualized with indirect immunofluorescence<br />

and NBD-phallacidin. Eur J Cell Biol 41:25–32<br />

Russo P, Juuti JT, Raudaskoski M (1992) Cloning, sequence and expression of a b-tubulin-encoding<br />

gene in the homobasidiomycete Schizophyllum commune. Gene<br />

119:175–182<br />

Salo V, Niini SS, Virtanen I, Raudaskoski M (1989) Comparative immunocytochemistry<br />

of the cytoskeleton in filamentous fungi with dikaryotic and multinucleate hyphae. J<br />

Cell Sci 94:11–24<br />

Schmit A-C, Lambert A-M (1990) Microinjected fluorescent phalloidin in vivo reveals<br />

the F-actin dynamics and assembly in higher <strong>plant</strong> mitotic cells. Plant Cell 2:129–138<br />

Schmidt A, Hall MN (1998) Signaling to the actin cytoskeleton. Annu Rev Cell Dev Biol<br />

14:305–338<br />

Seiler S, Nargang FE, Steinberg G, Schliwa M (1997) Kinesin is essential for cell morphogenesis<br />

and polarized secretion in Neurospora crassa. EMBO J 16:3025–3034<br />

Seiler S, Plamann M, Schliwa M (1999) Kinesin and dynein mutants provide novel<br />

insights into the roles of vesicle traffic during cell morphogenesis in Neurospora.Curr<br />

Biol 9:779–785<br />

Sheng J, Citovsky V (1996) Agrobacterium-<strong>plant</strong> cell DNA transport: Have virulence proteins,<br />

will travel. Plant Cell 8:1699–1710<br />

Shepherd VA, Orlovich DA,Ashford AE (1993) Cell-to-cell transport via motile tubules in<br />

growing hyphae of a fungus. J Cell Sci 105:1173–1178<br />

Slankis V (1949) Wirkung von alpha-indolylessigsäure auf die dichotomische Verzweigung<br />

isolierter wurzeln von Pinus sylvestris. Sven Bot Tidskr 43:603–607<br />

Smertenko AP, Jiang C-J, Simmons NJ,Weeds AG, Davies DR, Hussey PJ (1998) Ser6 in the<br />

maize actin-depolymerizing factor, ZmADF3, is phosphorylated by a calcium-stimulated<br />

protein kinase and is essential for the control of functional activity. Plant J<br />

14:187–193<br />

Smertenko A, Saleh N, Igarashi H, Mori H, Hauser-Hahn I, Jiang C-J, Sonobe S, Lloyd CW,<br />

Hussey P (2000) A new class of microtubule-associated proteins in <strong>plant</strong>s. Nat Cell<br />

Biol 2:750–753<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis. Second edition. Academic Press, San<br />

Diego, CA<br />

Snustad DP, Haas NA, Kopczak SD, Silflow CD (1992) The small genome of Arabidopsis<br />

contains at least nine expressed b-tubulin genes. Plant Cell 4:549–556<br />

Sonobe S, Shibaoka H (1989) Cortical fine actin filaments in higher <strong>plant</strong> cells visualized<br />

by rhodamine-phalloidin after pretreatment with m-maleimidobenzoyl N-hydroxysuccinimide<br />

ester. Protoplasma 148:80–86<br />

Staiger CJ (2000) Signaling to the actin cytoskeleton in <strong>plant</strong>s. Annu Rev Plant Physiol<br />

Plant Mol Biol 51:257–263<br />

Stamatas GN, McIntire LV (2001) Rapid flow-induced responses in endothelial cells.<br />

Biotechnol Prog 17:383–402<br />

Steinberg G (1997) A kinesin-like mechanoenzyme from the zygomycete Syncephalastrum<br />

racemosum shares biochemical similarities with conventional kinesin from<br />

Neurospora crassa. Eur J Cell Biol 73:124–131<br />

Steinberg G (1998) Organelle transport and molecular motors in fungi. Fungal Genet<br />

Biol 24:161–177<br />

Steinberg G (2000) The cellular roles of molecular motors in fungi. Trends Microbiol<br />

8:162–168


18 Mycorrhizal Development and Cytoskeleton 327<br />

Steinberg G, Schliwa M (1995) The Neurospora organelle motor: a distant relative of conventional<br />

kinesin with unconventional properties. Mol Biol Cell 6:1605–1618<br />

Steinberg G, Schliwa M (1996) Characterization of the biophysical and motility properties<br />

of kinesin from the fungus Neurospora crassa. J Biol Chem 271:7516–7521<br />

Steinberg G, Schliwa M, Lehmler C, Bölker M, Kahmann R, McIntosh JR (1998) Kinesin<br />

from the <strong>plant</strong> pathogenic fungus Ustilago maydis is involved in vacuole formation<br />

and cytoplasmic migration. J Cell Sci 111:2235–2246<br />

Straight AF, Marshall WF, Sedat JW, Murray AW (1997) Mitosis in living budding yeast:<br />

anaphase a but no metaphase plate. Science 277:574–578<br />

Straube A, Enard W, Berner A, Wedlich-Söldner R, Kahmann R, Steinberg G (2001) A<br />

split motor domain in a cytoplasmic dynein. EMBO J 20:5091–5100<br />

Suelmann R, Fischer R (2000) Nuclear migration in fungi – different motors at work. Res<br />

Microbiol 151:247–254<br />

Sundaram S, Kim SJ, Suzuki H, Mcquattie CJ, Hiremah ST, Podila GK (2001) Isolation and<br />

characterization of a symbiosis-regulated ras from the ectomycorrhizal fungus Laccaria<br />

bicolour. MPMI 14:618–628<br />

Tarkka MT (2001) Developmentally regulated proteins in Scots pine roots and ectomycorrhiza.<br />

PhD Thesis, University of Helsinki, Finland. Yliopistopaino, Helsinki, Finland.<br />

ISBN 951–45–9645–5<br />

Tarkka MT, Niini SS, Raudaskoski M (1998) Developmentally regulated proteins during<br />

differentiation of root system and ectomycorrhiza in Scots pine (Pinus sylvestris)<br />

with Suillus bovinus. Physiol Plant 104:449–455<br />

Tarkka MT, Vasara R, Gorfer M, Raudaskoski M (2000) Molecular characterization of<br />

actin genes from homobasidiomycetes: two different actin genes from Schizophyllum<br />

commune and Suillus bovinus. Gene 251:27–35<br />

Tarkka MT, Nyman TA, Kalkkinen N, Raudaskoski M (2001) Scots pine expresses shortroot-specific<br />

peroxidases during development. Eur J Biochem 268:86–92<br />

Timonen S, Finlay RD, Söderström B, Raudaskoski M (1993) Identification of cytoskeletal<br />

components in pine ectomycorrhizas. New Phytol 124:83–92<br />

Timonen S, Söderström B, Raudaskoski M (1996) Dynamics of cytoskeletal proteins in<br />

developing pine ectomycorrhiza. Mycorrhiza 6:423–429<br />

Tinsley JH, Minke PF, Bruno KS, Plamann M (1996) p150 Glued , the largest subunit of the<br />

dynactin complex, is nonessential in Neurospora but required for nuclear distribution.<br />

Mol Biol Cell 7:731–742<br />

Ueda K, Matsuyama T, Hashimoto T (1999) Visualization of microtubules in living cells<br />

of transgenic Arabidopsis thaliana. Protoplasma 206:201–206<br />

Uetake Y, Peterson RI (1997) Changes in actin filament arrays in protocorm cells of the<br />

orchid species, Spiranthes sinensis, induced by the symbiotic fungus Ceratobasidium<br />

cornigerum. Can J Bot 75:1661–1669<br />

Uetake Y, Peterson RI (1998) Association between microtubules and symbiotic fungal<br />

hyphae in protocorm cells of the orchid species, Spiranthes sinensis. New Phytol<br />

140:715–722<br />

Uetake Y, Farquhar ML, Peterson RL (1997) Changes in microtubule arrays in symbiotic<br />

orchid protocorms during fungal colonization and senescence. New Phytol 135:701–<br />

709<br />

Uribe X, Torres MA, Capellades M, Puigdomènech P, Rigau J (1998) Maize a-tubulin<br />

genes are expressed according to specific patterns of cell differentiation. Plant Mol<br />

Biol 37:1069–1078<br />

Vaillancourt LJ, Raudaskoski M, Specht CA, Raper CA (1997) Multiple genes encoding<br />

pheromones and a pheromone receptor define the Bb1 mating-type specificity in<br />

Schizophyllum commune. Genetics 146:541–551<br />

Vale RD, Milligan RA (2000) The way things move: looking under the hood of molecular<br />

motor proteins. Science 288:88–95


328<br />

Marjatta Raudaskoski, Mika Tarkka and Sara Niini<br />

Vantard M, Cowling R, Delichere C (2000) Cell cycle regulation of the microtubular<br />

cytoskeleton. Plant Mol Biol 43:691–703<br />

Versele M, Lemaire K, Thevelein JM (2001) Sex and sugar in yeast: two distinct GPCR<br />

systems. EMBO Rep 2:574–579<br />

Vidali L,Yokota E, Cheung AY, Shimmen T, Hepler PK (1999) The 135 kDa actin-bundling<br />

protein from Lilium longiflorum pollen is the <strong>plant</strong> homologue of villin. Protoplasma<br />

209:283–291<br />

Villemur R, Joyce CM, Haas NA, Goddard RH, Kopczak SD, Hussey PJ, Snustad DP, Silflow<br />

CD (1992) Alpha-tubulin gene family of maize (Zea mays L.). Evidence for two<br />

ancient alpha-tubulin genes in <strong>plant</strong>s. J Mol Biol 227:81–96<br />

Villemur R, Haas NA, Joyce CM, Snustad DP, Silflow CD (1994) Characterization of four<br />

new b-tubulin genes and their expression during male flower development in maize<br />

(Zea mays L.). Plant Mol Biol 24:295–315<br />

Wang W, Takezawa D, Narasimhulu SB, Reddy ASN, Poovaiah BW (1996) A novel kinesinlike<br />

protein with a calmodulin-binding domain. Plant Mol Biol 31:87–100<br />

Wasteneys GO (2000) The cytoskeleton and growth polarity. Curr Opin Plant Biol<br />

3:503–511<br />

Wasteneys GO (2002) Microtubule organization in the green kingdom: chaos or selforder?<br />

J Cell Sci 115:1345–1354<br />

Wendland J, Vaillancourt LJ, Hegner J, Lengeler KB, Laddison KJ, Specht CA, Raper CA,<br />

Kothe E (1995) The mating-type locus Ba1of Schizophyllum commune contains a<br />

pheromone receptor gene and putative pheromone genes. EMBO J 14:5271–5278<br />

Whittington AT, Vugrek O, Wei KJ, Hasenbein NG, Sugimoto K, Rashbrooke MC,<br />

Wasteneys GO (2001) MOR1 is essential for organizing cortical microtubules in<br />

<strong>plant</strong>s. Nature 411:610–613<br />

Wilcox HE (1968) Morphological studies of the roots of red pine, Pinus resinosa. II. Fungal<br />

colonization of roots and the development of mycorrhizae. Am J Bot 55:688–700<br />

Wood V, Gwilliam R, Rajandream MA, Lyne M, Lyne R, Stewart A, Sgouros J, Peat N,<br />

Hayles J, Baker S, Basham D, Bowman S, Brooks K, Brown D, Brown S, Chillingworth<br />

T, Churcher C, Collins M, Connor R, Cronin A, Davis P, Feltwell T, Fraser A, Gentles S,<br />

Goble A, Hamlin N, Harris D, Hidalgo J, Hodgson G, Holroyd S, Hornsby T, Howarth<br />

S, Huckle EJ, Hunt S, Jagels K, James K, Jones L, Jones M, Leather S, McDonald S,<br />

McLean J, Mooney P, Moule S, Mungall K, Murphy L, Niblett D, Odell C, Oliver K,<br />

O’Neil S, Pearson D, Quail MA, Rabbinowitsch E, Rutherford K, Rutter S, Saunders D,<br />

Seeger K, Sharp S, Skelton J, Simmonds M, Squares R, Squares S, Stevens K, Taylor K,<br />

Taylor RG, Tivey A, Walsh S, Warren T, Whitehead S, Woodward J,Volckaert G, Aert R,<br />

Robben J, Grymonprez B,Weltjens I,Vanstreels E, Rieger M, Schafer M, Muller-Auer S,<br />

Gabel C, Fuchs M, Fritzc C, Holzer E, Moestl D, Hilbert H, Borzym K, Langer I, Beck A,<br />

Lehrach H, Reinhardt R, Pohl TM, Eger P, Zimmermann W, Wedler H, Wambutt R,<br />

Purnelle B, Goffeau A, Cadieu E, Dreano S, Gloux S, Lelaure V, Mottier S, Galibert F,<br />

Aves SJ, Xiang Z, Hunt C, Moore K, Hurst SM, Lucas M, Rochet M, Gaillardin C, Tallada<br />

VA, Garzon A, Thode G, Daga RR, Cruzado L, Jimenez J, Sanchez M, del Rey F, Benito<br />

J, Dominguez A, Revuelta JL, Moreno S, Armstrong J, Forsburg SL, Cerrutti L, Lowe T,<br />

McCombie WR, Paulsen I, Potashkin J, Shpakovski GV, Ussery D, Barrell BG, Nurse P<br />

(2002) The genome sequence of Schizosaccharomyces pombe. Nature 415:871–880<br />

Wu Q, Sandrock TM, Turgeon BG, Yoder OC, Wirsel SG, Aist JR (1998) A fungal kinesin<br />

required for organelle motility, hyphal growth, and morphogenesis. Mol Biol Cell<br />

9:89–101<br />

Xiang X, Beckwith SM, Morris NR (1994) Cytoplasmic dynein is involved in nuclear<br />

migration in Aspergillus nidulans. Proc Natl Acad Sci USA 91:2100–2104<br />

Yamashita RA, May GS (1998) Constitutive activation of endocytosis by mutation of<br />

myoA, the myosin I gene of Aspergillus nidulans. J Biol Chem 273:14644–14648


18 Mycorrhizal Development and Cytoskeleton 329<br />

Ye XS, McGuire SL, Wolkow T, Tang A, Fincher R, Hamer JE, Osmani SA (1999) Interaction<br />

between developmental and cell cycle regulators is required to morphogenesis in<br />

Aspergillus nidulans. EMBO J 18:6994–7001<br />

Yokota E, Shimmen T (1999) The 135-kDa actin-bundling protein from lily pollen tubes<br />

arranges F-actin into bundles with uniform polarity. Planta 209:264–266<br />

Yokota E, Takahara K, Shimmen T (1998) Actin-bundling protein isolated from pollen<br />

tubes of lily: biochemical and immunocytochemical characterization. Plant Physiol<br />

116:1421–1429<br />

Yuan M, Shaw PJ, Warn RM, Lloyd CW (1994) Dynamic reorientation of cortical microtubules,<br />

from transverse to longitudinal, in living <strong>plant</strong> cells. Proc Natl Acad Sci USA<br />

91:6050–6053<br />

Zhang DH, Callaham DA, Hepler PK (1990) Regulation of anaphase chromosome<br />

motion in Tradescantia stamen hair cells by calcium and related signalling agents. J<br />

Cell Biol 111:171–182<br />

Zhang D, Wadsworth P, Hepler PK (1993) Dynamics of microfilaments are similar, but<br />

distinct from microtubules during cytokinesis in living, dividing <strong>plant</strong> cells. Cell<br />

Motil Cytoskeleton 24:151–155<br />

Zheng Z-L, Yang Z (2000) The Rop GTPase switch turns on polar growth in pollen.<br />

Trends Plant Sci 5:298–303


19 Functional Diversity of Arbuscular Mycorrhizal<br />

Fungi on Root Surfaces<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

1 Introduction<br />

Arbuscular mycorrhizal (AM) fungi can promote host <strong>plant</strong> growth by<br />

increasing phosphorus (P) uptake from soil while simultaneously obtaining<br />

carbon (C) from the photosynthate of the host <strong>plant</strong>. However, reductions in<br />

<strong>plant</strong> growth associated with AM fungi have also been recorded (e.g. Graham<br />

and Eissenstat 1998; Graham and Abbott 2000) which can be linked to carbon<br />

and phosphorus exchange (Koide and Elliott 1989). Both growth promotion<br />

and reduction depend upon the particular <strong>plant</strong>–fungal combination (Johnson<br />

et al. 1997) and soil conditions. P and C exchange between the host <strong>plant</strong><br />

and mycorrhizal fungus also depends on environmental and biological variables<br />

(Jakobsen 1998). The combined effects of P uptake and transfer to the<br />

<strong>plant</strong> and C release to the fungus are important considerations for the functioning<br />

of arbuscular mycorrhizas. While the mechanisms of nutrient<br />

exchange between AM fungi and the host <strong>plant</strong> remain speculative (Schwab et<br />

al. 1991; Saito 2000; Smith et al. 2001), more is known about the <strong>plant</strong> genes<br />

involved in P transfer (Harrison 1999) than the fungal genes (Rausch et al.<br />

2001). Symbiotic exchange of nutrients in arbuscular mycorrhizas, especially<br />

transport along hyphae and transfer to the host <strong>plant</strong>, has been reviewed<br />

(Saito 2000; Smith et al. 2001) and it has been pointed out that the mechanisms<br />

of symbiotic nutrient exchange may be more diverse than originally<br />

expected (Saito 2000).<br />

AM fungi occur in soil and in association with roots as communities of<br />

organisms that may simultaneously interact with the roots of one or several<br />

co-existing <strong>plant</strong> species. Species of AM fungi differ in their mode of colonisation<br />

and their capacity to form hyphae in soil and within the root (Abbott et<br />

al. 1992).<br />

Although hyphal characteristics may be distinctive for some fungi (Dodd<br />

et al. 2000), they are not usually present as discrete organisms and are difficult<br />

to distinguish from one another within and on the <strong>surface</strong> of roots. Although<br />

the fungi may have markedly different characteristics, they appear to function<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


332<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

in a similar manner, but with different levels of efficiency depending on their<br />

abundance as well as their intrinsic characteristics. Furthermore, their symbiotic<br />

response depends on environmental conditions and the relative abundance<br />

of other AM fungi associated with the roots of the same <strong>plant</strong>.<br />

The purpose of this review is to discuss the functional diversity of AM<br />

fungi and its significance in the context of interactions at root <strong>surface</strong>s and<br />

the potential consequences of this for <strong>plant</strong> growth and <strong>plant</strong> community<br />

structure.<br />

2 Mycorrhiza Formation and Ecological Specificity<br />

Arbuscular mycorrhizal fungi live symbiotically with the roots of approximately<br />

60 % of terrestrial <strong>plant</strong>s (Brundrett and Abbott 2002). About 200<br />

species of AM fungi have been described so far in the Glomales, but unequivocal<br />

evidence of their capacity to form mycorrhizas is not available for many<br />

species (Walker and Trappe 1993). A putative zygosporic stage has only been<br />

reported for the life cycle of Gigaspora decipiens (Tommerup and Sivasithamparam<br />

1990). Knowledge of genetic diversity of AM fungi is poorly defined.<br />

Associations between hosts and symbionts are usually non-specific in AM<br />

symbiosis (Mosse 1975). Most of the evidence for non-specificity in these<br />

associations has been demonstrated by inoculating roots with propagules of<br />

species of AM fungi in separate pot cultures (Smith and Read 1997). However,<br />

it may be possible for a <strong>plant</strong> grown in field soil to be preferentially colonised<br />

by one of the species of the AM fungi present. This could result from differences<br />

in the infectivity and/or quantity of propagules of each species, or from<br />

differences in the susceptibility of roots to colonisation by each fungus. This<br />

phenomenon has been defined as ‘ecological specificity’ by McGonigle and<br />

Fitter (1990).<br />

Arbuscular mycorrhizal fungi are obligate symbionts and they depend on<br />

the formation of mycorrhizas to take up carbon from the root for completing<br />

their life cycles. Fungal growth does not continue much in axenic culture in<br />

the absence of the host <strong>plant</strong>. The obligate status of AM fungi, the coenocytic<br />

nature of their spores (Becard and Pfeffer 1993) and the lack of demonstration<br />

of recombination (Rosendahl and Taylor 1997) limit the opportunities<br />

for fundamental research on their interactions with <strong>plant</strong> roots. Molecular<br />

techniques based on DNA analysis provide a number of possibilities to<br />

develop specific probes for AM fungi for determining phylogenetic relationships<br />

and diversity and for their identification in soil and <strong>plant</strong> roots (Jacquot<br />

et al. 2000).


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 333<br />

2.1 Establishment of the Symbiosis<br />

The establishment of a functional symbiosis between AM fungi and host<br />

<strong>plant</strong>s involves a sequence of recognition events between the fungus and the<br />

<strong>plant</strong> (Giovanetti et al. 1993a; Giovannetti and Sbrana 1998). Mycorrhizal<br />

colonisation has several phases (Tester et al. 1987; Gianinazzi-Pearson and<br />

Gianinazzi 1989) including spore germination, hyphal growth in soil, hyphal<br />

attachment to roots, appressorium formation, intraradical penetration and<br />

intraradical growth involving the formation of arbuscules and coils (Smith<br />

and Read 1997; Wegel et al. 1998). The developmental stages of fungal interaction<br />

with the <strong>plant</strong> are associated with <strong>plant</strong> signals inducing gene expression<br />

and recognition between the two partners of the symbiosis (Giovanetti and<br />

Sbrana 1998).<br />

2.2 Spore Germination and Hyphal Growth<br />

Spores of AM fungi can germinate under appropriate storage and environmental<br />

conditions. A host signal is not necessary for this step in the life cycle.<br />

The first response of a fungus to a host root is stimulation of hyphal growth.<br />

It is well documented that host roots can promote hyphal growth of AM fungi<br />

and induce changes in hyphal growth pattern and morphology by stimulating<br />

branching and inducing the formation of hyphal fans (Giovannetti et al.<br />

1993b). Hyphal contact with the <strong>surface</strong> of the host root occurs at random and<br />

the increased branching of a fungus near the root <strong>surface</strong> would increase the<br />

probability of root interception. Root architecture and root density also influence<br />

the likelihood of root and hypha interception (Abbott and Robson 1984).<br />

2.3 Role of Plant Root Exudates<br />

Although hyphal growth can be increased in response to root exudates from<br />

host <strong>plant</strong>s (Mosse 1962, 1988; Koske and Gemma 1992), there is no direct evidence<br />

for the release of inhibitory compounds from non-host roots. Indirect<br />

evidence has raised this as a possibility (Ocampo et al. 1980; Holliday 1989).<br />

The exudates from non-hosts appeared to lack factors which induced hyphal<br />

growth (Giovannetti and Sbrana 1998; Nagahashi 2000).<br />

Hyphal elongation of G. fasciculatus was enhanced by exudates from Trifolium<br />

repens when the <strong>plant</strong>s were grown under phosphate-deficient conditions<br />

(Elias and Safir 1987). This effect was reduced when phosphorus was<br />

added. Root exudates from both non-mycorrhizal and mycorrhizal peas<br />

inhibited hyphal growth of Gigaspora margarita (Balaji et al. 1994). In contrast,<br />

mycorrhizal Pisum sativum and its non-mycorrhizal isogenic mutant<br />

did not form root exudates that had different effects on Glomus mossae (Gio-


334<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

vanetti et al. 1993a). Generally, the amount, type and form of root exudates<br />

have the potential to influence fungal growth before the fungus meets the root<br />

as well as after the hyphae contact the root. In addition, the formation of<br />

appressoria and hyphal penetration of the root <strong>surface</strong> may involve recognition<br />

processes (Koske and Gemma 1992).<br />

3 Functioning of Arbuscular Mycorrhizas in Nutrient<br />

Exchange<br />

Nutrient exchange in arbuscular mycorrhizas can occur between the hyphae<br />

and host root cells. External hyphae penetrate the root <strong>surface</strong> and proceed<br />

into the root cortex forming arbuscules and coils (Saito 2000). The arbuscule<br />

is a complex intracellular hyphal structure formed in Arum-type mycorrhizas<br />

(Smith and Smith 1997). It has been assumed that the arbuscule is a likely site<br />

for symbiotic nutrient transfer in Arum-type mycorrhizas (Bonfante-Fasolo<br />

1987; Smith and Smith 1997), but carbon exchange may also occur across<br />

walls of non-arbuscular hyphae (Smith and Read 1997). Variation in arbuscule<br />

development in Arum-type mycorrhizas could reflect different characteristics<br />

of roots and fungi (Smith and Dickson 1991).<br />

Arbuscular mycorrhizal fungi with a range in demand for phosphorus have<br />

been isolated from south-western Australia. In particular, an isolate of S.<br />

calospora either increased or decreased <strong>plant</strong> growth depending on the phosphate<br />

status of the soil (Thomson et al. 1986). This fungus has a high demand<br />

for carbon from the <strong>plant</strong> relative to P transfer (Pearson et al. 1994), and forms<br />

considerably more external hyphae than some other fungi that are highly<br />

effective at enhancing P uptake across a range of P supply (Abbott and Robson<br />

1985). Indeed, under some circumstances, it can be inefficient in P transfer<br />

(Smith et al. 2000). In addition, S. calospora can interact with Glomus invermaium<br />

during colonisation of roots, restricting growth of G. invermaium in<br />

other parts of the same root system during some stages of its colonisation<br />

(Pearson et al. 1993). However, colonisation and activity of S. calospora can be<br />

stopped once sporulation has taken place (Pearson and Schweiger 1994),<br />

resulting in resumption of colonisation (and presumably P uptake and transfer)<br />

by G. invermaium. This example demonstrates the dynamics of activities<br />

of two AM fungi on root <strong>surface</strong>s, but the extent to which this occurs generally<br />

for all isolates of these or other species is not known. Unfortunately, relatively<br />

few isolates have been investigated for most species of AM fungi in any environment<br />

so the extent to which generalisations can be made, even for species,<br />

is unknown (Morton and Bentivenga 1994).<br />

In field soils, as several species of AM fungi would generally be involved in<br />

co-colonisation of roots, P uptake and transfer into the <strong>plant</strong> might be<br />

affected if one fungus interfered with colonisation by another. The outcome<br />

would depend on the functional diversity of the species present, i.e. their effi-


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 335<br />

ciency in P uptake and transfer. Circumstantial evidence of seasonal variation<br />

in the function of different species of AM fungi present in a woodland environment<br />

has been clearly demonstrated (Merryweather and Fitter 1998b).<br />

There have been few direct measurements of P uptake by communities of AM<br />

fungi (Jakobsen et al. 2001), but it is difficult to attribute P transferred to the<br />

activity of a particular fungus within a community.<br />

The relative abundance of AM fungi in field-grown <strong>plant</strong>s can be manipulated<br />

by environmental and management processes that occur in the field,<br />

where dual AM fungal occupancy of roots is the norm (Merryweather and Fitter<br />

1998a). With manipulation of P supply, the important observations of<br />

Thomson et al. (1986) can be used to identify the P status of the <strong>plant</strong> at which<br />

there is a point of transition from growth enhancement to growth depression<br />

in <strong>plant</strong>s colonised by S. calospora. In contrast, G. invermaium did not display<br />

such a physiological transition in this study.<br />

3.1 Metabolic Activity During Mycorrhiza Formation<br />

Changes in metabolic activities during mycorrhiza formation provide evidence<br />

for hypotheses related to biochemical mechanisms of C and P exchange<br />

between symbionts (Saito 2000). These have mostly been examined by comparing<br />

mycorrhizal and non-mycorrhizal roots, but without identifying the<br />

mechanisms of nutrient exchange. Changes in quantity and concentration of<br />

soluble carbohydrates in roots have not shown consistent trends with mycorrhizal<br />

colonisation (Pakovsky 1989; McArthur and Knowels 1993; Pearson et<br />

al. 1994; Solaiman and Saito 1997).<br />

The localisation of alkaline phosphatase (ALPase) in arbuscular hyphae<br />

was observed by histochemical study (Saito 1995). ALPase has also been<br />

located in vacuoles in intraradical hyphae of AM fungi (Gianinazzi et al.<br />

1979), and its activity varied with environmental conditions (Jabaji-Hare et al.<br />

1990). A correlation between the number of ALPase-active arbuscules and P<br />

uptake by mycorrhizal <strong>plant</strong>s indicates that arbuscular ALPase plays a significant<br />

role in phosphorus transformation from the AM fungus to the host <strong>plant</strong><br />

(Tijssen et al. 1983). Histochemical observation of intraradical hyphae has<br />

located lipid and polyphosphate (polyP) granules in hyphae. Although polyP<br />

molecules are likely to play an important role in P translocation, their existence<br />

has been contradicted. Recently, a successive extraction method showed<br />

that a granular fraction of polyP was contained in hyphae of Gigaspora margarita<br />

(Solaiman et al. 1999). The contribution of polyP was calculated from<br />

these data and it has been concluded that the contribution is not significant<br />

(Smith et al. 2001).


336<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

3.2 Gene Expression During Mycorrhiza Formation<br />

A phosphate (Pi) transporter GvPT was isolated from the AM fungus Glomus<br />

versiforme that resembles the yeast high-affinity Pi transporter PHO84 and<br />

the <strong>plant</strong> Pht1 transporter (Harrison and van Buuren 1995). This transporter<br />

gene is thought to be active in P uptake by fungal hyphae outside roots. The<br />

molecular mechanisms at the fungus–root interface involved in Pi efflux from<br />

the fungus into the apoplastic space and subsequently, into the cortical cells of<br />

the root are not well understood. It would be interesting to know whether<br />

these transporters comprise new protein families and to identify their site of<br />

expression. Observations so far are not conclusive. For example, Liu et al.<br />

(1998) concluded that the expression patterns of MtPT1 and MtPT2 cloned<br />

from Medicago truncatula roots are not consistent with their involvement at<br />

the symbiotic interface. Similarly, expression of the tomato Pi transporter<br />

gene LePT1 has been observed in cortical cells having arbuscules and it was<br />

thought to be involved in the uptake of Pi by the <strong>plant</strong> from the fungus (Rosewarne<br />

et al. 1999). However, LePT1 transcript levels were less in mycorrhizal<br />

compared to non-mycorrhizal <strong>plant</strong>s.A Pi transporter (MtPT1) from M. truncatula<br />

roots was consistent with its role in P transport at the root/soil interface<br />

(Chiou et al. 2001).<br />

The expression of a <strong>plant</strong> H + -ATPase gene was increased in barley roots<br />

colonised by G. intraradices (Murphy et al. 1997). This demonstrated the presence<br />

of a high H + -ATPase activity in the periarbuscular membrane of mycorrhizal<br />

roots. The isolated hexose transporter was of host origin, and in situ<br />

hybridisation showed it was expressed in cortical cells in the area colonised by<br />

the AM fungus (Harrison 1996). Therefore, the efflux of sugar from the host<br />

cell to the apoplast may be mediated by the transporter. Gene expression has<br />

been demonstrated for P transporters (Harrison and van Buuren 1995, Rosewarne<br />

et al. 1999), but the genes involved in carbon flows have not been identified.<br />

A model for the pathways and regulation of P uptake in mycorrhizal<br />

<strong>plant</strong>s was proposed by Rosewarne et al. (1999) based upon expression of P<br />

transporters in uncolonised roots (Liu et al. 1998), mycorrhizal roots (Rosewarne<br />

et al. 1999) and mycorrhizal fungi (Harrison and van Buuren 1995).<br />

According to the proposed model, cloning of the arbuscule-specific Pi transporter<br />

genes is required to investigate the mechanisms of P transfer in arbuscules<br />

in comparison with other parts of the symbiosis (such as intraradical<br />

and external hyphae), but phosphate can alter expression of the Pi transporter<br />

gene (Maldonado-Mendoza et al. 2001).<br />

3.3 Nutrient Exchange Mechanisms in Arbuscular Mycorrhizas<br />

Isolation of the fungus from host tissue in mycorrhizas has assisted in identifying<br />

the location of biochemical activities in nutrient exchange (Saito 2000).


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 337<br />

Intraradical hyphae were first isolated from host roots by enzymic digestion<br />

with cellulase and pectinase and hand-sorted under a dissecting microscope<br />

(Capaccio and Callow 1982). This is a time-consuming process which might<br />

reduce the metabolic activity of the hyphae (McGee and Smith 1990). Subsequently,<br />

a more suitable method was developed for isolating metabolically<br />

active intraradical hyphae from onion roots colonised by an AM fungus with<br />

only 1 h of enzymic digestion (Saito 1995). The metabolic activity of the isolated<br />

hyphae was not affected by this technique (Saito 1995; Solaiman and<br />

Saito 1997). The isolated arbuscules remained functional in membrane transport<br />

for at least 4 h. This corresponded with the in vivo NMR study by<br />

Shachar-Hill et al. (1995). Using this isolation technique, polyphosphate<br />

metabolism in isolated intraradical hyphae (Solaiman et al. 1999) and P efflux<br />

from the isolated intraradical hyphae have been observed (Solaiman and<br />

Saito 2001). P efflux from intraradical hyphae was coupled with polyP hydrolysis.<br />

A hyperarbuscule-forming <strong>plant</strong> mutant was screened from a large collection<br />

of nodulation mutants in model legume Lotus japonicus (Senoo et al.<br />

2000; Solaiman et al. 2000). The characteristic features of this mutant are: (1)<br />

it produces a higher number of arbuscules per unit length of roots compared<br />

to the wild-type <strong>plant</strong>, (2) the individual arbuscules formed in this mutant are<br />

well developed, and (3) metabolic activity of these arbuscules is higher than<br />

of those formed in the wild-type <strong>plant</strong>. Furthermore, a method of intact<br />

arbuscule isolation from the hyperarbuscule-forming mutant was developed<br />

without using enzymic digestion of colonised roots. The isolated arbuscules<br />

were metabolically active when tested with succinate dehydrogenase (SDH;<br />

Solaiman et al. 2000), alkaline phosphatase (ALP) and acid phosphatase<br />

(ACP) histochemical staining. This mutant is suitable for molecular genetic<br />

study and for further investigation of the exchange of P and C between the<br />

symbionts.<br />

Phosphorus in soil solution is absorbed by external hyphae through the Pi<br />

transporter, and the absorbed phosphate is condensed into polyP and translocated<br />

into the intraradical hyphae by protoplasmic streaming (Cooper and<br />

Tinker 1981; Harrison and van Buuren 1995). The factors potentially regulating<br />

the uptake, transport and transfer of phosphate from the fungus have<br />

been summarised by Saito (2000) as: (1) expression and regulation of the Pi<br />

transporter, (2) protoplasmic streaming of motile vacuoles, (3) synthesis and<br />

decomposition of polyP, and (4) release of Pi across the fungal membrane in<br />

the arbuscule. There has been no other information on the fungal Pi transporter<br />

in arbuscular mycorrhizas since the reports of Harrison and van<br />

Buuren (1995) and Maldonado-Mendoza et al. (2001). Synthesis and degradation<br />

of polyP in extraradical and intraradical hyphae (Solaiman et al. 1999;<br />

Ezawa et al. 2001) and an efflux of Pi from the intraradical hyphae have been<br />

demonstrated (Solaiman and Saito 2001). In spite of these advances, current<br />

knowledge is still based upon a limited number of host – fungus combina-


338<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

tions. Demonstrations of morphological and genetic diversity of AM fungi<br />

imply that the mechanism of symbiotic nutrient exchange might be diverse<br />

(Smith and Smith 1996), but the taxonomic diversity found might not necessarily<br />

be linked to the functional role of the AM fungi. Both inter- and intraspecific<br />

variation in the effectiveness of AM fungi has been reported (Franke-<br />

Snyder et al. 2001). Studies are required that show whether fungal diversity<br />

reflects quantitative rather than qualitative differences in functioning. The<br />

current understanding of functional diversity is that <strong>plant</strong>s can respond differently<br />

to different AM fungi, not only at the level of colonisation, nutrient<br />

uptake, growth, but also at the level of gene expression (Burleigh et al. 2002).<br />

4 Functional Diversity of Arbuscular Mycorrhizal Fungi in<br />

Root and Hyphal Interactions<br />

Functional diversity of AM fungi associated with roots of <strong>plant</strong>s in different<br />

ecosystems is not well understood. The dynamics of interactions between<br />

roots and hyphae provide a framework for predicting how diversity of AM<br />

fungi might be related to mycorrhiza function, but it is difficult to measure. It<br />

is almost impossible to predict the functional diversity of AM fungi at an<br />

ecosystem level based simply on what fungi are present in soil. On a theoretical<br />

basis, the functional diversity of AM fungi under field conditions cannot<br />

be assumed to be directly correlated with a measure of diversity of AM fungi,<br />

even if the fungi present differ in P uptake and transfer under controlled conditions.<br />

This is because of differences in processes such as rate of root colonisation,<br />

interactions between fungi during colonisation and other strategies<br />

that include shutting down of hyphal infectivity in association with sporulation,<br />

as can occur for both S. calospora and A. laevis (Jasper et al. 1993; Pearson<br />

and Schweiger 1993).<br />

There is increasing interest in the potential role of AM fungi in influencing<br />

<strong>plant</strong> community structure (Read 1990; van der Heijden et al. 1998a, b;<br />

Klironomos 2002; Franke-Snyder et al. 2001). However, as yet there is little evidence<br />

to support the hypothesis that the diversity of AM fungi is an important<br />

factor influencing <strong>plant</strong> community structure under natural field conditions.<br />

This would require extensive quantification of AM fungi within roots for<br />

time-intervals that are of significance to <strong>plant</strong> and fungal growth cycles following<br />

an appropriate approach (Merryweather and Fitter 1998a, b). On the<br />

contrary, there is considerable potential for the <strong>plant</strong> communities to influence<br />

the fungal community structure through preferential effects on colonisation<br />

by particular fungi and influences on sporulation (Sanders and Fitter<br />

1992).


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 339<br />

4.1 Diversity of Arbuscular Mycorrhizal Fungi Inside Roots<br />

The benefit to <strong>plant</strong>s through enhanced P uptake is expected to be markedly<br />

altered according to the effectiveness of dominant AM fungi inside roots.<br />

However, species diversity of AM fungi has generally been examined from the<br />

perspective of presence or absence of fungi in soil, not in roots (van der Heijden<br />

et al. 1998a; Bever et al. 2001). There is little evidence of a simple relationship<br />

between the relative abundance of morphotypes of AM fungi inside roots<br />

and spores in soil (Scheltema et al. 1987; Merryweather and Fitter 1998a). The<br />

effectiveness of AM fungi in taking up P in these studies is generally not considered,<br />

but neither is it easy to determine, particularly as effectiveness can<br />

change for the same fungus depending on the P status of the soil and <strong>plant</strong><br />

(Thomson et al. 1986). The diversity of AM fungi needs to be investigated further<br />

in relation to their relative abundance inside the roots (Abbott and Gazey<br />

1994). In addition, the methods applied to selecting combinations of species<br />

of AM fungi in studies of the role of species diversity need to be considered<br />

carefully (Wardle 1999). Another consideration is that AM fungi form associations<br />

with <strong>plant</strong>s that differ markedly in their growth habits and susceptibility<br />

to colonisation, and this makes assessment of the impact of diversity of<br />

AM fungi even more difficult to predict or measure at an ecosystem level.<br />

The diversity of AM fungi could be relevant to communities of AM fungi in<br />

the same way that high <strong>plant</strong> species diversity may help stabilise <strong>plant</strong> community<br />

structure and ecosystem processes (Klironomos et al. 2000; Tilman<br />

1996). However, high diversity can also lead to competitive exclusion and<br />

cause a reduction in the number of co-existing species (Huston 1994). For AM<br />

fungi, this may only reduce the abundance of some species below levels of<br />

detection (Bever et al. 2001). The competitive ability of species of AM fungi in<br />

roots is likely to be an important factor in determining the dominance of AM<br />

fungi inside roots as well as in soil. This is compounded by seasonal changes<br />

in infectivity of AM fungi (Merryweather and Fitter 1998b), which is influenced<br />

by fungal life cycles (Abbott and Gazey 1994) such as sporulation and<br />

associated changes in infectivity of the hyphae (Pearson and Schweiger 1993).<br />

As AM fungi occur as communities in soil and in roots, the extent to which<br />

they are likely to collectively contribute to P uptake (Jakobsen et al. 2001;<br />

Solaiman and Abbott 2003) depends on the mycorrhiza dependency of the<br />

host <strong>plant</strong> (van der Heijden et al. 1998b). Although there have been intensive<br />

studies of single AM fungus function, there have been few investigations of<br />

the contributions of communities of AM fungi. Reconstructed communities<br />

of AM fungi in soil can promote <strong>plant</strong> growth (Daft 1983; Daft and Hogart<br />

1983), or have no effect on <strong>plant</strong> growth (Sylvia et al. 1993). A correlation<br />

between the occurrence of AM fungal morphotypes and seasonal P uptake for<br />

AM fungi in a natural ecosystem was observed (Merryweather and Fitter<br />

1998b), which may be related to differences among fungi in the functional<br />

characteristics of hyphae they form in soil (Smith et al. 2000).


340<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

4.2 Relationship Between Hyphae in the Root and in the Soil<br />

The quantity of hyphae in the vicinity of roots associated with mycorrhizal<br />

roots can vary greatly (Sylvia 1986) and change with time (Bethlenfalvay et al.<br />

1982). Hyphae of AM fungi play key roles in the formation and functioning of<br />

mycorrhizas (Abbott et al. 1992). Hyphae in soil, originating from either an<br />

established hyphal network or from other propagules (spores, vesicles and<br />

root fragments), lead to the recognition and subsequent colonisation of roots.<br />

The distribution of hyphae and associated sporulation will determine where<br />

propagules are located in relation to newly formed roots. The roles of the<br />

hyphae in both P uptake and soil stabilisation are dependent on their distribution<br />

within the soil matrix and their interaction with the root <strong>surface</strong>.<br />

Abbott and Robson (1984) hypothesised for G. invermaium that low initial<br />

levels of infective hyphae in the soil would lead to small amounts of hyphae in<br />

soil relative to the amount formed inside the root. For high initial densities of<br />

infective hyphae of this fungus in soil, the exponential phase of colonisation<br />

of roots was expected to occur in parallel with extensive development of<br />

hyphae in the soil. In contrast, there was no similar relationship between the<br />

formation of hyphae in soil and within the root expected for S. calospora<br />

which consistently produced large amounts of hyphae in soil, irrespective of<br />

the density of hyphae within the root.<br />

There is relatively little information about the longevity of hyphae in soil<br />

(Sylvia 1988; Hamel et al. 1990). This would be important for predicting the<br />

activities of hyphae for both colonisation and P uptake. The majority of studies<br />

of mycorrhizas measure the extent of colonisation of roots at one point in<br />

time. This measure is of little value for understanding functional diversity of<br />

AM fungi because the AM fungi within the root may either be highly active or<br />

have ceased activity for some time. Most routine techniques for assessing<br />

mycorrhizas do not assess any functional attribute of the fungus. Therefore,<br />

care is required in extrapolating from levels of mycorrhizal root colonised to<br />

functional diversity of communities of AM fungi present in soil.<br />

5 Role of Arbuscular Mycorrhizal Fungi Associated with<br />

Roots in Soil Aggregation<br />

The AM fungal hyphae can enhance soil aggregation by using more than one<br />

mechanism. In clayey soils, entanglement of soil particles by hyphae can<br />

occur (Tisdall and Oades 1982; Oades 1984). There is insufficient hyphal<br />

length to extend around particles of sand and a more likely mechanism is<br />

cross-linking of particles by hyphae in sandy soils (Degens 1997). AM fungal<br />

hyphae may bind soil aggregates by exuding a glycoprotein (Wright et al.<br />

1996; Wright and Upadhyaya 1998). Some information on species differences<br />

in soil aggregation is available which indicates that AM fungi commonly pro-


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 341<br />

duce glomalin, but the amount varies considerably for different species<br />

(Wright and Upadhaya 1998). If AM fungal communities differ in their sensitivity<br />

to disturbance, the capacity of the species present to form hyphae as<br />

well as their ability to produce glomalin should influence the degree of soil<br />

aggregation.<br />

6 Environmental Influence on Functional Diversity of<br />

Arbuscular Mycorrhizal Fungi<br />

The soil environment includes many physical and chemical properties that<br />

are continuously being modified by dynamic biological processes. Rhizosphere<br />

soil is influenced by <strong>plant</strong> root exudates and microbial activity and the<br />

AM fungi that live in this habitat have adapted to a wide range of environmental<br />

conditions (Stahl and Christensen 1991; Giovannetti and Gianinazzi-<br />

Pearson 1994). Environmental stresses on AM fungi could include: (1) high or<br />

low levels of nutrients, (2) waterlog and drought, (3) soil acidity, (4) salinity,<br />

(5) high levels of toxic metals, (6) biotic factors (e.g. fauna that feed on<br />

hyphae), and (7) absence of suitable host <strong>plant</strong>s for long periods. AM fungi<br />

can adapt to both low and high levels of soil nutrients (Solaiman and Hirata<br />

1997). As they are aerobic, waterlogging has a considerable impact on their<br />

diversity and on their functioning. One of the most important soil factors<br />

influencing the distribution of species of AM fungi is soil pH (Robson and<br />

Abbott 1989). AM fungi show considerable diversity in their response to soil<br />

pH and changes in soil pH can affect the relative abundance of species inside<br />

roots (Sano et al. 2002). This has potential to influence the structure of communities<br />

of AM fungi in soil. There is also evidence that agricultural practices<br />

such as pesticide applications, cropping sequences and soil disturbance can<br />

affect diversity of AM fungi in soil (Dodd and Jeffries 1989; Sieverding 1991;<br />

Johnson et al. 1997).<br />

7 Role of Plant Mutants in Studying the Interactions<br />

Between Arbuscular Mycorrhizal Fungi and Roots<br />

In symbiotic associations between AM fungi and <strong>plant</strong> roots, genetic control<br />

imposed by each symbiont is poorly understood. Understanding of the<br />

genetic and molecular basis of this symbiosis has been prevented by the<br />

obligate nature of the fungal symbiont and by the lack of mycorrhiza formation<br />

on the model <strong>plant</strong> Arabidopsis thaliana. Recently, Medicago truncatula<br />

and Lotus japonicus have been chosen as model <strong>plant</strong>s for research of <strong>plant</strong> –<br />

microbe symbioses (Sagan et al. 1995; Jiang and Gresshoff 1997; Bonfante et<br />

al. 2000; Senoo et al. 2000). The use of molecular genetic approaches in model<br />

legumes will rapidly increase knowledge of host genetic determinants of


342<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

arbuscular mycorrhizas. An essential step in this process has been the generation,<br />

screening and analysis of mycorrhizal mutants (Marsh and Schultze<br />

2001).<br />

Plant mutants are valuable tools in unravelling complex events that occur<br />

during cell and tissue differentiation in <strong>plant</strong>s that show impaired formation<br />

of arbuscular mycorrhizas (Peterson and Guinel 2000). Since the first description<br />

of myc – mutants (Duc et al. 1989), there has been increasing interest in<br />

using them to address questions related to various key steps in the colonisation<br />

process (Senoo et al. 2000; Wyss et al. 1990). As these fungi are obligate<br />

symbionts, it has been difficult to study the interaction between the symbionts<br />

during colonisation. The interaction in early colonisation phases of<br />

Allium porrum L. (leek) roots by the AM fungus Glomus versiforme have been<br />

described (Garriock et al. 1989) and reviewed (Giovanetti et al. 1994). Mutants<br />

of pea (Pisum sativum L.) and faba bean (Vicia faba L.) were not colonised by<br />

AM fungi (Duc et al. 1989). These mycorrhizal (myc – ) mutants were also<br />

unable to form functional root nodules (nod – ). The myc – mutants have<br />

aborted infections (Gianinazzi-Pearson et al. 1991). In contrast, nod – mutants<br />

of soybean were colonised by Glomus mossae to the same extent as wild-type<br />

(nod + ) soybean <strong>plant</strong>s (Wess et al. 1990). The myc – mutants should be<br />

screened against different AM fungi in a range of soils to see whether resistance<br />

is horizontal or if some fungi can overcome resistance as in the case of<br />

certain nod – <strong>plant</strong>s in the presence of different Rhizobium populations (Lie<br />

and Timmermans 1983). Recently, it has been shown that some AM fungi can<br />

colonise the mutant of tomato, rmc, demonstrating that the fungi can overcome<br />

resistance to successful colonisation (Gao et al. 2001). This new tool<br />

would help exploration of genetic variability in AM fungi. It would also open<br />

the possibility of controlling <strong>plant</strong> – fungus specificity in the presence of communities<br />

of AM fungi in field soils.<br />

Mycorrhizal mutants were screened from the model <strong>plant</strong> Lotus japonicus<br />

(Senoo et al. 2000) after inoculation with Glomus sp. R-10. These mutants were<br />

characterized and categorized into mcbep (mycorrhizal colonisation blocked<br />

at epidermis) and mcbex (mycorrhizal colonisation blocked at exodermis)<br />

based on the detailed assessment of colonisation and microscopic observation<br />

(Senoo et al. 2000). Isolation and cloning of the gene will facilitate understanding<br />

of its function, and it could be used to probe a range of hosts to<br />

determine its distribution and expression. It is essential to expand the collection<br />

of mutants in order to build up a comprehensive description of the molecular<br />

genetic basis of successful mycorrhization.


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 343<br />

8 Conclusion and Future Research Needs<br />

Research is needed that integrates knowledge of ecological and genetic characteristics<br />

of arbuscular mycorrhizas for predicting P-related functions of<br />

AM fungi in soil communities. For example, the diversity among Arum-type<br />

AM fungi in arbuscule formation in roots in relation to the intensity of mycorrhizal<br />

colonisation and function is not known for fungi that differ in colonisation<br />

aggressiveness and response to P supply. Neither are the relationships<br />

understood between arbuscule formation and P/C exchange for AM fungi<br />

that differ in arbuscular, intraradical and external hyphal growth characteristics.<br />

It would be interesting to compare Pi transporter gene expression in<br />

arbuscules, intraradical and external hyphae in mycorrhizas formed by AM<br />

fungi that differ in (1) arbuscular and other colonisation characteristics, and<br />

(2) functional response to phosphate supply and the presence of co-colonising<br />

AM fungi that have different C demands. Finally, clarification is required<br />

of the actual roles of AM fungi in natural environments. The importance of<br />

AM fungal diversity is currently of considerable interest, but the limited evidence<br />

available is not yet sufficient to support claims that diversity of these<br />

potentially symbiotic organisms is of wide-scale significance in regulating the<br />

structure of <strong>plant</strong> communities.<br />

References and Selected Reading<br />

Abbott LK, Robson AD (1984) The effect of root density, inoculum placement and infectivity<br />

of inoculum on the development of vesicular-arbuscular mycorrhizas. New<br />

Phytol 97:285–299<br />

Abbott LK, Robson AD (1985) Formation of external hyphae in soil by four species of<br />

vesicular-arbuscular mycorrhizal fungi. New Phytol 99:245–255<br />

Abbott LK, Gazey C (1994) An ecological view of the formation of VA mycorrhizas. Plant<br />

Soil 159:69–78<br />

Abbott LK, Robson AD, Jasper DA, Gazey C (1992) What is the role of VA mycorrhizal<br />

hyphae in soil? In: Read DJ, Lewis DH, Fitter AH, Alexander IJ (eds) Mycorrhizas in<br />

ecosystems. CAB International, Wallingford, pp 37–41<br />

Balaji B, Ba AM, LaRue TA, Tepfer D, Piche Y (1994) Pisum sativum mutants insensitive<br />

to nodulation are also insensitive to invasion in vitro by the mycorrhizal fungus<br />

Gigaspora margarita. Plant Sci 102:195–203<br />

Becard G, Pfeffer PE (1993) State of nuclear division in arbuscular mycorrhizal fungi<br />

during in vitro development. Protoplasma 194:62–68<br />

Bethlenfalvay GJ, Brown MS, Pakovsky RS (1982) Relationships between host and endophyte<br />

development in mycorrhizal soybeans. New Phytol 90:537–543<br />

Bever JD, Schultz PA, Pringle A, Morton JB (2001) Arbuscular mycorrhizal fungi: more<br />

diverse than meets the eye, and the ecological tale of why. BioSci 51:923–931<br />

Bonfante-Fasolo P (1987) Vesicular arbuscular mycorrhizae: fungus-<strong>plant</strong> interactions<br />

at the cellular level. Symbiosis 3:249–268


344<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

Bonfante P, Genre A, Faccio A, Martin I, Schauser L, Stougaard J,Web J, Parniske M (2000)<br />

The Lotus japonicus Ljsym4 gene is required for the successful symbiotic infection of<br />

root epidermis cells. Mol Plant-Microbe Interact 13:1109–1120<br />

Brundrett MC, Abbott LK (2002) Arbuscular mycorrhizas in <strong>plant</strong> communities. In: Sivasithamparam<br />

K, Dixon KW, Barrett RL (eds) Microorganisms in Plant Conservation<br />

and Biodiversity. Kluwer, Dordrecht, pp 151–193<br />

Burleigh SH, CavagnaroT, Jakobsen I (2002) Functional diversity of arbuscular mycorrhizas<br />

extends to the expression of <strong>plant</strong> genes involved in P nutrition. J Exp Bot<br />

53:1593–1601<br />

Capaccio LCM, Callow JA (1982) The enzymes of polyphosphate metabolism in vesicular-arbuscular<br />

mycorrhizas. New Phytol 91:81–91<br />

Chiou TJ, Liu H, Harrison MJ (2001) The spatial expression patterns of a phosphate<br />

transporter (MtPT1) from Medicago truncatula indicate a role in phosphate transport<br />

at the root/soil interface. Plant J 25:281–293<br />

Cooper KM, Tinker PB (1981) Translocation and transfer of nutrients in vesicular-arbuscular<br />

mycorrhizas. IV. Effect of environmental variables on movement of phosphorus.<br />

New Phytol 88:327–339<br />

Daft MJ (1983) The influence of mixed inocula on endomycorrhizal development. Plant<br />

Soil 71:331–337<br />

Daft MJ, Hogart BG (1983) Competitive interactions amongst four species of Glomus on<br />

maize and onion. Trans Br Mycol Soc 80:339–345<br />

Degens BP (1997) Macroaggregation of soils by biological bonding and binding mechanisms<br />

and factors affecting this: a review. Aust J Soil Res 35:431–446<br />

Dodd JC, Jeffries P (1989) Effects of herbicides on three vesicular-arbuscular mycorrhizal<br />

fungi associated with winter wheat (Triticum aestivum L.). Biol Fertil Soils<br />

7:113–119<br />

Dodd JC, Boddington CL, Rodriguez A, Gonzalez-Chavez C, Mansur I (2000) Mycelium<br />

of arbuscular mycorrhizal fungi (AMF) from different genera: form, function and<br />

detection. Plant Soil 226:131–151<br />

Duc G, Trouvelot A, Gianinazzi-Pearson V, Gianinazzi S (1989) First report of non-mycorrhizal<br />

<strong>plant</strong> mutants (myc – ) obtained in pea (Pisum sativum L.) and faba bean<br />

(Vicia faba L.). Plant Sci 60:215–222<br />

Elias KS, Safir GR (1987) Hyphal elongation of Glomus fasciculatus in response to root<br />

exudates. Appl Environ Microbiol 53:1928–1933<br />

Ezawa T, Smith SE, Smith FA (2001) Differentiation of polyphosphate metabolism<br />

between the extra- and intraradical hyphae of arbuscular mycorrhizal fungi. New<br />

Phytol 149:555–563<br />

Franke-Snyder M, Douds Jr DD, Galvez L, Phillips JG, Wagoner P, Drinkwater L, Morton<br />

JB (2001) Diversity of communities of arbuscular mycorrhizal (AM) fungi present in<br />

conventional versus low-input agricultural sites in eastern Pennsylvania, USA. Appl<br />

Soil Ecol 16:35–48<br />

Gao L-L, Delp G, Smith SE (2001) Colonization patterns in a mycorrhiza-defective<br />

mutant tomato vary with different arbuscular-mycorrhizal fungi. New Phytol 151:<br />

477–491<br />

Garriock ML, Peterson RL, Ackerley CA (1989) Early stages in colonization of Allium<br />

porrum (leek) roots by the vesicular-arbuscular mycorrhizal fungus, Glomus versiforme.<br />

New Phytol 112:85–92<br />

Gianinazzi S, Gianinazzi-Pearson V, Dexheimer J (1979) Enzyme studies on the metabolism<br />

of vesicular-arbuscular mycorrhiza. 3. Ultrastructural localization of acid and<br />

alkaline phosphatase in onion roots infected by Glomus mosseae (Nicol. and Gerd.).<br />

New Phytol 82:127–132<br />

Gianinazzi-Pearson V, Gianinzzi S (1989) Cellular and genetical aspects of interactions<br />

between host and fungal symbionts in mycorrhizae. Genome 31:336–341


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 345<br />

Gianinazzi-Pearson V, Gianinazzi S, Guillemin JP, Trouvelot A, Duc G (1991) Genetic and<br />

cellular analysis of resistance of vesicular-arbuscular mycorrhizal fungi in pea<br />

mutants. In: Hennecke, H, Varma DPS (eds) Advances in molecular genetics of <strong>plant</strong><br />

– microbe interactions. Kluwer, Dordrecht, pp 336–342<br />

Giovannetti M, Gianinazzi-Pearson V (1994) Biodiversity in arbuscular mycorrhizal<br />

fungi. Mycol Res 98:705–715<br />

Giovannetti M, Sbrana S, Logi C (1994) Early processes involved in host recognition by<br />

arbuscular mycorrhizal fungi. New Phytol 127:703–709<br />

Giovannetti M, Sbrana C (1998) Meeting a non-host: the behaviour of AM fungi. Mycorrhiza<br />

8:123–130<br />

Giovannetti M, Sbrana C, Avio L, Citernesi AS, Logi C (1993a) Differential hyphal morphogenesis<br />

in arbuscular mycorrhizal fungi during preinfection stages. New Phytol<br />

125:587–593<br />

Giovanetti M, Avio L, Sbrana L, Citernesi AS (1993b) Factors affecting appressorium<br />

development in the vesicular-arbuscular mycorrhizal fungus Glomus mosseae (Nicol.<br />

and Gerd.) Gerd. and Trape. New Phytol 123:114–122<br />

Graham JH, Eissenstat DM (1998) Field evidence for the carbon cost of citrus mycorrhizas.<br />

New Phytol 140:103–110<br />

Graham JH, Abbott LK (2000) Wheat responses to aggressive and non-aggressive arbuscular<br />

mycorrhizal fungi. Plant Soil 220:207–218<br />

Hamel C, Fyles H, Smith DL (1990) measurement of development of endomycorrhizal<br />

mycelium using three different vital stains. New Phytol 115:297–302<br />

Harrison MJ (1996) A sugar transporter from Medicago truncatula: altered expression<br />

pattern in roots during vesicular-arbuscular (VA) mycorrhizal associations. Plant J<br />

9:491–503<br />

Harrison MJ (1999) Molecular and cellular aspects of the arbuscular mycorrhizal symbiosis.<br />

Ann Rev Plant Physiol Plant Mol Biol 50:361–389<br />

Harrison MJ, van Buuren ML (1995) A phosphate transporter from the mycorrhizal fungus<br />

Glomus versiforme. Nature 378:626–629<br />

Holliday P (1989) A dictionary of <strong>plant</strong> pathology. Cambridge University Press, Cambridge,<br />

pp 369<br />

Huston MA (1994) Biological diversity: the coexistence of species on changing landscapes.<br />

Cambridge University Press, Cambridge, pp 1–681<br />

Jabaji-Hare SH, Therien J, Charest PM (1990) High resolution cytochemical study of the<br />

vesicular-arbuscular mycorrhizal association, Glomus clarum-Allium porrum. New<br />

Phytol 114:481–496<br />

Jakobsen I (1998) Transport of phosphorus and carbon in arbuscular mycorrhizas. In:<br />

Varma A, Hock B (eds) Mycorrhiza: structure, function, molecular biology and<br />

biotechnology. Springer, Berlin Heidelberg New York, pp 305–332<br />

Jakobsen I, Gazey C, Abbott LK (2001) Phosphate transport by communities of arbuscular<br />

mycorrhizal fungi in intact soil cores. New Phytol 149:95–103<br />

Jacquot E, van Tuinen D, Gianinazzi S, Gianinazzi-Pearson V (2000) Monitoring species<br />

of arbuscular mycorrhizal fungi in <strong>plant</strong>a and in soil by nested PCR: application to<br />

the study of the impact of sewage sludge. Plant Soil 226:179–188<br />

Jasper DA, Abbott LK, Robson AD (1993) The survival of infective hyphae of vesiculararbuscular<br />

mycorrhizal fungi in dry soil: an interaction with sporulation. New Phytol<br />

124:473–479<br />

Jiang Q, Gresshoff PM (1997) Classical and molecular genetics of the model legume<br />

Lotus japonicus. Mol Plant-Microbe Interact 10:59–68<br />

Johnson NC, Graham JH, Smith FA (1997) Functioning of mycorrhizal associations along<br />

the mutualism-parasitism continuum. New Phytol 135:575–585<br />

Klironomos JN (2002) Feedback with soil biota contributes to <strong>plant</strong> rarity and invasiveness<br />

in communities. Nature 417:67–70


346<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

Klironomos JN, McCune J, Hart M, Neville J (2000) The influence of arbuscular mycorrhizae<br />

on the relationship between <strong>plant</strong> diversity and productivity. Ecol Lett<br />

3:137–141<br />

Koide R, Elliott G (1989) Cost, benefit and efficiency of the vesicular-arbuscular mycorrhizal<br />

symbiosis. Funct Ecol 3:252–255<br />

Koide RT, Schreiner RP (1992) Regulation of the vesicular-arbuscular mycorrhizal symbiosis.<br />

Ann Rev Plant Physiol Plant Mol Biol 43:557–581<br />

Koske RE, Gemma JN (1992) Fungal reactions to <strong>plant</strong>s prior to mycorrhiza formation.<br />

In: Allen MF (ed) Mycorrhizal functioning. Routledge, Chapman and Hall, New York,<br />

pp. 3–36<br />

Lie TA, Timmermans PCJM (1983) Host-genetic control of nitrogen fixation in the<br />

legume-Rhizobium symbiosis: complication in the genetic analysis due to material<br />

effects. Plant Soil 75:449–453<br />

Liu H, Trieu AT, Blaylock LA, Harrison MJ (1998) Cloning and characterization of two<br />

phosphate transporters from Medicago truncatula roots: regulation in response to<br />

phosphate and to colonisation by arbuscular mycorrhizal (AM) fungi. Mol Plant-<br />

Microbe Interact 11:14–22<br />

Maldonado-Mendoza IE, Dewbre GR, Harrison MJ (2001) A phosphate transporter gene<br />

from the extra-radical mycelium of an arbuscular mycorrhizal fungus Glomus<br />

intraradices is regulated in respect to phosphate in the environment. Mol Plant-<br />

Microbe Interact 4:1140–1148<br />

Marsh JF, Schultze M (2001) Analysis of arbuscular mycorrhizas using symbiosis-defective<br />

<strong>plant</strong> mutants. New Phytol 150:525–532<br />

McArthur DAJ, Knowels NR (1993) Influence of vesicular-arbuscular mycorrhizal fungi<br />

on the response of potato to phosphorus deficiency. Plant Physiol 101:147–160<br />

McGee PA, Smith SE (1990) Activity of succinate dehydrogenase in vesicular-arbuscular<br />

mycorrhizal fungi after enzymic digestion from roots of Allium porrum. Mycol Res<br />

94:305–308<br />

McGonigle T, Fitter AH (1990) Ecological specificity of vesicular-arbuscular mycorrhizal<br />

associations. Mycol Res 94:120–122<br />

Merryweather JW, Fitter AH (1998a) The arbuscular mycorrhizal fungi of Hyacinthoides<br />

non-scripta-I. Diversity of fungal taxa. New Phytol 138:117–129<br />

Merryweather JW, Fitter AH (1998b) The arbuscular mycorrhizal fungi of Hyacinthoides<br />

non-scripta-II. Seasonal and spatial patterns of fungal populations. New Phytol<br />

138:131–142<br />

Morton JB, Bentivenga SP (1994) Levels of diversity in endomycorrhizal fungi (Glomales,<br />

Zygomycetes) and their role in defining taxonomic and non-taxonomic groups.<br />

Plant Soil 159:47–59<br />

Mosse B (1962) The establishment of vesicular-arbuscular mycorrhiza under aseptic<br />

conditions. J Gener Microbiol 27:509–520<br />

Mosse B (1975) Specificity in VA mycorrhizas. In: Sanders FE, Mosse B, Tinker PB (eds)<br />

Endomycorrhizas. Academic Press, London, pp 469–484<br />

Mosse B (1988) Some studies relating to “independent growth of vesicular-arbuscular<br />

endophytes”. Can J Bot 66:2533–2540<br />

Murphy PJ, Langridge P, Smith SE (1997) Cloning <strong>plant</strong> genes differentially expressed<br />

during colonisation of roots of Hordeum vulgare by the vesicular-arbuscular mycorrhizal<br />

fungus Glomus intraradices. New Phytol 135:291–301<br />

Nagahashi G (2000) In vitro and in situ techniques to examine the role of roots and root<br />

exudates during AM fungus-host interactions. In: Kapulnik Y, Douds Jr DD (eds)<br />

Arbuscular mycorrhizas: physiology and function. Kluwer, Dordrecht, pp 287–300<br />

Oades JM (1984) Soil organic matter and structural stability: Mechanisms and implications<br />

for management. Plant Soil 76:319–337


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 347<br />

Ocampo JA, Martin J, Hayman DS (1980) Influence of <strong>plant</strong> interactions on vesiculararbuscular<br />

mycorrhizal infections. I. Host and non-host <strong>plant</strong>s grown together. New<br />

Phytol 84:27–35<br />

Pakovsky RS (1989) Carbohydrate, protein amino acid status of Glycine-Glomus-<br />

Bradyrhizobium symbiosis. Physiol Plant 72:733–746<br />

Pearson JN, Schweiger P (1993) Scutellospora calospora (Nicol. and Gerd.) Walker and<br />

Sanders associated with subterranean clover: dynamics of colonisation, sporulation<br />

and soluble carbohydrates. New Phytol 124:215–219<br />

Pearson JN, Schweiger P (1994) Scuttelospora calospora (Nicol. and Gerd.) Walker and<br />

Sanders associated with subterranean clover produces non-infective hyphae during<br />

sporulation. New Phytol 127:697–701<br />

Pearson JN, Abbott LK, Jasper DA (1993) Mediation of competition between two colonizing<br />

VA mycorrhizal fungi by the host <strong>plant</strong>. New Phytol 123:93–98<br />

Pearson JN,Abbott LK, Jasper DA (1994) Phosphate, soluble carbohydrates and the competition<br />

between two arbuscular mycorrhizal fungi colonizing subterranean clover.<br />

New Phytol 127:101–106<br />

Peterson RL, Guinel FC (2000) The use of <strong>plant</strong> mutants to study regulation of colonization<br />

by AM fungi. In: Kapulnik Y, Douds Jr DD (eds) Arbuscular mycorrhizas: physiology<br />

and function. Kluwer, Dordrecht, pp 147–171<br />

Rausch C, Daram P, Brunner S, Jansa J, Laloi M, Leggewie G,Amrhein N, Bucher M (2001)<br />

A phosphate transporter expressed in arbuscule-containing cells in potato. Nature<br />

414:462–466<br />

Read DJ (1990) Mycorrhizas in ecosystems – Nature’s response to the ‘Law of the minimum’.<br />

In: Hawksworth DL (ed) Frontiers in mycology. CAB International, Wallingford,<br />

pp 101–130<br />

Robson AD, Abbott LK (1989) The effect of soil acidity on microbial activity in soil. In:<br />

Robson AD (ed) Soil acidity and <strong>plant</strong> growth. Academic Press, Sydney, pp 139–165<br />

Rosendahl S, Taylor JW (1997) Development of multiple genetic markers for studies of<br />

genetic variation in arbuscular mycorrhizal fungi using AFLP. Mol Ecol 6:821–829<br />

Rosewarne GM, Barker SJ, Smith SE, Smith FA, Schachtman DP (1999) A Lycopersicon<br />

esculentum phosphate transporter (LePT1) involved in phosphorus uptake from a<br />

vesicular-arbuscular mycorrhizal fungus. New Phytol 144:507–516<br />

Sagan M, Morandi D, Tarenghi E, Duc G (1995) Selection of nodulation and mycorrhizal<br />

mutants in the model <strong>plant</strong> Medicago truncatula (Gaertn.) after g-ray mutagenesis.<br />

Plant Sci 111:63–71<br />

Saito M (1995) Enzyme activities of the internal hyphae and germinated spores of an<br />

arbuscular mycorrhizal fungus, Gigaspora margarita Becker and Hall. New Phytol<br />

129:425–431<br />

Saito M (2000) Symbiotic exchange of nutrients in arbuscular mycorrhizas: Transport<br />

and transfer of phosphorus. In: Kapulnik Y, Douds Jr DD (eds) Arbuscular mycorrhizas:<br />

physiology and function. Kluwer, Dordrecht, pp 85–105<br />

Sanders IR, Fitters AH (1992) Evidence for differential responses between host-fungus<br />

combinations of vesicular-arbuscular mycorrhizas from a grassland. Mycol Res<br />

96:415–419<br />

Sano SM, Abbott LK, Solaiman MZ, Robson AD (2002) Influence of liming, inoculum<br />

level and inoculum placement on root colonization of subterranean clover. Mycorrhiza<br />

12:285–290<br />

Scheltema MA, Abbott LK, Robson AD (1987) Seasonal variation in infectivity of VA<br />

mycorrhizal fungi in annual pastures in a Mediterranean environment. Aust J Agric<br />

Res 38:707–715<br />

Schwab SM, Menge JA, Tinker PB (1991) Regulation of nutrient transfer between host<br />

and fungus in vesicular mycorrhizas. New Phytol 117:387–398


348<br />

M. Zakaria Solaiman and Lynette K. Abbott<br />

Senoo K, Solaiman MZ, Kawaguchi M, Imaizumi-Anraku H, Akao S, Tanaka A, Obata H<br />

(2000) Isolation of two different phenotypes of mycorrhizal mutants in the model<br />

legume <strong>plant</strong> Lotus japonicus after EMS-treatment. Plant Cell Physiol 41:726–732<br />

Shachar-Hill Y, Pfeffer PE, Douds D, Osman SF, Doner LW, Ratcliffe RG (1995) Partitioning<br />

of intermediary carbon metabolism in vesicular-arbuscular mycorrhizal leek.<br />

Plant Physiol 108:7–15<br />

Sieverding E (1991) Vesicular-arbuscular mycorrhiza management in tropical agroecosystems.<br />

Deutsche Gesellschaft für Technische Zusammenarbeit. Eschborn, Germany,<br />

pp 371<br />

Smilauer P (2001) Communities of arbuscular mycorrhizal fungi in grassland: seasonal<br />

variability and effects of environment and host <strong>plant</strong>s. Folia Geobot 36:243–263<br />

Smith FA, Smith SE (1997) Structural diversity in (vesicular)-arbuscular mycorrhizal<br />

symbioses. New Phytol 137:373–388<br />

Smith FA, Jakobsen I, Smith SE (2000) Spatial differences in acquisition of soil phosphate<br />

between two arbuscular mycorrhizal fungi in symbiosis with Medicago truncatula.<br />

New Phytol 147:357–366<br />

Smith SE, Dickson S (1991) Quantification of active vesicular-arbuscular mycorrhizal<br />

infection using image analysis and other techniques. Aust J Plant Physiol 18:737–648<br />

Smith SE, Smith FA (1996) Mutualism and parasitism: diversity in function and structure<br />

in the “arbuscular” (VA) mycorrhizal symbiosis. Adv Bot Res 22:1–43<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis. Academic Press, London, pp 1–605<br />

Smith SE, Dickson S, Smith FA (2001) Nutrient transfer in arbuscular mycorrhizas: how<br />

are fungal and <strong>plant</strong> processes integrated? Aust J Plant Physiol 28:683–694<br />

Solaiman MZ, Hirata H (1997) Effect of arbuscular mycorrhizal fungi inoculation of rice<br />

seedlings at the nursery stage upon performance in the paddy field and greenhouse.<br />

Plant Soil 191:1–12<br />

Solaiman MZ, Saito M (1997) Use of sugars by intraradical hyphae of arbuscular mycorrhizal<br />

fungi revealed by radiorespirometry. New Phytol 136:533–538<br />

Solaiman MZ, Saito M (2001) Phosphate efflux from the intraradical hyphae of an arbuscular<br />

mycorrhizal fungus, Gigaspora margarita, in vitro and its implication to phosphorus<br />

translocation in the hyphae. New Phytol 151:525–533<br />

Solaiman MZ,Abbott LK (2003) Phosphorus uptake by a community of arbuscular mycorrhizal<br />

fungi in jarrah forest. Plant Soil 248:313–320<br />

Solaiman MZ, Ezawa T, Kojima T, Saito M (1999) Polyphosphates in intraradical and<br />

extraradical hyphae of arbuscular mycorrhizal fungi. Appl Environ Microbiol<br />

65:5604–5606<br />

Solaiman MZ, Senoo K, Kawaguchi M, Imaizumi-Anraku H, Akao S, Tanaka A, Obata H<br />

(2000) Characterization of mycorrhizas formed by Glomus sp. on roots of hypernodulating<br />

mutants of Lotus japonicus. J Plant Res 113:443–448<br />

Sylvia DM (1986) Spatial and temporal distribution of vesicular-arbuscular mycorrhizal<br />

fungi associated with Uniola paniculata in Florida foredunes. Mycologia 78:728–734<br />

Sylvia DM (1988) Activity of external hyphae vesicular arbuscular mycorrhizal fungi.<br />

Soil Biol Biochem 20:39–43<br />

Stahl PD, Christensen M (1991) Population variation in the mycorrhizal fungus Glomus<br />

mosseae: breath of environmental tolerance. Mycol Res 95:300–3007<br />

Sylvia DM, Wilson DO, Graham JH, Maddox JJ, Millner P, Morton JB, Skipper HD, Wright<br />

SF, Jarstfer AG (1993) Evaluation of vesicular arbuscular mycorrhizal fungi in diverse<br />

<strong>plant</strong>s and soils. Soil Biol Biochem 25:705–713<br />

Tester M, Smith SE, Smith FA (1987) The phenomenon of non-mycorrhizal <strong>plant</strong>s. Can J<br />

Bot 65:419–431<br />

Thomson BD, Robson AD, Abbott LK (1986) Effects of phosphorus on the formation of<br />

mycorrhizas by Gigaspora margarita and Glomus fasciculatum in relation to root carbohydrates.<br />

New Phytol 103:751–765


19 Functional Diversity of Arbuscular Mycorrhizal Fungi on Root Surfaces 349<br />

Tijssen JPF, Dubbelman TMAR, Van Steveninak J (1983) Isolation and characterization<br />

of polyphosphate from the yeast cell <strong>surface</strong>. Biochem Biophysics Acta 760:143–148<br />

Tilman D (1996) Biodiversity: population versus ecosystem stability. Ecology 77:350–363<br />

Tisdall JM, Oades JM (1982) Organic matter and water-stable aggregates in soils. J Soil<br />

Sci 33:141–163<br />

Tommerup IC, Sivasithamparam K (1990) Zygospores and asexual spores of Gigaspora<br />

decipiens, an arbuscular mycorrhizal fungus. Mycol Res 94:897–900<br />

Van der Heijden MGA, Klironomos JN, Ursic M, Moutoglis P, Streitwolf-Engel, Boller T,<br />

Wiemken A, Sanders IR (1998a) Mycorrhizal fungal diversity determines <strong>plant</strong> biodiversity,<br />

ecosystem variability and productivity. Nature 396:69–72<br />

Van der Heijden MGA, Boller T, Wiemken A, Sanders IR (1998b) Different arbuscular<br />

mycorrhizal fungal species are potential determinants of <strong>plant</strong> community structure.<br />

Ecology 79:2082–2091<br />

Walker C, Trappe JM (1993) Names and epithets in the glomales and endogonales. Mycol<br />

Res 97:339–344<br />

Wardle DA (1999) Is “sampling effect” a problem for experiments investigating biodiversity<br />

– ecosystem function relationships? Oikos 87:403–407<br />

Wegel E, Schauser L, Sandal N, Stougaard J, Parniske M (1998) Mycorrhiza mutants of<br />

Lotus japonicus define genetically independent steps during symbiotic infection. Mol<br />

Plant-Microbe Interact 11:933–936<br />

Wright SF, Upadhyaya A (1998) A survey of soils for aggregate stability and glomalin, a<br />

glycoprotein produced by hyphae of arbuscular mycorrhizal fungi. Plant Soil 198:97–<br />

107<br />

Wright SF, Franke-Snyder M, Morton JB, Upadhyaya A (1996) Time-course study and<br />

partial characterization of a protein on hyphae of arbuscular mycorrhizal fungi during<br />

active colonization of roots. Plant Soil 181:193–203<br />

Wyss P, Mellor RB, Wiemken A (1990) Vesicular-arbuscular mycorrhizas of wild-type<br />

soybean and non-nodulating mutants with Glomus mossae contain symbiosis-specific<br />

polypeptides (mycorrhizins), immunologically cross-reactive with nodulins.<br />

Planta 182:22–26


20 Mycorrhizal Fungi and Plant Growth Promoting<br />

Rhizobacteria<br />

José-Miguel Barea, Rosario Azcón<br />

and Concepción Azcón-Aguilar<br />

1 Introduction<br />

Soil microbial communities are crucial in maintaining a biological balance in<br />

soil, a key issue for the sustainability of either natural ecosystems or agroecosystems<br />

(Kennedy and Smith 1995). When provided with available carbon<br />

substrates, soil microorganisms are able to develop a range of activities in the<br />

microhabitats where they flourish and some of these activities are of great relevance<br />

for <strong>plant</strong> growth and health and for soil quality (Bowen and Rovira<br />

1999). Soil-borne microbes are found bound to the <strong>surface</strong> of soil particles or<br />

in the soil aggregates, while others interact specifically with <strong>plant</strong> roots (Glick<br />

1995). Particularly important from the point of view of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong><br />

are the interactions at the root – soil interface where microorganisms,<br />

<strong>plant</strong> roots and soil constituents interact (Lynch 1990; Azcón-Aguilar and<br />

Barea 1992; Linderman 1992; Kennedy 1998; Bowen and Rovira 1999; Barea<br />

2000; Gryndler 2000) to develop a dynamic environment what is known as the<br />

rhizosphere (Hiltner 1904). The rhizosphere, therefore, is the zone of influence<br />

of <strong>plant</strong> roots on the soil microbiota; a microcosm with physical, chemical<br />

and biological properties different from those of the root-free bulk soil<br />

(Bowen and Rovira 1999; Gryndler 2000; Barea 2000). A characteristic of the<br />

rhizosphere is that microbial diversity is altered and that the activity and<br />

number of microorganisms is increased (Kennedy 1998).<br />

The supply of photosynthates and decaying <strong>plant</strong> material to the root-associated<br />

microbiota is a key issue for rhizosphere formation and functioning.<br />

The release of organic material is known to occur mainly as root exudates,<br />

acting as either signals or growth substrates (Werner 1998). However, once<br />

the microbial population is established, rhizosphere developments are<br />

affected by microbially induced changes on rooting patterns and by the supply<br />

of available nutrients to <strong>plant</strong>s, which in turn modify the quality and<br />

quantity of root exudates (Bowen and Rovira 1999; Barea 2000; Gryndler<br />

2000). Microbial interactions in the rhizosphere are known to markedly influence<br />

<strong>plant</strong> fitness and soil quality (Lynch 1990; Bethlenfalvay and Schüepp<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


352<br />

José-Miguel Barea et al.<br />

1994). In particular, microorganisms associated with <strong>plant</strong> roots help the host<br />

<strong>plant</strong> adapt to stress conditions concerning water and mineral nutrition, and<br />

soil-borne <strong>plant</strong> pathogens (Jeffries and Barea 2001).<br />

From the point of view of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>, the compartmentalization<br />

of the rhizosphere (broad sense) is important. Kennedy (1998) suggested<br />

that there are three separate, but interacting components, namely the<br />

rhizosphere (soil), the rhizoplane, and the root itself. The rhizosphere is the<br />

zone of soil influenced by roots through the release of substrates that affect<br />

microbial activity. The rhizoplane is actually the root <strong>surface</strong>, but also<br />

includes the strongly adhering soil particles. The root itself is a part of the system<br />

because certain microorganisms, the endophytes, are able to colonize<br />

root tissues. Microbial colonization of the rhizoplane and/or the root tissues<br />

is known as root colonization, while the colonization of the adjacent volume<br />

of soil under the influence of the root is known as rhizosphere colonization<br />

(Kloepper et al. 1991).<br />

2 Main Types of Rhizosphere Microorganisms<br />

In spite of several very different types of microorganisms living in the root –<br />

soil interface microhabitats, most studies on rhizosphere <strong>microbiology</strong> refer<br />

to only bacteria and fungi (Bowen and Rovira 1999; Gryndler 2000). Two main<br />

groups of microorganisms can be distinguished: saprophytes and symbionts.<br />

Both of them comprise detrimental, neutral and beneficial bacteria and fungi.<br />

Detrimental microbes include the major <strong>plant</strong> pathogens, as well as minor<br />

parasitic and nonparasitic, deleterious rhizosphere organisms, either bacteria<br />

or fungi, (Weller and Thomashow 1994; Nehl et al. 1996). Beneficial microorganisms<br />

are known to play fundamental roles in agroecosystem and natural<br />

ecosystem sustainability, and some of them can be used as inoculants to benefit<br />

<strong>plant</strong> growth and health (Alabouvette et al. 1997; Barea et al. 1997; Cordier<br />

et al. 1999; Barea 2000; Dobbelaere et al. 2001; Probanza et al. 2002).<br />

Saprophytic bacteria and fungi colonize subterranean <strong>plant</strong> <strong>surface</strong>s. Root<br />

colonization by rhizosphere bacteria has been extensively studied. It appears<br />

to be a strain-specific, active process that is exhibited by a subset of the total<br />

rhizosphere bacterial community, termed rhizobacteria, which is known to<br />

display a specific ability for root colonization (Kloepper 1994, 1996). The beneficial<br />

root colonizing rhizosphere bacteria, the so-called <strong>plant</strong> growth promoting<br />

rhizobacteria (PGPR), carry out important activities in the root/soil<br />

interfaces (Probanza et al. 2002).<br />

The endophytic microorganisms colonizing the root tissues develop activities<br />

involved in <strong>plant</strong> growth promotion and <strong>plant</strong> protection (Kloepper<br />

1994; Chanway 1996; Sturz et al. 2000; Sturz and Novak 2000). Even nonsymbiotic<br />

microorganisms may be endophytes and colonize the root tissues (Duijff<br />

et al. 1997; Van Loon et al. 1998). Piriformospora indica (Basidiomycota) has


een described as a <strong>plant</strong>-growth-promoting root endophyte (Varma et al.<br />

1999). However, because this chapter deals only with rhizobacteria, these, and<br />

other fungal endophytes, will not be dealt with further.<br />

Plant symbiotic bacteria and fungi are recognized and can either include<br />

pathogens or mutualistic organisms. Mycorrhizal fungi and nitrogen (N 2)-fixing<br />

bacteria are the main mutualistic symbionts (Barea 1997). This chapter<br />

will focus only on mycorrhizal fungi and PGPRs.<br />

3 Mycorrhizal Fungi<br />

20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 353<br />

The roots of most <strong>plant</strong> species associate with certain soil fungi and establish<br />

what are known as mycorrhiza (Smith and Read 1997). Mycorrhizal functions<br />

include improvement of <strong>plant</strong> establishment, enhancement of nutrient<br />

uptake, protection against cultural and environmental stresses, and the<br />

improvement of soil structure (Barea et al. 1997).<br />

Mycorrhizal symbiosis can be found in nearly all types of ecological situations,<br />

and most <strong>plant</strong> species are able to form this symbiosis naturally, the<br />

most common type involved in the normal cropping systems is the arbuscular<br />

mycorrhizal (AM) type (Smith and Read 1997). The responsible AM fungi<br />

belong to the order Glomales in the Zygomycetes (Morton and Redecker<br />

2001), and are a very common group of soil-borne fungi whose origin and<br />

divergence date back 100–400 million years ago (Simon et al. 1993; Morton<br />

2000; Redecker et al. 2000). However, a new fungal phylum, the Glomeromycota<br />

have recently been proposed (Schübler et al. 2001). Because this chapter<br />

focuses only on arbuscular mycorrhizas, the term “AM fungi” will be used to<br />

refer to “arbuscular mycorrhizal fungi”.<br />

During the process of AM formation (Giovannetti 2000), in which the <strong>plant</strong><br />

“accepts” the fungal colonization without any significant rejection reaction<br />

(Dumas-Gaudot et al. 2000), a series of root–fungus interactions allows the<br />

integration of both organisms. The establishment of the symbiosis is the<br />

result of a continuous molecular “dialogue” between <strong>plant</strong> and fungus, as<br />

exerted through the exchange of both recognition and acceptance signals<br />

(Vierheilig and Piché 2002). The result of this dialogue will finally depend on<br />

the genome expression of both partners (Gianinazzi-Pearson et al. 1996;<br />

Franken and Requena 2001).<br />

After the biotrophic colonization of the root cortex, AM fungi develop an<br />

external mycelium which is a bridge connecting the root with the surrounding<br />

soil microhabitats. Such mycorrhizal (fungal-root) symbiosis is critical in<br />

nutrient cycling in soil–<strong>plant</strong> systems (Smith and Read 1997). In cooperation<br />

with other soil organisms, the external AM fungal mycelium forms water-stable<br />

aggregates necessary for good soil tilth (Miller and Jastrow 2000; Requena<br />

et al. 2001). The AM symbiosis also improves <strong>plant</strong> health through increased<br />

protection against biotic and abiotic stresses (Bethlenfalvay and Linderman


354<br />

José-Miguel Barea et al.<br />

1992; Azcón-Aguilar and Barea 1996; Linderman 2000; Miller and Jastrow<br />

2000; Augé 2001; Requena et al. 2001; Werner et al. 2002).<br />

Recent developments in molecular biology are being applied to the genetic<br />

characterization of AM fungi based on PCR-based approaches (Sanders et al.<br />

1996; Helgason et al. 1998; Ferrol et al. 2000). During the last few years, the<br />

analysis of ribosomal genes (rRNA) has demonstrated the polymorphism of<br />

these genes in AM fungi, particularly those corresponding to the small ribosomal<br />

subunit 18S, therefore permitting phylogeny and diversity studies<br />

(Clapp et al. 1995; Redecker et al. 1997; van Tuinen et al. 1998; Redecker et al.<br />

2000; Daniell et al. 2001; Schübler et al. 2001). Novel techniques currently<br />

developed for microbial molecular ecology studies, such as PCR-single-strand<br />

conformation polymorphism (SSCP) and PCR-temperature gradient gel electrophoresis<br />

(TGGE), are being adapted for the characterization of different<br />

ecotypes of AM fungi, both in soil and in roots (Kjoller and Rosendahl 2000).<br />

Since AM fungi are obligate symbionts, they must multiply on living roots.<br />

This is a limitation for inocula production (Azcón-Aguilar and Barea 1997).<br />

However, several substrates and procedures have been described for inoculum<br />

production and application in horticulture/fruit culture/forestry (Gianinazzi<br />

et al. 1990; Vestberg and Estaun 1994; Lobato et al. 1995; Varma and<br />

Schüepp 1995; Calvet et al. 1996; Sylvia 1998; Azcón-Aguilar et al. 2000;<br />

Mohammad et al. 2000). The Federation of European Mycorrhizal Inoculum<br />

Producers has been established.<br />

4 Plant Growth Promoting Rhizobacteria<br />

The beneficial root colonizing rhizosphere bacteria, the so-called <strong>plant</strong><br />

growth promoting rhizobacteria (PGPR), are defined by three intrinsic characteristics:<br />

(1) they must have the ability to undergo root colonization, (2)<br />

they must survive and multiply in microhabitats associated with the root <strong>surface</strong>,<br />

in competition with native microbiota, at least for the time needed to<br />

express their <strong>plant</strong> promotion activities, and (3) they must have the ability to<br />

promote <strong>plant</strong> growth (Kloepper 1994). The PGPR are known to carry out<br />

many important ecosystem processes, such as those involved in the biological<br />

control of <strong>plant</strong> pathogens, nutrient cycling and/or seedling establishment<br />

(Haas et al. 1991; Kloepper et al. 1991; Lugtenberg et al. 1991; Lemanceau and<br />

Alabouvette 1993; O’Gara et al. 1994; Weller and Thomashow 1994; Broek and<br />

Vanderleyden 1995; Glick 1995; Bashan and Holguin 1998; Barea 2000;<br />

Probanza et al. 2002). Many bacterial taxa include PGPR strains with<br />

Pseudomonas and Bacillus as the most commonly described genera possessing<br />

PGPR ability, and some strains from these and other genera are used as<br />

seed inoculants (Kloepper 1994; Bertrand et al. 2001; Probanza 2002).<br />

Azospirillum sp. are considered PGPR (Bashan 1999; Bashan and Gonzalez<br />

1999) and are used as seed inoculants under field conditions (Dobbelaere et


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 355<br />

al. 2001). The main activity of these bacteria is associated with the production<br />

of auxin-type phytohormones (Dobbelaere et al. 1999). The production and<br />

significance of auxins have been investigated at the molecular level (van de<br />

Broek et al. 1999; Lambrecht et al. 2000).<br />

Several systems for studying rhizosphere colonization by PGPR have been<br />

proposed, the one described by Simons et al. (1996) appears interesting, not<br />

only for testing the PGPR ability, but also to investigate PGPR/<strong>plant</strong> root interactions.<br />

It is known that several bacterial cell <strong>surface</strong> properties could be involved<br />

in the adhesion of PGPR to roots that may be either nonspecific based on electrostatic<br />

forces, or involve specific recognition between the <strong>surface</strong>s. In this<br />

context, root <strong>surface</strong> glyco-proteins and several different bacterial exo-polysaccharides<br />

could be involved (Weller and Thomashow 1994). In some PGPR,<br />

fimbriae (pili) may function in the adherence of cells to roots, but the contribution<br />

of flagella to the colonization process apparently depends on the PGPR<br />

strain, <strong>plant</strong> species and type of soil (Kloepper 1994).A sugar-binding protein<br />

system has been described in Azospirillum related to chemostaxis and root<br />

colonization (van Bastelaere et al. 1999).<br />

Novel techniques for microbial community fingerprinting are being developed<br />

(Kozdroj and van Elsas, 2000), and these are being adapted for the<br />

genetic characterization of PGPR. For example, the approach used by Zinniel<br />

et al. (2002), based on the 16S rRNA gene amplification and sequencing, has<br />

been proposed for the genetic characterization of endophytic bacteria, while<br />

Bertrand et al. (2001) identify PGPR 16S rDNA sequence analysis. The molecular<br />

bases of rhizosphere colonization have recently been reviewed (Lugtenberg<br />

et al. 2001). Siciliano and Germida (1998) proposed the use of BIOLOG<br />

analysis and fatty acid methyl ester profiles to study PGPR behavior after<br />

inoculation, particularly the effects on root-associated microbial populations.<br />

Confocal laser scanning microscopy is being used for studying the<br />

microorganisms/<strong>plant</strong> root interactions, for example, to detect a currently<br />

used marker such as the green fluorescent protein (Lagopodi et al. 2002) or to<br />

localize colonizing bacteria by fluorescence and in situ hybridization, after<br />

staining with the fluorescent Live/Dead BacLight dye (Bianciotto et al. 2000,<br />

2001).<br />

The molecular bases of the biocontrol ability of these rhizobacteria have<br />

been investigated in the last few years (Keel et al. 1992; O’Gara et al. 1994;<br />

Cook et al. 1995; Tomashow and Weller 1995; Chin-A-Woeng et al. 2001;<br />

Moenne-Loccos et al. 2001), and systemic induced resistance has been argued<br />

as a mechanism of disease suppression by endophytes (Duijff et al. 1998), or<br />

other PGPR (Defago and Keel 1995; Chin-A-Woeng et al. 2001).


356<br />

José-Miguel Barea et al.<br />

5 Reasons for Studying Arbuscular Mycorrhizal Fungi and<br />

Plant Growth Promoting Rhizobacteria Interactions and<br />

Main Scenarios<br />

Since they share common habitats, i.e., the root <strong>surface</strong>, and common functions,<br />

the AM fungi and PGPR have to interact during their processes of root<br />

colonization or functioning as root-associated microorganisms. In fact, just<br />

like any soil other inhabitant, the AM fungi are immersed in the framework of<br />

microbial interactions characteristic of soil microbiota relationships (Barea<br />

1997). Soil microorganisms, particularly PGPR, can influence AM formation<br />

and function and, conversely, mycorrhizas can affect the microbial populations,<br />

particularly PGPR in the rhizosphere (Azcón-Aguilar and Barea 1992;<br />

Linderman 1992, 1994; Fitter and Garbaye 1994; Barea 1997, 2000). The analysis<br />

of microbe – microbe interactions is crucial to an understanding of the<br />

events which occur at the root – soil interface and, particularly, to those<br />

related to the microbial colonization of the root <strong>surface</strong>, or the processes of<br />

root infection/colonization by pathogens or mutualistic symbionts (Lynch<br />

1990).<br />

These interactions must be taken into consideration when trying to manage<br />

AM fungi and PGPR for the biological control of <strong>plant</strong> pathogens or for<br />

the biogeochemical cycling of <strong>plant</strong> nutrients (Barea et al. 1997, 2002).<br />

Information is accumulating with regard to cell-to-cell interactions<br />

between AM fungi and PGPR. Bianciotto et al. (1996a, b, 2000, 2001) and Bonfante<br />

and Perotto (2000) investigated whether PGPR attach to the structures<br />

of the AM fungi by means of a direct cell-to-cell interaction. Attachment of<br />

rhizobia and pseudomonads to the spores and fungal mycelium of Gigaspora<br />

margarita was visualized by a combination of electron and confocal microscopy.<br />

The results showed that both rhizobia and pseudomonads adhere to<br />

spores and hyphae of AM fungi germinated in vitro, although the degree of<br />

attachment depended upon the strain. Bianciotto et al. (1996b) showed that<br />

extracellular material of bacterial origin containing cellulose, which was produced<br />

around the attached bacteria, may mediate fungal/bacterial interactions.<br />

They also support the fact that AM fungi could act as a vehicle for the<br />

colonization of <strong>plant</strong> roots by PGPR, as previously suggested (Boddey et al.<br />

1991).<br />

Bianciotto et al. (1996b) demonstrated that there were no specific receptors<br />

for the bacteria on the fungal structures and that physicochemical factors<br />

govern attachment to fungal <strong>surface</strong>s. Electrostatic interactions may, therefore,<br />

play a key role in the early stages of adhesion and cellulose fibrils may be<br />

later involved to guarantee a stable attachment. The complex interactions<br />

involving the tripartite system composed by AM fungi/bacteria/<strong>plant</strong> have<br />

recently being reviewed (Bonfante and Perotto 2000).


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 357<br />

6 Effect of Plant Growth Promoting Rhizobacteria on<br />

Mycorrhiza Formation<br />

Microbial populations in the rhizosphere are known to either interfere with or<br />

benefit AM establishment (Germida and Walley 1996; Vosátka and Gryndler<br />

1999). Deleterious rhizosphere bacteria (Nehl et al. 1996) and mycoparasitic<br />

relationships (Jeffries 1997), have been found to interfere with AM formation,<br />

while many microorganisms can improve AM formation and/or functioning<br />

(Barea 1997). One example of the beneficial effects is that of the so-called<br />

mycorrhiza-helper-bacteria which are known to stimulate mycelial growth of<br />

mycorrhizal fungi and/or enhance mycorrhizal formation (Garbaye 1994;<br />

Azcón-Aguilar and Barea 1995; Barea 1997; Frey-Klett et al. 1997; Gryndler<br />

and Hrselova 1998; Gryndler 2000; Gryndler et al. 2000). Soil microorganisms<br />

can produce compounds that increase root cell permeability and are able to<br />

increase the rates of root exudation. This, in turn, would stimulate mycorrhizal<br />

fungal mycelia in the rhizosphere or facilitate root penetration by the<br />

fungus. Plant hormones, as produced by soil microorganisms, are known to<br />

affect mycorrhiza establishment (Azcón-Aguilar and Barea 1992, 1995; Barea<br />

1997, 2000).<br />

Rhizosphere microorganisms are also known to affect the pre-symbiotic<br />

stages (Giovannetti 2000) of mycorrhizal developments, like spore germination<br />

rate and mycelial growth (Azcón-Aguilar and Barea, 1992, 1995).<br />

It is noteworthy that antibiotic-producing Pseudomonas sp. (Barea et al.<br />

1998; Vazquez et al. 2000) did not interfere with mycorrhiza formation or<br />

functioning.<br />

7 Effect of Mycorrhizas on the Establishment of Plant<br />

Growth Promoting Rhizobacteria in the Rhizosphere<br />

The establishment of the AM fungus in the root cortex is known to change<br />

many key aspects of <strong>plant</strong> physiology. These include the mineral nutrient<br />

composition in <strong>plant</strong> tissues, the hormonal balance and the patterns of C allocation<br />

(Harley and Smith 1983; Azcón-Aguilar and Bago 1994; Smith et al.<br />

1994). Therefore, the AM symbiotic status changes the chemical composition<br />

of root exudates while the development of an AM soil mycelium introduces<br />

physical modifications into the environment surrounding the roots. The AM<br />

soil mycelium represents a carbon source to microbial communities which is<br />

an important contribution through interactions with components of the<br />

microbiota to improve <strong>plant</strong> growth and health, and soil quality (Bethlenfalvay<br />

and Schüepp 1994).<br />

Arbuscular mycorrhizal-induced changes in <strong>plant</strong> physiology affect, both<br />

quantitatively and qualitatively, the microbial populations in either the rhizosphere<br />

and/or the rhizoplane (Azcón-Aguilar and Barea 1992; Linderman


358<br />

José-Miguel Barea et al.<br />

1992; Barea 1997; Cordier et al. 1999; Barea 2000; Gryndler 2000). This situation<br />

creates the “rhizosphere of a mycorrhizal <strong>plant</strong>”. However, there are specific<br />

modifications in the environment surrounding the AM mycelium itself,<br />

which develop what it known as the mycorrhizosphere (Linderman 1992;<br />

Barea 2000; Grynler 2000). In addition to this term, the soil space affected by<br />

extraradical hyphae is also called the mycosphere (Linderman 1988) or<br />

hyphosphere as an analogy to the term rhizosphere (Gryndler 2000). Large<br />

numbers of bacteria (including actinomycetes) and fungi can be associated<br />

with AM fungal structures (Filippi et al. 1998; Budi et al. 1999). Since the AM<br />

mycelium releases energy-rich organic compounds, an increased growth and<br />

activity of microbial saprophytes are expected to occur in the mycorrhizosphere.<br />

However, the enrichment of this particular environment with organic<br />

compounds is much lower than that of the rhizosphere, which corresponds to<br />

lower counts of bacteria in mycorrhizospheric soil, compared to those in the<br />

rhizosphere (Andrade et al. 1997).Apparently, there is a preferential establishment<br />

of Gram-negative bacteria in the hyphosphere (Vosátka 1996).<br />

It has been demonstrated that mycorrhizal colonization changes some<br />

morphological parameters in developing root systems (Atkinson et al. 1994;<br />

Berta et al. 1995), with a greater root branching as the most commonly<br />

described effect. Undoubtedly these changes must affect the establishment<br />

and activity of microorganisms in the mycorrhizosphere environment.<br />

The establishment of PGPR inoculants in the rhizosphere can be affected<br />

by AM fungal co-inoculation (Christensen and Jakobsen 1993; Puppi et al.<br />

1994; Barea 1997; Andrade et al. 1998; Ravnskov et al. 1999). In particular, AM<br />

inoculation improved the establishment of both inoculated and indigenous<br />

phosphate-solubilizing rhizobacteria (Toro et al. 1997; Barea et al. 2002).<br />

Interestingly, mycorrhizal fungi improved rhizosphere colonization by<br />

Pseudomonas sp. and root colonization by Azospirillum sp. (Klyuchnikov and<br />

Kozhevin 1990). Moreover, several experiments, reviewed by Nehl et al. (1996),<br />

suggest that mycorrhizal colonization may affect whether a given rhizobacterium<br />

functions as a PGPR, or as a DRB.<br />

In spite of the fact that most of the reports support the beneficial effects of<br />

mycorrhizas on the establishment of PGPR inoculants, detrimental effects<br />

have also been found (Waschkies et al. 1994; Marschner and Crowley 1996a, b;<br />

Barea et al. 1997).<br />

As indicated previously, an extreme case of close interactions is that of<br />

Burkholderia-like bacteria as endosymbionts in AM fungi of the Gigasporaceae<br />

(Bianciotto et al. 2000; Ruiz-Lozano and Bonfante 2000).


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 359<br />

8 Interactions Involved in Nutrient Cycling and Plant<br />

Growth Promotion<br />

It is well known that soil microorganisms are able to change the bioavailability<br />

of mineral <strong>plant</strong> nutrients, and this ability has been shown by soil bacteria<br />

probed to have PGPR activity and, in some cases, be used as <strong>plant</strong> inoculants<br />

(Barea et al. 1997; Probanza et al. 2002). Several experiments have described<br />

the improvement of <strong>plant</strong> growth and nutrition by means of synergistic interactions<br />

between PGPR and AM fungi (Barea 2000). It has also been suggested<br />

that a certain level of selectivity (“specificity”) is involved in these interactions<br />

(Azcón 1989). From the perspective of sustainability, the re-establishment<br />

of nutrient cycles after any process of soil degradation is of interest, as<br />

is the understanding of the microbial interactions responsible for the subsequent<br />

management of such natural resources, either for a low-input agricultural<br />

technology (Bethlenfalvay and Linderman 1992; Gianinazzi and<br />

Schüepp 1994; Jeffries and Barea 2001), or for the re-establishment of the natural<br />

vegetation in a degraded area (Miller and Jastrow 1994, 2000; Requena et<br />

al. 2001). Most of the information on this topic concerns N and P cycling<br />

(Barea 2000).<br />

In spite of this, Rhizobium sp. (general term) are not considered among the<br />

PGPR types; due to the relevance of their interaction with AM fungi, it would<br />

be interesting to make some comments on this. A great deal of work has been<br />

carried out on the tripartite symbiosis legume (general term) – mycorrhiza-<br />

Rhizobium (Azcón-Aguilar and Barea 1992; Barea et al. 1992; Barea 2000). The<br />

inoculation of AM fungi has been shown to improve nodulation and N 2 fixation.<br />

Using the isotope 15 N has made it possible to ascertain and quantify the<br />

amount of N which is actually fixed in a particular situation, and measure the<br />

contribution of the AM symbiosis to the process (Barea et al. 1992). The physiological<br />

and biochemical mechanisms underlying the AM fungi x Rhizobium<br />

interactions to improve legume productivity have also been discussed. In<br />

spite of the main AM effect in enhancing Rhizobium activity mediated by a<br />

generalized stimulation of host nutrition, more localized effects may occur at<br />

the root or nodule level (Barea et al. 1992). Interactions can also take place at<br />

either the pre-colonization stages, when both microorganisms interact as rhizosphere<br />

inhabitants, or during the development of the tripartite symbiosis<br />

(Azcón-Aguilar and Barea 1992). The influence of host and/or bacterial genotypes<br />

in these interactions has also been discussed, suggesting a certain level<br />

of specificity (Azcón et al. 1991; Ruiz-Lozano and Azcón 1993; Monzón and<br />

Azcón 1996).<br />

Multimicrobial interactions including AM fungi, Rhizobium sp. and PGPR<br />

have also been tested (Requena et al. 1997). Target microorganisms were isolated<br />

from a representative area of a desertification-threatened semi-arid<br />

ecosystem in the south-east of Spain. Microbial isolates were characterized<br />

and screened for effectiveness in soil microcosms. Anthyllis cytisoides L., an


360<br />

José-Miguel Barea et al.<br />

AM-dependent pioneer legume, dominant in the target Mediterranean<br />

ecosystem, was the test <strong>plant</strong>. Several microbial cultures from existing collections<br />

were also included in the screening process. In general, the results support<br />

the importance of physiological and genetic adaptation of microbes to<br />

the environment, thus the use of efficient local isolates is recommended. Several<br />

microbial combinations were effective in improving <strong>plant</strong> development,<br />

nutrient uptake, N 2 -fixation ( 15 N) and root system quality.<br />

The interactions between AM fungi and Rhizobium have been demonstrated<br />

to be beneficial under drought conditions (Goicoechea at al. 1997,<br />

1998; Ruiz-Lozano et al. 2001).<br />

There is also evidence that Rhizobium strains are able to colonize the rhizosphere<br />

of nonlegume hosts where they establish positive interactions with<br />

AM fungi and behave as PGPR (Schloter et al. 1997; Galleguillos et al. 2000).<br />

In spite of the fact that Azospirillum are known to benefit <strong>plant</strong> development,<br />

N acquisition and yield under appropriate conditions (Okon 1994;<br />

Bashan 1999; Dobbelaere et al. 2001), it has been demonstrated that these bacteria<br />

mainly act by influencing the morphology, geometry and physiology of<br />

the root system. Interactions between AM fungi and Azospirillum have been<br />

reviewed by Volpin and Kapulnik (1994) and it has been demonstrated that<br />

Azospirillum could enhance mycorrhizal formation and response while AM<br />

fungi may improve Azospirillum establishment in the rhizosphere.<br />

Since some PGPR may improve nodulation by Rhizobium sp. (Halverson<br />

and Handelsman 1991; Staley et al. 1992; Azcón 1993), certain PGPR-Rhizobium<br />

interactions could be relevant to mycorrhizosphere interactions.<br />

The interactions related to P-cycling have also received much attention.<br />

These are based on the fact that phosphate ions solubilized by free-living<br />

microorganisms from sparingly soluble inorganic and organic P compounds<br />

(Whitelaw 2000) increase the soil phosphate pools available for the extraradical<br />

AM mycelium to benefit <strong>plant</strong> nutrition (Smith and Read 1997). Several<br />

experiments have demonstrated synergistic microbial interactions involving<br />

phosphate-solubilizing rhizobacteria (PSB) and mycorrhizal fungi (Barea et<br />

al. 1997; Kim et al. 1998). The interactive effect of PSB and mycorrhizal fungi<br />

on <strong>plant</strong> use of soil P sources of low bioavailability was evaluated by using 32 P<br />

isotopic dilution approaches (Toro et al. 1997, 1998). The PSB behaved as mycorrhiza-helper-bacteria,<br />

promoting mycorrhiza establishment by both the<br />

indigenous and the inoculated mycorrhiza. Conversely, mycorrhiza formation<br />

increased the size of the PSB population. Because the bacteria did not change<br />

root weight, length or specific root length, they probably acted by improving<br />

the pre-colonization stages of mycorrhiza formation. The dual inoculation<br />

treatment significantly increased biomass and N and P accumulation in <strong>plant</strong><br />

tissues and these dually inoculated <strong>plant</strong>s displayed lower specific activity<br />

( 32 P/ 31 P) than their comparable controls, suggesting that the mycorrhizal and<br />

bacterized <strong>plant</strong>s were using P sources (endogenous or added as rock phosphate)<br />

otherwise unavailable to the <strong>plant</strong>. It, therefore appears that these rhi-


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 361<br />

zosphere/mycorrhizosphere interactions contributed to the biogeochemical P<br />

cycling, thereby promoting <strong>plant</strong> nutrition. The interactive effect of PSB, AM<br />

fungi and Rhizobium with regard to improving the agronomic efficiency of<br />

rock phosphate for legume crops (Medicago sativa), was evaluated by using<br />

isotopic techniques under controlled conditions, and further validated under<br />

field conditions (Barea et al. 2002). It was demonstrated that the microbial<br />

interactions tested improved <strong>plant</strong> growth and N and P acquisition under<br />

normal cultivation. Similar results were obtained by using Medicago arborea,<br />

a woody legume of interest for revegetation and biological reactivation of<br />

desertified semi-arid Mediterranean ecosystems (Valdenegro et al. 2001).<br />

9 Interactions for the Biological Control of Root Pathogens<br />

Once the AM status has been established in <strong>plant</strong> roots, reduced damage<br />

caused by soil-borne <strong>plant</strong> pathogens has been shown. To account for this,<br />

several mechanisms have been suggested to explain the enhancement of <strong>plant</strong><br />

resistance/tolerance in mycorrhizal <strong>plant</strong>s (Linderman 1994, 2000; Azcón-<br />

Aguilar and Barea 1992, 1996). One of the proposed mechanisms is based on<br />

the microbial changes produced in the mycorrhizosphere. In this context,<br />

there is strong evidence that these microbial shifts occur, and that the resulting<br />

microbial equilibria could influence the growth and health of the <strong>plant</strong>s.<br />

Although this effect has not been assessed specifically as a mechanism for<br />

AM-associated biological control, there are indications that such a mechanism<br />

could be involved (Azcón-Aguilar and Barea 1992, 1996; Linderman<br />

1994, 2000). In any case, it has been demonstrated that such an effect is dependent<br />

on the AM fungus involved, as well as the substrate and host <strong>plant</strong><br />

(Azcón-Aguilar and Barea 1996; Linderman 2000).<br />

Since specific PGPR antagonistic to root pathogens are being used as biological<br />

control agents (Alabouvette et al. 1997), it has been proposed to try to<br />

exploit the prophylactic ability of AM fungi in association with these antagonists<br />

(Linderman 1994, 2000; Azcón-Aguilar and Barea 1996; Barea et al. 1998;<br />

Budi et al. 1999). Experimental evidence is accumulating, but the information<br />

is still too scarce to make general conclusions.<br />

Several studies have demonstrated that microbial antagonists of fungal<br />

pathogens, either fungi or PGPR, do not exert any anti-microbial effect<br />

against AM fungi (Calvet et al. 1993; Barea et al. 1998; Edwards et al. 1998;<br />

Vazquez et al. 2000; Werner et al. 2002). This is a key point to exploit the possibilities<br />

of dual (AM fungi and PGPR) inoculation in <strong>plant</strong> defense against<br />

root pathogens.<br />

In particular, Barea et al. (1998) carried out a series of experiments to test<br />

the effect of Pseudomonas spp. producing 2,4-diacetylphloroglucinol (DAPG)<br />

on AM formation and functioning. Three Pseudomonas strains were tested for<br />

their effects on AM fungi: a wild type (F113) producing the antifungal com-


362<br />

José-Miguel Barea et al.<br />

pound DAPG; the genetically modified strain (F113G22), a DAPG-negative<br />

mutant of F113; and another genetically modified strain [F113 (pCU203)], a<br />

DAPG-overproducer. The results from in vitro and in soil experiments<br />

demonstrate no negative effects of these Pseudomonas strains on spore germination,<br />

and a stimulation of hyphal growth of the AM fungus Glomus<br />

mosseae. Concentrations of the antifungal compound DAPG which were far in<br />

excess of those reached in the rhizosphere of Pseudomonas-inoculated <strong>plant</strong>s<br />

exhibited negative effects on germination of AM fungal spores, but more realistic<br />

concentrations of DAPG did not affect AM fungal development. A soil<br />

microcosm system was also used to evaluate the effect of these bacteria on the<br />

process of AM formation. No significant difference in AM formation on<br />

tomato <strong>plant</strong>s between F113, F113G22 and F113 (pCU203) was observed, with<br />

the F113 and F113G22 strains resulting in a significant increase in the percentage<br />

of the root system becoming mycorrhizal. Therefore, these strains<br />

behaved as MHB. In a field experiment, none of these Pseudomonas strains<br />

affected: (1) number and diversity of AM fungal native population; (2) the<br />

percentage of root length that became mycorrhizal; (3) AM performance. Furthermore,<br />

the antifungal Pseudomonas improved <strong>plant</strong> growth and nutrient<br />

(N and P) acquisition by the mycorrhizal <strong>plant</strong>s (Barea et al. 1998).<br />

Acknowledgements. This work was supported by CICyT (REN2000–1506 project), Spain,<br />

and GENOMYCA (QLK5–2000–01319 project), ECO-SAFE (QLK3–2000–31759 project),<br />

and INCO-DEV (ICA4-CT-2001–10057) UE.<br />

References and Selected Reading<br />

Alabouvette C, Schippers B, Lemanceau P, Bakker PAHM (1997) Biological control of<br />

fusarium-wilts: towards development of commercial product. In: Boland GJ, Kuykendall<br />

LD (eds) Plant microbe interactions and biological control. Marcel Dekker,<br />

New York, pp 15–36<br />

Andrade G, Mihara KL, Linderman RG, Bethlenfalvay GJ (1997) Bacteria from rhizosphere<br />

and hyphosphere soils of different arbuscular mycorrhizal fungi. Plant Soil<br />

192:71–79<br />

Andrade G, Mihara KL, Linderman RG, Bethlenfalvay GJ (1998) Soil aggregation status<br />

and rhizobacteria in the mycorrhizosphere. Plant Soil 202:89–96<br />

Atkinson S, Berta G, Hooker JE (1994) Impact of mycorrhizal colonisation on root architecture,<br />

root longevity and the formation of growth regulators. In: Gianinazzi S,<br />

Schüepp H (eds) Impact of arbuscular mycorrhizas on sustainable agriculture and<br />

natural ecosystems. ALS, Birkhäuser, Basel, Switzerland, pp 47–60<br />

Augé RM (2001) Water relations, drought and vesicular-arbuscular mycorrhizal symbiosis.<br />

Mycorrhiza 11:3–42<br />

Azcón R (1989) Selective interaction between free-living rhizosphere bacteria and vesicular-arbuscular<br />

mycorrhizal fungi. Soil Biol Biochem 21:639–644<br />

Azcón R (1993) Growth and nutrition of nodulated mycorrhizal and non-mycorrhizal<br />

Hedysarum coronarium as a result of treatments with fractions from a <strong>plant</strong> growthpromoting<br />

rhizobacteria. Soil Biol Biochem 25:1037–1042


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 363<br />

Azcón R, Rubio R, Barea JM (1991) Selective interactions between different species of<br />

mycorrhizal fungi and Rhizobium meliloti strains, and their effects on growth, N 2 -fixation<br />

( 15 N) and nutrition of Medicago sativa L. New Phytol 117:399–404<br />

Azcón-Aguilar C, Barea JM (1992) Interactions between mycorrhizal fungi and other<br />

rhizosphere microorganisms. In: Allen MJ (eds) Mycorrhizal functioning.An integrative<br />

<strong>plant</strong>-fungal process. Routledge, Chapman and Hall, New York, pp 163–198<br />

Azcón-Aguilar C, Bago B (1994) Physiological characteristics of the host <strong>plant</strong> promoting<br />

an undisturbed functioning of the mycorrhizal symbiosis. In: Gianinazzi S,<br />

Schüepp H (eds) Impact of arbuscular mycorrhizas on sustainable agriculture and<br />

natural ecosystems. ALS, Birkhäuser, Basel, Switzerland, pp 47–60<br />

Azcón-Aguilar C, Barea JM (1995) Saprophytic growth of arbuscular-mycorrhizal fungi.<br />

In: Hock B, Varma A (eds) Mycorrhiza structure function, molecular biology and<br />

biotechnology. Springer, Berlin Heidelberg New York, pp 391–407<br />

Azcón-Aguilar C, Barea JM (1996) Arbuscular mycorrhizas and biological control of<br />

soil-borne <strong>plant</strong> pathogens. An overview of the mechanisms involved. Mycorrhiza<br />

6:457–464<br />

Azcón-Aguilar C, Barea JM (1997) Applying mycorrhiza biotechnology to horticulture:<br />

significance and potentials. Sci Hortic 68:1–24<br />

Azcón-Aguilar C, Palenzuela EJ, Barea JM (2000) Substrato para la producción de inóculos<br />

de hongos formadores de micorrizas. Patente N. 9901814, Spain, CSIC<br />

Barea JM (1997) Mycorrhiza/bacteria interactions on <strong>plant</strong> growth promotion. In:<br />

Ogoshi A, Kobayashi L, Homma Y, Kodama F, Kondon N, Akino S (eds) Plant growthpromoting<br />

rhizobacteria, present status and future prospects. OECD, Paris, pp<br />

150–158<br />

Barea JM (2000) Rhizosphere and mycorrhiza of field crops. In: Toutant JP, Balazs E,<br />

Galante E, Lynch JM, Schepers JS, Werner D, Werry PA (eds) Biological resource management:<br />

connecting science and policy (OECD). INRA, Editions and Springer, Berlin<br />

Heidelberg New York, pp 110–125<br />

Barea JM, Azcón R, Azcón-Aguilar C (1992) Vesicular-arbuscular mycorrhizal fungi in<br />

nitrogen-fixing systems. In: Norris JR, Read DJ,Varma AK (eds) Methods in <strong>microbiology</strong>.<br />

Academic Press, London, pp 391–416<br />

Barea JM, Azcón-Aguilar C, Azcón R (1997) Interactions between mycorrhizal fungi and<br />

rhizosphere microorganisms within the context of sustainable soil-<strong>plant</strong> systems. In:<br />

Gange AC, Brown VK (eds) Multitrophic interactions in terrestrial systems. Blackwell<br />

Science, Oxford, pp 65–77<br />

Barea JM, Andrade G, Bianciotto V, Dowling D, Lohrke S, Bonfante P, O’Gara F, Azcón-<br />

Aguilar C (1998) Impact on arbuscular mycorrhiza formation of Pseudomonas strains<br />

used as inoculants for the biocontrol of soil-borne <strong>plant</strong> fungal pathogens. Appl Environ<br />

Microbiol 64:2304–2307<br />

Barea JM, Toro M, Orozco MO, Campos E,Azcón R (2002) The application of isotopic ( 32 P<br />

and 15 N) dilution techniques to evaluate the interactive effect of phosphate-solubilizing<br />

rhizobacteria, mycorrhizal fungi and Rhizobium to improve the agronomic efficiency<br />

of rock phosphate for legume crops. Nutr Cycl Agroecosyst 63:35–42<br />

Bashan Y (1999) Interactions of Azospirillum spp. in soils: a review. Biol Fertil Soils<br />

29:246–256<br />

Bashan Y, Holguin G (1998) Proposal for the division of <strong>plant</strong> growth-promoting rhizobacteria<br />

into two classifications: biocontrol-PGPB (<strong>plant</strong> growth-promoting bacteria)<br />

and PGPB. Soil Biol Biochem 30:1225–1228<br />

Bashan Y, Gonzalez LE (1999) Long-term survival of the <strong>plant</strong>-growth-promoting bacteria<br />

Azospirillum brasilense and Pseudomonas fluorescens in dry alginate inoculant.<br />

Appl Microbiol Biotechnol 51:262–266


364<br />

José-Miguel Barea et al.<br />

Berta G, Trotta A, Fusconi A, Hooker JE, Munro M, Arkinson D, Giovannetti M, Morini S,<br />

Fortuna P, Tisserant B, Gianinazzi-Pearson V, Gianinazzi S (1995) Arbuscular mycorrhizal<br />

induced changes to <strong>plant</strong> growth and root system morphology in Prunus<br />

cerasifera. Tree Physiol 15:281–293<br />

Bertrand H, Nalin R, Bally R, Cleyet-Marel JC (2001) Isolation and identification of the<br />

most efficient <strong>plant</strong> growth-promoting bacteria associated with canola (Brassica<br />

napus). Biol Fertil Soils 33:152–156<br />

Bethlenfalvay GJ, Linderman RG (1992) Mycorrhizae in sustainable agriculture. ASA<br />

Special publication No. 54, Madison, Wisconsin<br />

Bethlenfalvay GJ, Schüepp H (1994) Arbuscular mycorrhizas and agrosystem stability.<br />

In: Gianinazzi S, Schüepp H (eds) Impact of arbuscular mycorrhizas on sustainable<br />

agriculture and natural ecosystems. Birkhäuser, Basel, pp 117–131<br />

Bianciotto V, Bandi C, Minerdi D, Sironi M, Tichy HV, Bonfante P (1996a) An obligately<br />

endosymbiotic mycorrhizal fungus itself harbors obligately intracellular bacteria.<br />

Appl Environ Microbiol 62:3005–3010<br />

Bianciotto V, Minerdi D, Perotto S, Bonfante P (1996b). Cellular interactions between<br />

arbuscular mycorrhizal fungi and rhizosphere bacteria. Protoplasma 193:123–131<br />

Bianciotto V, Lumini E, Lanfranco L, Minerdi D, Bonfante P, Perotto S (2000) Detection<br />

and identification of bacterial endosymbionts in arbuscular mycorrhizal fungi<br />

belonging to the family Gigasporaceae. Appl Environ Microbiol 66:4503–4509<br />

Bianciotto V, Andreotti S, Balestrini R, Bonfante P, Perotto S (2001) Mucoid mutants of<br />

the biocontrol strain Pseudomonas fluorescens CHA0 show increased ability in<br />

biofilm formation on mycorrhizal and nonmycorrhizal carrot roots. Mol Plant<br />

Microbe Interact 14:255–260<br />

Boddey RM, Urquiaga S, Reis V, Döbereiner J (1991). Biological nitrogen fixation associated<br />

with sugar cane. Plant Soil 137:111–117<br />

Bonfante P, Perotto S (2000) Outside and inside the roots: cell-to-cell interactions among<br />

arbuscular mycorrhizal fungi, bacteria and host <strong>plant</strong>s. In: Podila GK, Douds DD Jr<br />

(eds) Current advances in mycorrhizae research. APS Press, St. Paul, MN, pp 141–155<br />

Bowen GD, Rovira AD (1999) The rhizosphere and its management to improve <strong>plant</strong><br />

growth. Adv Agron 66:1–102<br />

Broek AV,Vanderleyden J (1995) Genetics of the Azospirillum-<strong>plant</strong> root association. Crit<br />

Rev Plant Sci 14:445–466<br />

Budi SW,Van Tuinen D, Martinotti G, Gianinazzi S (1999) Isolation from Sorghum bicolor<br />

mycorrhizosphere of a bacterium compatible with arbuscular mycorrhiza development<br />

and antagonistic towards soilborne fungal pathogens. Appl Environ Microbiol<br />

65:5148–5150<br />

Calvet C, Pera J, Barea JM (1993) Growth response of marigold (Tagetes erecta L.) to inoculation<br />

with Glomus mosseae, Trichoderma aureoviride and Phythium ultimum in a<br />

peat-perlite mixture. Plant Soil 148:1–6<br />

Calvet C, Camprubi A, Rodriguez-Kabana R (1996) Inclusion of arbuscular mycorrhizal<br />

fungi in alginate films for experimental studies and <strong>plant</strong> inoculation. Hortscience<br />

31:285<br />

Chanway CP (1996) Endophytes: they’re not just fungi! Can J Bot 74:321–322<br />

Chin-A-Woeng TFC, Thomas-Oates JE, Lugtenberg BJJ, Bloemberg GV (2001) Introduction<br />

of the phzH gene of Pseudomonas chlororaphis PCL 1391 extends the range of<br />

biocontrol ability of phenazine-1 carboxylic acid producing Pseudomonas. Mol Plant-<br />

Microbe Interact 14:1006–1015<br />

Christensen H, Jakobsen I (1993) Reduction of bacterial growth by a vesicular-arbuscular<br />

mycorrhizal fungus in the rhizosphere of cucumber (Cucunis sativus L). Biol Fertil<br />

Soils 15:253–258<br />

Clapp JP, Young JPW, Merryweather JW, Fitter AH (1995) Diversity of fungal symbionts<br />

in arbuscular mycorrhizas from a natural community. New Phytol 130:259–265


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 365<br />

Cook RJ, Thomashow LS, Weller DM, Fujimoto D, Mazzola M, Bangera G, Kim DS (1995)<br />

Molecular mechanisms of defense by rhizobacteria against root disease. Proc Natl<br />

Acad Sci USA 92:4197–4201<br />

Cordier C, Lemoine MC, Lemanceau P, Gianinazzi-Pearson V, Gianinazzi S (1999) The<br />

beneficial rhizosphere: a necessary strategy for micro<strong>plant</strong> production. Acta Hortic<br />

530:259–265<br />

Daniell TJ, Husband R, Fitter AH, Young JPW (2001) Molecular diversity of arbuscular<br />

mycorrhizal fungi colonising arable crops. FEMS Microbiol Ecol 36: 203–209<br />

Defago G, Keel C (1995) Pseudomonads as biocontrol agents of diseases caused by soilborne<br />

pathogens In: Hokkanen HMT, Lynch JM (eds) Benefits and risks of introducing<br />

biocontrol agents. University Press, Cambridge<br />

Dobbelaere S, Croonenborghs A, Thys A, Vande Browk A, Vanderleyden J (1999) Phytostimulatory<br />

effect Azospirillum brasilense strains and auxins on wheat. Plant Soil<br />

212:155–164<br />

Dobbelaere S, Croonenborghs A, Thys A, Ptacek D, Vanderleyden J, Dutto P, Labandera-<br />

Gonzalez C, Caballero-Mellado J, Aguirre JF, Kapulnik Y, Brener S, Burdman S,<br />

Kadouri D, Sarig S, Okon Y (2001) Response of agronomically important crops to<br />

inoculation with Azospirillum. Aust J Plant Physiol 28:1–9<br />

Duijff BJ, Gianinazzi-Pearson V, Lemanceau P (1997) Involvement of the outer membrane<br />

lipopolysaccharides in the endophytic root colonization of tomato roots by<br />

biocontrol Pseudomonas fluorescens strain WCS417r. New Phytol 135:325–334<br />

Duijff BJ, Pouhair D, Olivain C, Alabouvette C, Lemanceau P (1998) Implication of systemic<br />

induced resistance in the suppression of fusarium wilt of tomato by<br />

Pseudomonas fluorescens WCS417r and nonpathogenic Fusarium oxysporum Fo47.<br />

Eur J Plant Pathol. 104:903–910<br />

Dumas-Gaudot E, Gollotte A, Cordier C, Gianinazzi S, Gianinazzi-Pearson V (2000) Modulation<br />

of host defence systems In: Kapulnick Y, Douds Jr DD (eds) Arbuscular mycorrhizas:<br />

physiology and functions. Kluwer, Dordrecht, pp 121–140<br />

Edwards SG,Young JPW, Fitter AH (1998) Interactions between Pseudomonas fluorescens<br />

biocontrol agents and Glomus mosseae, an arbuscular mycorrhizal fungus, within the<br />

rhizosphere. FEMS Microbiol Lett 116:297–303<br />

Ferrol N, Barea JM, Azcón-Aguilar C (2000) The plasma membrane H + -ATPase genes<br />

family in the arbuscular mycorrhizal fungus Glomus mosseae. Curr Genet 37:112–118<br />

Filippi C, Bagnoli G, Citernesi AS, Giovannetti M (1998) Ultrastructural spatial distribution<br />

of bacteria associated with sporocarps of Glomus mosseae. Symbiosis 24:1–12<br />

Fitter AH, Garbaye J (1994). Interactions between mycorrhizal fungi and other soil<br />

organisms. Plant Soil 159: 123–132<br />

Franken P, Requena N (2001) Analysis of gene expression in arbuscular mycorrhizas:<br />

new approaches and challenges. New Phytol 150:517–523<br />

Frey P, Frey-Klett P, Garbaye J, Berge O, Heulin T (1997) Metabolic and genotypic fingerprinting<br />

of fluorescent Pseudomonads associated with the Douglas Fir-Laccaria biocolor<br />

mycorrhizosphere. Appl Environ Microbiol 63:1852–1860<br />

Galleguillos C, Aguirre C, Barea JM, Azcón R (2000) Growth promoting effect of two<br />

Sinorhizobium meliloti strains (a wild type and its genetically modified derivative) on<br />

a non-legume <strong>plant</strong> species in specific interaction with two arbuscular mycorrhizal<br />

fungi. Plant Sci 159:57–63<br />

Garbaye J (1994) Helper bacteria: A new dimension to the mycorrhizal symbiosis. New<br />

Phytol 128:197–210<br />

Germida JJ, Walley FL (1996) Plant growth-promoting rhizobacteria alter rooting patterns<br />

and arbuscular mycorrhizal fungi colonization of field-grown spring wheat.<br />

Biol Fertil Soils 23:113–120<br />

Gianinazzi S, Schüepp H (1994) Impact of arbuscular mycorrhizas on sustainable agriculture<br />

and natural ecosystems. ALS, Birkhäuser, Basel


366<br />

José-Miguel Barea et al.<br />

Gianinazzi S, Gianinazzi-Pearson V, Trouvelot A (1990) Potentialities and procedures for<br />

the use of endomycorrhizas with special emphasis on high value crops. In: Whipps<br />

JM, Lumsden B (eds) Biotechnology of fungi for improving <strong>plant</strong> growth. Cambridge<br />

University Press, Cambridge, pp 41–54<br />

Gianinazzi-Pearson V, Dumas-Gaudot E, Gollotte A, Tahiri-Alaoui A, Gianinazzi S (1996)<br />

Cellular and molecular defence-related root responses to invasion by arbuscular mycorrhizal<br />

fungi. New Phytol 133:45–57<br />

Giovannetti M (2000) Spore germination and pre-symbiotic mycelial growth In: Kapulnick<br />

Y, Douds DD Jr (eds) Arbuscular mycorrhizas: physiology and functions. Kluwer,<br />

Dordrecht, pp 47–68<br />

Glick BR (1995) The enhancement of <strong>plant</strong> growth by free-living bacteria. Can J Microbiol<br />

41:109–117<br />

Goicoechea N, Antolín MC, Sánchez-Díaz M (1997) Influence of arbuscular mycorrhizae<br />

and Rhizobium on nutrient content and water relations in drought stressed alfalfa.<br />

Plant Soil 192:261–268<br />

Goicoechea N, Szalai G, Antolín MC, Sánchez-Díaz M, Paldi E (1998) Influence of arbuscular<br />

mycorrhizae and Rhizobium on free polyamines and proline levels in waterstressed<br />

alfalfa. J Plant Physiol 153:706–711<br />

Gryndler M (2000) Interactions of arbuscular mycorrhizal fungi with other soil organisms<br />

In: Kapulnick Y, Douds Jr DD (eds) Arbuscular mycorrhizas: physiology and<br />

functions. Kluwer, Dordrecht, pp 239–262<br />

Gryndler M, Hrselova H (1998) Effect of diazotrophic bacteria isolated from a mycelium<br />

of arbuscular mycorrhizal fungi on colonization of maize roots by Glomus fistulosum.<br />

Biol Plant 41:617–621<br />

Gryndler M, Hrselová H, Stríteská D (2000) Effect of soil bacteria on growth of hyphae<br />

of the arbuscular mycorrhizal (AM) fungus Glomus claroideum. Folia Microbiol<br />

45:545–551<br />

Haas D, Keel C, Laville J, Maurhofer M, Oberliansli T, Schnider U,Voisard C, Wüthrich B,<br />

Defago G. (1991) Secondary metabolites of Pseudomonas fluorescens strain CHA0<br />

involved in the suppression of root diseases. In: Hennecke H, Verma DPS (eds)<br />

Advances in molecular genetics of <strong>plant</strong>-microbe interactions. Kluwer, Dordrecht, pp<br />

450–456<br />

Halverson LJ, Handelsman J (1991) Enhancement of soybean nodulation by Bacillus<br />

ceresus UW95 in the field and in a growth chambers. Appl Environ Microbiol<br />

57:2767–2770<br />

Harley JL, Smith SE (1983) Mycorrhizal symbiosis. Academic Press, New York<br />

Helgason T, Daniell TJ, Husband R, Fitter AH,Young JPW (1998) Ploughing up the woodwide<br />

web? Nature 394:431<br />

Hiltner L (1904) Über neuere Erfahrungen und Probleme auf dem Gebiet der Bodenbakteriologie<br />

und unter besonderer Berücksichtigung der Gründüngung und Brache.<br />

Arb Dtsch Landwirtsch Ges 98:59–78<br />

Jeffries P (1997) Mycoparasitism. In: Wicklow DT, Södertröm BE (eds) The Mycota IV.<br />

Environmental and microbial relationships. Springer, Berlin Heidelberg New York, pp<br />

149–164<br />

Jeffries P, Barea JM (2001) Arbuscular mycorrhiza – a key component of sustainable<br />

<strong>plant</strong>-soil ecosystems. In: Hock B (ed) The Mycota. vol. IX. Fungal Associations.<br />

Springer, Berlin Heidelberg New York, pp 95–113<br />

Keel C, Schnider U, Maurhofer M, Vorsand C, Laville J, Burger U, Wirthuer P, Hass D,<br />

Defago G (1992) Suppression of root diseases by Pseudomonas fluorescens CHAO:<br />

Importance of the bacterial secondary metabolite 2,4-diacetylphloroglucinol. Mol<br />

Plant-Microbe Interact 5:4–13


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 367<br />

Kennedy AC (1998) The rhizosphere and spermosphere In: Sylvia DM, Fuhrmann JJ,<br />

Hartel PG, Zuberer DA (eds) Principles and applications of soil <strong>microbiology</strong>. Prentice<br />

Hall, Upper Saddle River, New Jersey, pp 389–407<br />

Kennedy AC, Smith KL (1995) Soil microbial diversity and the sustainability of agricultural<br />

soils. Plant Soil 170:75–86<br />

Kim KY, Jordan D, McDonald GA (1998) Effect of phosphate-solubilizing bacteria and<br />

vesicular-arbuscular mycorrhizae on tomato growth and soil microbial activity. Biol<br />

Fertil Soils 26:79–87<br />

Kjoller R, Rosendahl S (2000) Detection of arbuscular mycorrhizal fungi (Glomales) in<br />

roots by nested PCR and SSCP (single stranded conformation polymorphism). Plant<br />

Soil 226:189–196<br />

Kloepper JW (1994) Plant growth-promoting rhizobacteria (other systems) In: Okon Y<br />

(ed) Azospirillum/<strong>plant</strong> associations. CRC Press, Boca Raton, pp 111–118<br />

Kloepper JW (1996) Host specificity in microbe-microbe interactions. BioScience<br />

46:406–409<br />

Kloepper JW, Zablotowick RM, Tipping EM, Lifshitz R (1991) Plant growth promotion<br />

mediated by bacterial rhizosphere colonizers. In: Keister DL, Cregan PB (eds) The rhizosphere<br />

and <strong>plant</strong> growth. Kluwer, Dordrecht, pp 315–326<br />

Klyuchnikov AA, Kozhevin PA (1990). Dynamics of Pseudomonas fluorescens and<br />

Azospirillum brasiliense populations in the formation of vesicular arbuscular mycorrhiza.<br />

Microbiologia 59:651–655<br />

Kozdrój J, Elsas JD van (2000) Application of polymerase chain reaction-denaturing gradient<br />

gel electrophoresis for comparison of direct and indirect extraction methods of<br />

soil DNA used for microbial community fingerprinting. Biol Fertil Soils 31:372–378<br />

Lagopodi AL, Ram AFJ, Lamers GEM, Punt PJ, Van den Hondel CAMJJ, Lugtenberg BJJ,<br />

Bloemberg GV (2002) Novel aspects of tomato root colonization and infection by<br />

Fusarium oxysporum f. sp. radicis-lycopersici revealed by confocal laser scanning<br />

microscopic analysis using the green fluorescent protein as a marker. Mol Plant-<br />

Microbe Interact 15:172–179<br />

Lambrecht M, Okon Y,Vande Broek A,Vanderleyden J (2000) Indole-3-acetic acid, a reciprocal<br />

signalling molecule in bacteria-<strong>plant</strong> interactions. Trends Microbiol 8:298–<br />

300<br />

Lemanceau P, Alabouvette C (1993). Suppression of fusarium-wilts by fluorescent<br />

pseudomonads: mechanisms and applications. Biocontrol Sci Technol 3:219–234<br />

Linderman RG (1988) Mycorrhizal interactions with the rhizosphere microflora. The<br />

mycorrhizosphere effects. Phytopathology 78:366–371<br />

Linderman RG (1992) Vesicular-arbuscular mycorrhizae and soil microbial interactions.<br />

In: Bethlenfalvay GJ, Linderman RG (eds) Mycorrhizae in sustainable agriculture.<br />

ASA Spec, Madison, Wisconsin, pp 45–70<br />

Linderman RG (1994) Role of VAM fungi in biocontrol. In: Pfleger FL, Linderman RG<br />

(eds) Mycorrhizae and <strong>plant</strong> health. APS Press, St Paul, pp 1–26<br />

Linderman RG (2000) Effects of mycorrhizas on <strong>plant</strong> tolerance to diseases. In: Kapulnik<br />

Y, Douds DD Jr (eds) Arbuscular mycorrhizas: physiology and function. Kluwer, Dordrecht,<br />

pp 345–365<br />

Lovato PE, Schüepp H, Trouvelot A, Gianinazzi S (1995) Application of arbuscular mycorrhizal<br />

fungi (AMF) in orchard and ornamental <strong>plant</strong>s. In: Varma A, Hock B (eds)<br />

Mycorrhiza structure, function, molecular biology and biotechnology. Springer,<br />

Berlin Heidelberg New York, pp 521–559<br />

Lugtenberg BJJ, Weger de LA, Bennett JW (1991) Microbial stimulation of <strong>plant</strong> growth<br />

and protection from disease. Curr Opin Microbiol 2:457–464<br />

Lugtenberg BJJ, Dekkers L, Bloemberg GV (2001) Molecular determinants of rhizosphere<br />

colonization by bacteria. Annu Rev Phytopathol 39:461–490


368<br />

José-Miguel Barea et al.<br />

Lynch JM (1990) The rhizosphere. Wiley, New York, p 462<br />

Marschner P, Crowley DE (1996a) Root colonization of mycorrhizal and nonmycorrhizal<br />

pepper (Capsicum annuum) by Pseudomonas fluorescens 2–79RL. New Phytol<br />

134:115–122<br />

Marschner P, Crowley DE (1996b) Physiological activity of a bioluminescent<br />

Pseudomonas fluorescens (strain 2–79) in the rhizosphere of mycorrhizal and nonmycorrhizal<br />

pepper (Capsicum annuum L). Soil Biol Biochem 28:869–876<br />

Miller RM, Jastrow JD (1994) Vesicular-arbuscular mycorrhizae and biogeochemical<br />

cycling. In: Pfleger FL, Linderman RG (eds) Mycorrhizae and <strong>plant</strong> health. APS Press,<br />

St. Paul, MN, pp 189–212<br />

Miller RM, Jastrow JD (2000) Mycorrhizal fungi influence soil structure. In: Kapulnik Y,<br />

Douds DD Jr (eds) Arbuscular mycorrhizas: physiology and functions. Kluwer, Dordrecht,<br />

pp 3–18<br />

Moenne-Loccos Y, Tichy HV, O’Donnell A, Simon R, O’Gara F (2001) Impact of 2,4diacetylphloroglucinol-producing<br />

biocontrol strain Pseudomonas fluorescens F113<br />

on intraspecific diver of resident culturable fluorescent pseudomonads associate with<br />

the roots of field-grown sugar beet seedlings. Appl Environ Microbiol 67:3418–3425<br />

Mohammad A, Khan AG, Kuek C (2000) Improved aeroponic culture of inocula of arbuscular<br />

mycorrhizal fungi. Mycorrhiza 9:337–339<br />

Monzón A, Azcón R (1996) Relevance of mycorrhizal fungal origin and host <strong>plant</strong> genotype<br />

to inducing growth and nutrient uptake in Medicago species. Agric Ecosyst Environ<br />

60:9–15<br />

Morton J.B. (2000) Evolution of endophytism in arbuscular mycorrhizal fungi of Glomales.<br />

In: Bacon CW, White Jr JE (eds) Microbial endophytes. Marcel Dekker, New York,<br />

pp 121–140<br />

Morton JB, Redecker D (2001) Two new families of Glomales, Archaeosporaceae and<br />

Paraglomaceae, with two new genera Archaeospora and Paraglomus, based on concordant<br />

molecular and morphological characters. Mycologia 93:181–195<br />

Nehl DB, Allen SJ, Brown JF (1996) Deleterious rhizosphere bacteria: an integrating perspective.<br />

Appl Soil Ecol 5:1–20<br />

O’Gara F, Dowling DN, Boesten B (1994) Molecular ecology of rhizosphere microorganisms.VCH,<br />

Weinheim, p 173<br />

Okon Y (1994) Azospirillum/Plant associations. CRC Press, Boca Raton, p 175<br />

Probanza A, Lucas García JA, Ruiz Palomino M, Ramos B, Gutiérrez Mañero FJ (2002)<br />

Pinus pinea L. seedling growth and bacterial rhizosphere structure after inoculation<br />

with PGPR Bacillus (B. licheniformis CECT 5106 and B. pumillus CECT 5105). Appl<br />

Soil Ecol 20:75–84<br />

Puppi G, Azcón R, Höflich G (1994) Management of positive interactions of arbuscular<br />

mycorrhizal fungi with essential groups of soil microorganisms. In: Gianinazzi S,<br />

Schüepp H (eds) Impact of arbuscular mycorrhizas on sustainable agriculture and<br />

natural ecosystems. ALS, Birkhäuser, Basel, pp 201–215<br />

Ravnskov S, Nybroe O, Jakobsen I (1999) Influence of an arbuscular mycorrhizal fungus<br />

on Pseudomonas fluorescens DF57 in rhizosphere and hyphosphere soil. New Phytol<br />

142:113–122<br />

Redecker D, Thierfelder H, Walker C, Werner D (1997) Restriction analysis of PCRamplified<br />

internal transcribed spacers of ribosomal DNA as a tool for species identification<br />

in different genera of the order Glomales. Appl Environ Microbiol<br />

63:1756–1761<br />

Redecker D, Morton JB, Bruns TD (2000) Ancestral lineages of arbuscular mycorrhizal<br />

fungi (Glomales). Mol Phylogenet Evol 14:276–284<br />

Requena N, Jimenez I, Toro M, Barea JM (1997) Interactions between <strong>plant</strong>-growth- promoting<br />

rhizobacteria (PGPR), arbuscular mycorrhizal fungi and Rhizobium spp. in


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 369<br />

the rhizosphere of Anthyllis cytisoides, a model legume for revegetation in Mediterranean<br />

semi-arid ecosystems. New Phytol 136:667–677<br />

Requena N, Perez-Solis E, Azcón-Aguilar C, Jeffries P, Barea JM (2001) Management of<br />

indigenous <strong>plant</strong>-microbe symbioses aids restoration of desertified. Appl Environ<br />

Microbiol 67:495–498<br />

Ruiz-Lozano JM, Azcón R (1993) Specificity and functional compatibility of VA mycorrhizal<br />

endophytes in association with Bradyrhizobium strains in Cicer arietinum.<br />

Symbiosis 15:217–226<br />

Ruiz-Lozano JM, Bonfante P (2000) A Burkholderia strain living inside the arbuscular<br />

mycorrhizal fungus Gigaspora margarita possesses the vacB gene, which is involved<br />

in host cell colonization by bacteria. Microbial Ecol 39:137–144<br />

Ruiz-Lozano JM, Collados C, Barea JM, Azcón R (2001) Arbuscular mycorrhizal symbiosis<br />

can alleviate drought-induced nodule senescence in soybean <strong>plant</strong>s. New Phytol<br />

151:493–502<br />

Sanders IR, Clapp JP,Wiemken A (1996) The genetic diversity of arbuscular mycorrhizal<br />

fungi in natural ecosystems – A key to understanding the ecology and functioning of<br />

the mycorrhizal symbiosis. New Phytol 133:123–134<br />

Schloter M, Wiehe W, Assmus B, Steindl H, Becke H, Höflich G, Hartmann A (1997) Root<br />

colonization of different <strong>plant</strong>s by <strong>plant</strong>-growth-promoting Rhizobium leguminosarum<br />

bv. trifolii R39 studied with monospecific polyclonal antisera. Appl Environ<br />

Microbiol 63:2038–2046<br />

Schübler A, Schwarzott D, Walker C (2001) A new fungal phylum, the Glomeromycota:<br />

phylogeny and evolution. Mycol Res 105:1413–1421<br />

Siciliano SD, Germida JJ (1998) BIOLOG analysis and fatty acid methyl ester profiles<br />

indicate that pseudomonad inoculants that promote phytoremediation alter the rootassociated<br />

microbial community of Bromus biebersteinii. Soil Biol Biochem<br />

30:1717–1723<br />

Simon L, Bousquet J, Lévesque RC, Lalonde M (1993). Origin and diversification of<br />

endomycorrhizal fungi and coincidence with vascular land <strong>plant</strong>s. Nature 363:67–69<br />

Simons M, van der Bij AJ, Brand I, de Weger LA, Wijffelman CA, Lugtenberg BJJ (1996)<br />

Gnotobiotic system for studying rhizosphere colonization by <strong>plant</strong> growth-promoting<br />

Pseudomonas bacteria. Mol Plant-Microbe Interact 9:600–607<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis. Academic Press, London<br />

Smith SE, Gianinazzi-Pearson V, Koide R, Cairney JWG (1994) Nutrient transport in<br />

mycorrhizas: structure, physiology and consequences for efficiency of the symbiosis.<br />

In: Robson AD, Abbott LK, Malajczuk N (eds) Management of mycorrhizas in agriculture,<br />

horticulture and forestry. Kluwer, Dordrecht, pp 103–113<br />

Staley TW, Lawrence EG, Nance EL (1992) Influence of a <strong>plant</strong> growth-promoting<br />

pseudomonad and vesicular-arbuscular mycorrhizal fungus on alfalfa and birdsfoot<br />

trefoil growth and nodulation. Biol Fertil Soils 14:175–180<br />

Sturz AV Nowak J (2000) Endophytic communities of rhizobacteria and the strategies<br />

required to create yield enhancing associations with crops. Appl Soil Ecol 15:183–190<br />

Sturz AV, Christie BR, Nowak J (2000) Bacterial endophytes: potential role in developing<br />

sustainable systems of crop production. Crit Rev Plant Sci 19:1–30<br />

Sylvia DM (1998) Mycorrhizal symbioses. In: Sylvia DM, Fuhrmann JJ, Hartel PG,<br />

Zuberer DA (eds) Principles and applications of soil <strong>microbiology</strong>. Prentice Hall,<br />

Upper Saddle River, New Jersey, pp 408–426<br />

Thomashow LS,Weller DM (1995) Current concepts in the use of introduced bacteria for<br />

biological control: mechanisms and antifungal metabolites. In: Stacey G, Keen N (eds)<br />

Plant-microbe interactions. Chapman and Hall, New York, pp 187–235<br />

Toro M,Azcón R, Barea JM (1997) Improvement of arbuscular mycorrhizal development<br />

by inoculation with phosphate-solubilizing rhizobacteria to improve rock phosphate<br />

bioavailability ( 32 P) and nutrient cycling. Appl Environ Microbiol 63: 4408–4412


370<br />

José-Miguel Barea et al.<br />

Toro M,Azcón R, Barea JM (1998) The use of isotopic dilution techniques to evaluate the<br />

interactive effects of Rhizobium genotype, mycorrhizal fungi, phosphate-solubilizing<br />

rhizobacteria and rock phosphate on nitrogen and phosphorus acquisition by Medicago<br />

sativa. New Phytol 138:265–273<br />

Tuinen D van, Jacquot E, Zhao B, Gollotte A, Gianinazzi-Pearson V (1998) Characterization<br />

of root colonization profiles by a microcosm community of arbuscular mycorrhizal<br />

fungi using 25S rDNA-targeted nested PCR. Mol Ecol 7:879–887<br />

Valdenegro M, Barea JM,Azcón R (2001) Influence of arbuscular-mycorrhizal fungi, Rhizobium<br />

meliloti strains and PGPR inoculation on the growth of Medicago arborea<br />

used as model legume for re-vegetation and biological reactivation in a semi-arid<br />

Mediterranean area. Plant Growth Regul 34:233–240<br />

Van Bastelaere E, Lambrecht M, Vermeiren H, Keijers V, de Wilde P, Proost P, Van Dommelen<br />

A, Varderleyden J (1999) Characterization of a sugar-binding protein from<br />

Azospirillum brasilense mediating chemotaxis to and uptake of sugars. Mol Microbiol<br />

32:703–714<br />

Van de Broek A, Lambrecht M, Vanderleyden J (1999) Auxins upregulate expression of<br />

the indole-3-pyruvate decaboxylase gene from Azospirillum brasilense. J Bacteriol<br />

181:1338–1342<br />

Van Loon LC, Bakker PAHM, Pieterse CMJ (1998) Systemic resistance induced by rhizosphere<br />

bacteria. Annu Rev Phytopathol 36:453–483<br />

Varma A, Schüepp H (1995) Mycorrhization of the commercially important micropropagated<br />

<strong>plant</strong>s. Crit Rev Biotechnol 15:313–328<br />

Varma A,Verma S, Sudha, Sahay N, Bütehorn B, Franken P (1999) Piriformospora indica,<br />

a cultivable <strong>plant</strong>-growth-promoting root endophyte. Appl Environ Microbiol<br />

65:2741–2744<br />

Vázquez MM, Cesar S, Azcón R, Barea JM (2000) Interactions between arbuscular mycorrhizal<br />

fungi and other microbial inoculants (Azospirillum, Pseudomonas, Trichoderma)<br />

and their effects on microbial population and enzyme activities in the rhizosphere<br />

of maize <strong>plant</strong>s. Appl Soil Ecol 15:261–272<br />

Vierheilig H, Piché Y (2002) Signalling in arbuscular mycorrhiza: facts and hypotheses<br />

In: Buslig B, Manthey J (eds) Flavonoids in cell functions. Kluwer Academic/Plenum<br />

Publishers, New York, pp 23–29<br />

Vestberg M, Estaún V (1994) Micropropagated <strong>plant</strong>s, an opportunity to positively manage<br />

mycorrhizal activities. In: Gianinazzi S, Schüepp H (eds) Impact of arbuscular<br />

mycorrhizas on sustainable agriculture and natural ecosystems. Birkhäuser, Basel, pp<br />

217–226<br />

Volpin H, Kapulnik Y (1994) Interaction of Azospirillum with beneficial soil microorganisms.<br />

In: Okon Y (ed) Azospirillum/<strong>plant</strong> associations. CRC Press, Boca Raton, pp<br />

111–118<br />

Vosátka M (1996) Soil bacteria – a component of <strong>plant</strong>, soil and arbuscular mycorrhizal<br />

fungal interactions. In: Azcon-Aguilar C, Barea JM (Eds) Mycorrhizas in integrated<br />

systems – from genes to <strong>plant</strong> development. (pp 613–618). European Commission<br />

Report EUR 16728, Brussels, Luxembourg<br />

Vosátka M, Gryndler M (1999) Treatment with culture fractions from Pseudomonas<br />

putida modifies the development of Glomus fistulosum mycorrhiza and the response<br />

of potato and maize <strong>plant</strong>s to inoculation. Appl Soil Ecol 11:245–251<br />

Waschkies C, Schropp A, Marschner H (1994). Relations between grapevine re<strong>plant</strong> disease<br />

and root colonization of grapevine (Vitis sp.) by fluorescent pseudomonads and<br />

endomycorrhizal fungi. Plant Soil 162:219–227<br />

Weller DM, Thomashow LS (1994) Current challenges in introducing beneficial microorganisms<br />

into the rhizosphere. In: O’Gara F, Dowling DN, Boesten B (eds) Molecular<br />

ecology of rhizosphere microorganisms biotechnology and the release of GMOs.<br />

VCH, Weinheim, pp 1–18


20 Mycorrhizal Fungi and Plant Growth Promoting Rhizobacteria 371<br />

Werner D (1998) Organic signals between <strong>plant</strong>s and microorganisms. In: Pinton R,<br />

Varanini Z, Nannipieri P (eds) The rhizosphere: biochemistry and organic substances<br />

at the soil-<strong>plant</strong> interfaces. Marcel Dekker, New York<br />

Werner D, Barea J-M, Brewin NJ, Cooper P, Katinakis P, Lindström K, O’Gara F, Spaink<br />

HP, Truchet G, Müller P (2002) Symbiosis and defence in the interaction of <strong>plant</strong>s with<br />

microorganisms. Symbiosis 32:83–104<br />

Whitelaw MA (2000) Growth promotion of <strong>plant</strong>s inoculated with phosphate-solubilizing<br />

fungi. Adv Agron 69:99–151<br />

Zinniel DK, Lambrecht P, Harris NB, Feng Z, Kuczmarski D, Higley P, Ishimaru CA,<br />

Arunakumari A, Barletta RG, Vidaver AK (2002) Isolation and characterization of<br />

endophytic colonizing bacteria from agronomic crops and prairie <strong>plant</strong>s. Appl Environ<br />

Microbiol 68:2198–2208


21 Carbohydrates and Nitrogen: Nutrients and<br />

Signals in Ectomycorrhizas<br />

Uwe Nehls<br />

1 Introduction<br />

Due to <strong>plant</strong> litter, forest soil is rich in complex carbohydrates (e.g., cellulose<br />

and lignin). Nevertheless, these carbohydrates are only slowly degraded by<br />

specialized microorganisms and thus forest soils are rather poor in readily<br />

cleavable carbohydrates that are necessary for the growth of the majority of<br />

microbes including ectomycorrhizal fungi.<br />

Basidiomycetes are able to transfer nutrients and metabolites over long distances.<br />

Exploring a rich source of readily utilizable carbohydrates would thus<br />

favor the colonization of other soil areas, too. The association with fine roots<br />

of woody <strong>plant</strong>s forming ectomycorrhizas is a way that secures exclusive<br />

access to such a rich carbohydrate source for ectomycorrhizal fungi.<br />

Organic compounds contained in root exudates are candidates for the carbon<br />

transfer from the host to the mycorrhizal fungus. Low-molecular-weight<br />

root exudates comprise soluble sugars, carboxylic acids and amino acids<br />

(Marschner 1995; Smith and Read 1997; Hampp and Schaeffer 1999). The best<br />

growth of ectomycorrhizal fungi (ECM) fungi occurs on the hexoses glucose,<br />

fructose, and mannose. Sucrose, which is the preferred transport sugar in<br />

most host <strong>plant</strong>s cannot be used by ECM investigated so far (e.g., Salzer and<br />

Hager 1991), Laccaria bicolor being possibly an exception (Tagu et al. 2000).<br />

Even if <strong>plant</strong>-derived hexoses are most important for ectomycorrhizal<br />

fungi, there is ample evidence that soil carbon sources are also intensively<br />

used. Among these are starch, dextrins, glucans, oligosaccharides or sugar<br />

alcohols (Palmer and Hacskaylo 1970; Cao and Crawford 1993; Berredjem et<br />

al. 1998), proteins (Abuzinadah and Read 1986), or even cellulose or lignin<br />

(Norkrans 1950; Trojanowski et al. 1984; Taber and Taber 1987; Haselwandter<br />

et al. 1990).<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


374<br />

Uwe Nehls<br />

2 Trehalose Utilization by Ectomycorrhizal Fungi<br />

Trehalose is a nonreducing disaccharide that is found in a wide variety of<br />

organisms including bacteria, fungi, protozoa, nematodes, insects and <strong>plant</strong>s<br />

(Elbein 1974; Mellor 1992, Müller et al. 1994). Trehalose has been shown to be<br />

a good carbon source for a number of ectomycorrhizal fungi (Lewis and<br />

Harley 1965; Palmer and Hacskaylo 1970).<br />

Amanita muscaria hyphae excrete an acid trehalase into the growth<br />

medium to break down external trehalose (Wisser et al. 2000).With its apparent<br />

molecular mass of 165 kDa, a pI of 3.7, a pH optimum of about 4.0 and an<br />

apparent Km value for trehalose of 0.38 mM, it is comparable to that of other<br />

fungi.<br />

Excreted acid trehalases, in general, are very stable. Owing to the low Km<br />

value of the enzyme, and the acidic pH optimum, trehalose hydrolysis in<br />

acidic forest soils should be very efficient. The resulting monosaccharides are<br />

then taken up by the highly efficient monosaccharide import system of A.<br />

muscaria (Chen and Hampp 1993; Nehls et al. 1998, see below).<br />

Carbohydrate-starved mycelia excreted about four times more acid trehalase<br />

into the growth medium than mycelia that were well supported with<br />

sugar (Wisser 2000), indicating an up-regulated expression of acid trehalase<br />

with regard to poor carbon nutrition.<br />

Trehalose is one of the main storage carbohydrates of basidiomycetes, but<br />

is also found in bacteria in large quantities. The utilization of trehalose by<br />

ectomycorrhizal fungi might thus be important for two reasons:<br />

1. The availability of an additional carbon source would improve ectomycorrhizal<br />

fungal growth in soil.<br />

2. By utilization of a soil carbon source that otherwise could be used by other<br />

microorganisms, ectomycorrhizal fungi could reduce the growth of their<br />

putative competitors for other nutrients (e.g., nitrogen or phosphate).<br />

3 Carbohydrate Uptake<br />

A prerequisite for a rapid uptake of monosaccharides are membrane transport<br />

systems. Experiments with suspension-cultured hyphae of ectomycorrhizal<br />

fungi (Salzer and Hager 1991) and with protoplasts (Chen and Hampp<br />

1993) indicated that most basidiomycotic ectomycorrhizal fungi have no system<br />

for sucrose import or hydrolysis, but for the uptake of glucose and fructose<br />

(Palmer and Hacskaylo 1970; Salzer and Hager 1991).<br />

To date, only two hexose transporter genes from A. muscaria (Fig. 1),<br />

AmMst1 (Nehls et al. 1998) encoding a protein of 520 amino acids and<br />

AmMst2 encoding a protein of 519 amino acids, and one hexose transporter<br />

gene from Tuber borchii (Agostini and Stocchi, pers. comm.) have been identified.<br />

While AmMst1 reveals the highest sequence homology with Hxt1 from


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 375<br />

Fig. 1. Dendrogram of the alignment of the deduced protein sequence of AmMST1 and<br />

AmMst2 with known fungal monosaccharide transporters. The relationship of the<br />

deduced protein sequence of AmMST1 and AmMst2 to other fungal monosaccharide<br />

transporters was determined by multiple alignment using ClustalW (version 1.8)<br />

Uromyces fabae, AmMst2 has the best homologies to Stl1 of S. cerevisiae.The<br />

homology between both deduced Amanita proteins is low (33.3 % identity,<br />

56.8 % similarity). Interestingly, the noncoding region of the cDNAs revealed<br />

a higher identity (approx. 60 %) than the coding region (approx. 50 %). The<br />

expression patterns with regard to carbon and nitrogen nutrition are identical<br />

for both genes (Nehls, unpublished). When expressed in yeast, AmMst1<br />

revealed K M values of 0.46 mM for glucose and 4.2 mM for fructose, indicating<br />

a strong preference for glucose (Wiese et al. 2000). Also, A. muscaria hyphae<br />

strongly favored glucose uptake even in the presence of a large excess of fructose<br />

(20 mM vs. 1 mM; Nehls et al. 2001 c). A similar preference for glucose<br />

uptake was also observed for the ectomycorrhizal ascomycete Cenococcum<br />

geophilum (Stülten et al. 1995), indicating that this behavior might be com-


376<br />

Uwe Nehls<br />

mon to ectomycorrhizal fungi. Since A. muscaria revealed only one type of<br />

hexose uptake kinetic, mostly resembling that of AmMst1 when expressed in<br />

yeast, it is very likely that AmMst1 and AmMst2 have similar transport properties.<br />

The investigated A. muscaria hyphae are heterocaryotic, containing<br />

nuclei of two different origins. It could thus be questioned whether AmMst1<br />

and AmMst2 are encoding different genes from one nucleus or both genes<br />

belong to different nuclei.<br />

4 Carbohydrate Metabolism<br />

As in other organisms phosphofructokinase (ATP-dependent) is the rate-limiting<br />

step in fungal glycolysis (Kowallik et al. 1998). In A. muscaria, this<br />

enzyme is activated by fructose 2,6-bisphosphate (F26BP; k a about 30 nM;<br />

Schaeffer et al. 1996) which is similar to the properties of the corresponding<br />

enzyme from yeast or animal cells, but differs from <strong>plant</strong> phosphofructokinases.<br />

It has been shown that A. muscaria mycelia grown in the presence of<br />

high hexose concentrations as well as mycorrhizal roots have increased<br />

amounts of F26BP (Schaeffer et al. 1996; Hoffmann et al. 1997). This could<br />

indicate increased rates of glycolysis in hyphae under elevated hexose supply,<br />

e.g., hyphae of the Hartig net (Hampp and Schaeffer 1999).<br />

In yeast cells, levels of fructose-2,6 bisphosphate, and thus the predominance<br />

of glycolysis over gluconeogenesis, are controlled by the formation of<br />

cyclic AMP (cAMP). Increased glucose supply causes an increase of activity of<br />

adenylate cyclase (Thevelein 1991) and thus of the cAMP content in hyphae.<br />

cAMP activates a cAMP-dependent protein kinase (PKA) which, via phosphorylation,<br />

activates F26BP formation while inhibiting F26BP degradation<br />

(Thevelein 1991; d’Enfert 1997; RadisBaptista et al. 1998).<br />

At least the initial steps of glucose-dependent regulation of glycolysis also<br />

exist in A. muscaria. Changes in pool sizes of cAMP were detected in relation<br />

to glucose supply (Hoffmann et al. 1997). When suspension cultures of A.<br />

muscaria were transferred from medium containing low (1 mM) to high<br />

(40 mM) glucose concentrations, both cAMP pools as well as rates of activity<br />

of protein kinase A increased (Nehls et al. 2001 c).<br />

5 Carbohydrate Storage<br />

In addition to their relevance for carbon storage, storage carbohydrates have<br />

additional functions in fungi,e.g.,the rapid conversion of carbohydrates that are<br />

taken up (to maintain a carbon sink) or membrane and protein protection (e.g.,<br />

trehalose). Two different pools of storage carbohydrates can be distinguished in<br />

ectomycorrhizal fungi: short chain carbohydrates (trehalose) or polyols (mannitol,arabitol,erythritol),and<br />

the long chain carbohydrate glycogen.


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 377<br />

Hexoses that are taken up by ectomycorrhizal fungi are either introduced<br />

into glycolysis (e.g., formation of amino acids) or converted into short chain<br />

carbohydrates and polyols (Martin et al. 1985, 1987, 1988, 1998). Growth of A.<br />

muscaria (suspension culture) on glucose as a carbon source resulted in an<br />

increase in the trehalose content until the external glucose concentration was<br />

below 4 mM, followed by a depletion of trehalose concentration over time. In<br />

contrast, the glycogen content was stable during the investigation (4 weeks;<br />

Wallenda 1996). In Lactarius sp. the glycogen content was high during winter,<br />

declined until summer and was restored during the autumn (Genet et al.<br />

2000). Thus, trehalose and polyols are presumably short term storage compounds,<br />

revealing high fluctuation rates, while glycogen is the long term storage<br />

carbohydrate of hyphae that is only mobilized when the short term pools<br />

are empty.<br />

Ectomycorrhizal fungal colonies could become quite large, and support<br />

from <strong>plant</strong>-derived carbohydrates has been shown to be necessary for fungal<br />

growth in soil (Leake et al. 2001). Thus, long-distance transport of carbon is of<br />

great interest for fungal physiology. In P. involutus ectomycorrhizas, glycogen<br />

particles were observed in the Hartig net and the inner and outer hyphal layers<br />

of the fungal sheath (Jordy et al. 1998). Since glycogen is stored in the cytoplasm<br />

as large, nonmobile granules it is rather likely that glycogen is not the<br />

long-distance transport form of carbohydrates. Polyols and trehalose are, in<br />

addition to glucose, present in large quantities in fungal hyphae, and are thus<br />

good candidates for long-distance transport carbohydrates between different<br />

parts of the fungal colony.<br />

6 Carbohydrates as Signal, Regulating Fungal Gene<br />

Expression in Ectomycorrhizas<br />

Sugar-regulated gene expression was mainly investigated in saprophytic<br />

ascomycetes (Jennings 1995). Here, the external monosaccharide concentration<br />

regulates fungal gene expression, e.g., that of monosaccharide transporters<br />

at the transcriptional as well as the posttranscriptional (protein<br />

degradation rate) level. Two different transcriptional control mechanisms,<br />

induction/enhancement or repression of gene expression were described<br />

(Felenbok and Kelley 1996; Özcan et al. 1996).<br />

In A. muscaria, the expression of the hexose transporter genes is up-regulated<br />

by a threshold response mechanism depending on the extracellular concentration<br />

of monosaccharides (Nehls et al. 1998). In A. muscaria hyphae<br />

grown in the presence of glucose concentrations up to 2 mM, the glucose<br />

transporter genes AmMst1 and AmMst2 are expressed at a basal level, while<br />

monosaccharide concentrations above that threshold triggered at least a fourfold<br />

increase in the transcript levels. This up-regulation could not be further<br />

enhanced by hexose concentrations of up to 100 mM. Since the increase of


378<br />

Uwe Nehls<br />

monosaccharide transporter gene expression is a slow process, it could be<br />

interpreted as an adaptation to the elevated hexose concentrations usually<br />

only found at the <strong>plant</strong>/fungus interface, but not in the soil.<br />

In yeast, sugar dependent induction/enhancement of gene expression is<br />

controlled by two monosaccharide transporter-like proteins, RGT2 and SNF3,<br />

sensing the external sugar concentration (Celenza et al. 1988; Özcan et al.<br />

1996). These transporters have a C-terminal extension containing a conserved<br />

amino acid motive thought to be involved in the transduction of the external<br />

sugar signal. Due to their different glucose affinities, SNF3 senses low external<br />

glucose concentrations while RGT2 senses high concentrations. In contrast to<br />

yeast, the putative monosaccharide transporter RCO3 (Madi et al. 1997) that is<br />

presumably also acting as monosaccharide sensor in Neurospora crassa,does<br />

not contain any extension. The signal cascade, transforming the sugar signal<br />

into modified gene expression, is not fully understood. To date, two elements<br />

have been identified, the transcription factor RGT1 (Özcan et al. 1996) and a<br />

signal transduction mediator, the SCF complex (Özcan and Johnston 1999).<br />

Without the sugar signal, RGT1 is a repressor for glucose-induced genes while<br />

activation via the SCF complex (in response to a sugar signal) modifies RGT1<br />

function to that of a transcriptional activator (Johnston 1999).<br />

The signal regulating the hexose-dependent, enhanced AmMst1 expression<br />

is still unknown, but in contrast to yeast, it seems to be transmitted by an<br />

internal and not an external sensor. Glucose analogues that are imported by<br />

AmMst1 and phosphorylated, but not further metabolized, did not increase<br />

the AmMst1 transcript level as glucose did (Wiese et al. 2000). Furthermore,<br />

the result of these experiments makes it rather likely that the signal must be<br />

generated downstream of hexokinase activity, in glycolysis or carbon storage<br />

pathways.<br />

While AmMst1 expression is an example of sugar-dependent enhancement<br />

of gene expression in A. muscaria, a second gene (AmPAL) was identified that<br />

revealed sugar-dependent gene repression (Nehls et al. 1999a). PAL is a key<br />

enzyme of secondary metabolism and thus of the production of phenolic<br />

compounds. ECM-forming fungi have been reported to use phenolic compounds<br />

for both their own protection and that of their host against bacterial<br />

or fungal attacks (Marx 1969; Chakravarty and Unestam 1987; Garbaye 1991).<br />

In A. muscaria, the transcript of AmPAL was abundant in hyphae grown at<br />

low external glucose concentrations, but exhibited a significant decrease in<br />

hyphae cultured at glucose concentrations of above 2 mM (less than 1/30 of<br />

the transcript level at low glucose). Unlike AmMst1, AmPAL-expression is<br />

probably regulated by sugar phosphorylation via hexokinase as sugar sensor<br />

(Nehls et al. 1999a).<br />

Also in saprophytic ascomycetes the monosaccharide-dependent gene<br />

repression is regulated via a hexokinase-dependent signaling pathway (Ronne<br />

1995; Gancedo 1998). The molecular mechanism of signal initiation is still<br />

unclear, but a hexokinase (in yeast mainly hex2) initiates the signal in


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 379<br />

response to the C-flux through the enzyme. Whether hexokinase phosphorylation<br />

or its association with other proteins is responsible for signal generation<br />

is still a mater of debate, but it is rather likely that the conformational<br />

changes of the enzyme during its enzymatic activity are sensed and not the<br />

generated hexose phosphates per se. The signaling pathways involve the activation/deactivation<br />

of the SNF1 protein complex that is presumably mediated<br />

by phosphorylation/dephosphorylation (AMPK kinase and REG1/GLC7<br />

phosphatase complex, respectively; Lesage et al. 1996; Johnston 1999). The<br />

sugar-dependent gene repression is mediated by a DNA-binding protein like<br />

MIG1 (yeast) or CREA (Aspergillus, Neurospora; Felenbok and Kelly 1996),<br />

acting as repressors.<br />

While in the pure fungal culture AmMst1 was induced and AmPAL<br />

repressed by elevated hexose concentrations, both genes were strongly<br />

expressed in entire mycorrhizas (Nehls et al. 1998, 1999a). It could thus be<br />

concluded that in mycorrhizas the sugar-dependent regulation of both genes<br />

is either modified by developmental events, or different in the sheath and Hartig<br />

net hyphae. To address this question, ectomycorrhizas were dissected and<br />

gene expression was investigated separately for hyphae of the fungal sheath<br />

and the Hartig net (Nehls et al. 2001a). Similar to low external hexose concentrations<br />

in pure fungal culture, AmMst1 was expressed only at the basal level<br />

in hyphae of the fungal sheath. In contrast, AmPAL revealed a high transcript<br />

level in this fungal structure. For Hartig net hyphae the opposite expression<br />

pattern was observed. As for hyphae in pure culture in the presence of high<br />

external hexose concentrations, the transcript level of AmMst1 was sixfold<br />

enhanced while the expression of AmPAL was only barely detectable.<br />

Owing to the opposite regulation of both genes in hyphae of fungal sheath<br />

and Hartig net that resembles the hexose-dependent expression of these<br />

genes in pure culture, different hexose concentrations in the apoplast of the<br />

fungus/<strong>plant</strong> interface (hexose concentration >2 mM) and the apoplast of the<br />

fungal sheath (hexose concentration 2 mM) could be assumed in<br />

the Hartig net that would trigger the observed hexose-dependent fungal gene<br />

expression. Fructose withdrawal from the apoplast presumably takes place<br />

mainly within the innermost one or two layers of the fungal sheath since fructose<br />

uptake by A. muscaria hyphae is rather efficient when the glucose concentration<br />

is


380<br />

Uwe Nehls<br />

Fig. 2. Spatial distribution of hexose uptake by fungal hyphae in ectomycorrhizas: a<br />

model. Sucrose hydrolysis in the apoplast of the Hartig net results in high glucose and<br />

fructose concentrations.We assume that here glucose is taken up preferentially, since the<br />

uptake of fructose is inhibited (by glucose concentrations above 0.5 mM). In the innermost<br />

one or two layers of the fungal sheath glucose concentration is low, due to efficient<br />

uptake by fungal hyphae of the Hartig net. Thus, most probably fructose is taken up. In<br />

the apoplast of the majority of the fungal sheath, glucose as well as fructose concentrations<br />

are low due to the efficient hexose uptake by hyphae of the Hartig net and the inner<br />

layers of the sheath<br />

sion of AmMst1 and a repression of AmPAL) are present in the apoplast of the<br />

majority of fungal sheath hyphae.<br />

“Metabolic zonation” and “physiological heterogeneity” have already been<br />

discussed as important concepts for a functional understanding of ectomycorrhizal<br />

symbiosis (Martin et al. 1992; Cairney and Burke 1996; Timonen and<br />

Sen 1998). Differences in the apoplastic hexose concentration at the Hartig<br />

net vs. fungal sheath could thus be supposed to generate a signal that might<br />

regulate fungal physiological heterogeneity in ectomycorrhizas, in addition to<br />

the developmental program.<br />

7 Nitrogen<br />

The ability of ectomycorrhizal fungi to take up inorganic nitrogen is well<br />

established (Melin and Nilsson 1952; France and Reid 1983; Plassard et al.


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 381<br />

1986; Finlay et al. 1988; Chalot and Brun 1998). In accordance with the predominant<br />

occurrence of ammonium as inorganic nitrogen source in the soil,<br />

most ectomycorrhizal fungi grow better on ammonium than on nitrate in<br />

pure culture (France and Reid 1984; Finlay et al. 1992). Nevertheless, even in<br />

mature forests nitrate could be present in large amounts as a result of bacterial<br />

activity (e.g., open forest areas) or as a result of fertilization in areas with<br />

extensive agriculture (Gessler et al. 1998).<br />

In many forest ecosystems, rates of nitrogen mineralization of litter are low<br />

and consequently, the supply of inorganic nitrogen is often limited (Read<br />

1991). In addition, nitrification is usually slow and the poorly mobile ammonium<br />

ion (Keeney 1980) predominates together with organic nitrogen (e.g.,<br />

amino acids or protein). Important for the establishment of forest ecosystems<br />

is thus, the capability of ectomycorrhizal fungi to exploit (in collaboration<br />

with other soil organisms) organic debris (e.g., litter) as a nutrient source<br />

(Nasholm and Persson 2001).<br />

8 Utilization of Inorganic Nitrogen<br />

For a number of ectomycorrhizal fungi growth on nitrate as sole nitrogen<br />

source (France and Reid 1984; Littke et al. 1984; Plassard et al. 1986) as well<br />

as the presence of nitrate reductase activity (Wagner et al. 1989; Sarjala<br />

1990) have been shown. In saprophytic ascomycetes, the expression of<br />

nitrate reductase is repressed in the presence of a reduced nitrogen source<br />

(e.g., ammonium) and induced only the presence of nitrate. In contrast,<br />

nitrate reductase activity of the ectomycorrhizal fungus Hebeloma cylindrosporum<br />

was similar for nitrate and ammonium-fed hyphae (Scheromm et<br />

al. 1990), indicating a different type of regulation. A nitrate transporter gene<br />

has been identified so far only from H. cylindrosporum (Marmeisse, pers.<br />

comm.).<br />

Two ammonium transporter genes of H. cylindrosporum (HcAMT2 and<br />

HcAMT3) were isolated and functionally characterized in yeast (Javelle et al.<br />

2001). HcAMT2 revealed a K M value of 58 mM and HcAMT3A of 260 mM.<br />

When the fungus was grown under optimal nitrogen conditions (ammonium<br />

concentration >2 mM) the expression of both transporter genes was only<br />

barely detectable while gene expression strongly increases under nitrogen<br />

starvation. Similar results were found in Paxillus involutus, where N starvation<br />

triggered a fourfold increase in methylamine transport after 2 h incubation<br />

in nitrogen-free media (Javelle et al. 1999).<br />

In addition, one ammonium transporter gene (TbAMT1) was isolated from<br />

the ascomycete Tuber borchii (Montanini et al. 2002). Heterologous expression<br />

in yeast revealed a K M value of 2 mM. When exposed to ammonium or nitrate,<br />

the gene was expressed at a basal level while nitrogen depletion resulted in a<br />

slow and only slight increase in gene expression. This expression profile is


382<br />

Uwe Nehls<br />

quite untypical for fungi where good nitrogen nutrition usually results in a<br />

strong repression of ammonium transporter genes.<br />

9 Utilization of Organic Nitrogen<br />

Important for the establishment of forest ecosystems is the capability of ectomycorrhizal<br />

fungi to exploit (in collaboration with other soil organisms)<br />

organic debris (e.g., litter) as a nutrient source (Nasholm and Persson 2001).<br />

10 Proteolytic Activities of Ectomycorrhizal Fungi<br />

Ericoid fungi (Bajwa et al. 1985; Leake and Read 1990), but also some ectomycorrhizal<br />

fungi (Abuzinadah and Read 1986; El-Badaoui and Botton 1989; Zhu<br />

1990; Spägele 1992; Zhu et al. 1994; Bending and Read 1996) are able to utilize<br />

protein not only as a nitrogen, but also as a carbon source (for a review, see<br />

Smith and Read 1997).<br />

Two proteins with proteolytic activities and molecular masses of about<br />

45 kDa (AmProt1) and 100 kDa (AmProt2) are excreted by A. muscaria (Nehls<br />

et al. 2001b). AmProt1 was mainly released at pH-values up to pH 5.4 and<br />

revealed a narrow pH-optimum around 3.0. It resembles thus, proteases<br />

released by H. crustuliniforme (Zhu 1990) and the ericoid fungus Hymenoscyphus<br />

ericea (Leake and Read 1990). AmProt2 was only excreted at pH-values<br />

between 5.4 and 6.3 and reveals a broad pH-optimum between 3 and 6. A.<br />

muscaria is mainly growing in the litter layer of both acidic and less acidic<br />

forest soils. Since forest litter layers are, in addition to fungi, intensively colonized<br />

by biofilm-forming bacteria (Berg et al. 1998), where the microenvironment<br />

is adapted to bacterial growth (e.g., pH 5–6; Fletcher 1996), expression<br />

of a protease that is active at a less acidic pH would favor the mobilization of<br />

bacteria-derived proteins by ectomycorrhizal fungi.<br />

A cDNA presumably encoding AmProt1 was identified in an EST project<br />

(Nehls et al. 2001b). AmProt1 was not only regulated by the external pH, but<br />

also by carbon as well as nitrogen availability. Nitrogen starvation alone<br />

increased AmProt1 expression by a factor of 3 to 4. However, the absence of a<br />

carbon source increased the transcript level of the gene by a factor of approximately<br />

12, independent of the presence or absence of nitrogen. The expression<br />

of AmProt1 reflects thus the nutritional status of fungal hyphae with<br />

respect to carbon (major regulatory effect) and nitrogen (minor regulatory<br />

effect).


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 383<br />

11 Uptake of Amino Acids<br />

Amino acids (as a result of protein degradation) are frequently found in forest<br />

soils and are thus of great importance for nitrogen nutrition. The ability to<br />

take up amino acids with high efficiency has been frequently shown for ectomycorrhizal<br />

fungi (Abuzinadah and Read 1988; Chalot et al. 1995, 1996; Wallenda<br />

and Read 1999).<br />

Fungal amino acid importer genes have been isolated to date from A. muscaria<br />

(AmAAP1; Nehls et al. 1999b) and H. cylindrosporum (Wipf et al. 2002).<br />

As determined by heterologous expression in yeast, these genes encode high<br />

affinity H + /amino acid symporter with a broad amino acid spectrum.<br />

AmAAP1 has a higher affinity to basic and aromatic amino acids compared to<br />

acidic or neutral amino acids. These differences in affinity might reflect the<br />

fact that basic amino acids are present in soil in significantly lower concentrations<br />

(8–30 mM) than neutral amino acids (70–80 mM; Scheller 1996).<br />

In contrast to AmProt1 (Nehls et al. 2001b, see above), carbon catabolite<br />

repression is not involved in regulation of AmAAP1 expression (Nehls et al.<br />

1999b). This is in agreement with results obtained for the ectomycorrhizal<br />

fungus P. involutus (Chalot et al. 1995).<br />

Good nitrogen support of fungal hyphae by amino acids as well as ammonium<br />

(not imported by AmAAP1) resulted in a low, constitutive AmAAP1<br />

expression (Nehls et al. 1999b). In contrast, AmAAP1 expression increased<br />

tenfold at low external nitrogen concentrations. It could thus be concluded<br />

that AmAAP1 expression is regulated by the endogenous nitrogen status of<br />

fungal cells, and not by the nitrogen source.<br />

As shown for a yeast mutant lacking arginine uptake activity, the reduced<br />

re-import capacity for this amino acid resulted in a net arginine loss of the<br />

cells (Grenson 1973). The strongly enhanced expression of AmAAP1 under<br />

nitrogen starvation conditions (even in the absence of amino acids) could<br />

also indicate that AmAAP1, in addition to amino acid uptake for nitrogen<br />

nutrition, might be important in the reduction of amino acid loss by hyphal<br />

leakage.<br />

12 Regulation of Fungal Nitrogen Export in Mycorrhizas by<br />

the Nitrogen-Status of Hyphae<br />

The nitrogen-dependent expression profile of nitrogen importer genes of<br />

ectomycorrhizal fungi (A. muscaria: Nehls et. al. 1999; H. cylindrosporum:<br />

Javelle et al. 2001; Wipf et al. 2002) resembles that of ascomycetes (yeast: Ter<br />

Schure et. al. 1998; Aspergillus: Sophianopoulou and Diallinas 1995). Here,<br />

nitrogen importer gene expression is regulated at the transcriptional level by<br />

two mechanisms: nitrogen repression in the presence of a good nitrogen<br />

source (ammonium or glutamine) and the induction of genes necessary for


384<br />

Uwe Nehls<br />

the utilization of alternative nitrogen sources under nitrogen limitation (e.g.,<br />

Tazebay 1997). Nitrogen-dependent gene repression is presumably regulated<br />

by the internal nitrogen status of cells, and not the external nitrogen availability.<br />

Either, the intracellular ammonium concentration (Ter Schure et. al.<br />

2000) and/or the activity of the glutamine synthetase (Sophianopoulou and<br />

Diallinas 1995) are supposed to sense the endogenous nitrogen status.<br />

In ectomycorrhizal fungi, nitrogen importer gene expression is presumably<br />

also regulated by the internal nitrogen status of the hyphae (Nehls et. al. 1999;<br />

Javelle et al. 2001; Wipf et al. 2002). This could indicate how nitrogen uptake by<br />

soil-growing hyphae and nitrogen export by hyphae of the Hartig net might<br />

be managed (Fig. 3). Since the nitrogen content of forest soil is quite low and<br />

part of the nitrogen is transported to other parts of the growing fungal colony<br />

(e.g., mycorrhizas), soil-growing hyphae are presumably nitrogen-limited,<br />

resulting in a low endogenous nitrogen status and a strong expression of<br />

nitrogen importer genes. On the other hand, mycorrhizas are well supplied<br />

with nitrogen by soil-growing hyphae, thus revealing a high nitrogen status<br />

and a strongly reduced nitrogen importer gene expression. This nitrogen-<br />

Fig. 3. Regulation of fungal nitrogen uptake from soil and nitrogen excretion at the<br />

<strong>plant</strong>/fungus interface: a model. Nitrogen export to other parts of the fungal colony<br />

together with a low nitrogen content in soil results in a low endogenous nitrogen state in<br />

soil growing hyphae. In consequence, nitrogen importer genes are highly expressed and<br />

nitrogen uptake capacity is high. Nitrogen import from soil growing hyphae causes a<br />

high endogenous nitrogen status in hyphae of the Hartig net. This results in a repression<br />

of nitrogen importer gene expression and, together with posttranslational inactivation<br />

processes, in a low nitrogen uptake capacity. Together with export mechanisms, this<br />

leads to a net export of nitrogen at the <strong>plant</strong>/fungus interface


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 385<br />

dependent repression of amino acid transporter gene expression (indicated<br />

by AmAAP1), together with posttranslational events (e.g., increased degradation<br />

of plasma membrane transport proteins) that are described for yeasts<br />

(Springael and Andre 1998), could thus result in a highly reduced fungal<br />

capacity for re-uptake of amino acids at the <strong>plant</strong>/fungus interface. In combination<br />

with efflux mechanisms (e.g., nitrogen leakage), this would thus result<br />

in a net export of nitrogen.<br />

13 Carbohydrate and Nitrogen-Dependent Regulation of<br />

Fungal Gene Expression<br />

Carbohydrates as well as nitrogen are essential components of biological molecules<br />

(e.g., amino acids or nucleotides), and obviously have a great impact on<br />

fungal gene expression (e.g., Gonzales et al. 1997).<br />

With regard to carbon and nitrogen nutrition, four different patterns of<br />

regulation have been observed in A. muscaria. The amino acid importer gene<br />

AmAAP1 is only regulated by nitrogen nutrition, while the hexose transporters<br />

AmMst1 and AmMst2 (Nehls et al. 1998) are only regulated by carbohydrate<br />

nutrition. On the other hand, AmProt1 (protease; Nehls et al. 2001b)<br />

and AmTPS1 (trehalose-6-phosphate synthase) are regulated by both nitrogen<br />

as well as carbon nutrition. Nevertheless, the impact of carbon and nitrogen<br />

nutrition differs significantly for both genes. While AmProt1 is mainly<br />

regulated by nitrogen, AmTPS1 is mainly regulated by carbon availability.<br />

Comparable gene expression patterns have been described for fungi (Gonzales<br />

et al. 1997) as well as <strong>plant</strong>s (Coruzzi and Zhou 2001), revealing a universal<br />

and phylogenetically old regulation strategy.<br />

14 Conclusions<br />

Since large EST projects of ectomycorrhizal model systems are currently<br />

under progress (Tagu and Martin 1995; Johansson et al. 2000; Voiblet et al.<br />

2001; Wipf et al. 2003), macro- and micro-array hybridization will enable an<br />

overview of the general impact of carbon and nitrogen nutrition on gene<br />

expression for different ectomycorrhizal fungi.<br />

Present data suggest that carbon- and nitrogen-dependent gene repression<br />

in ectomycorrhizal fungi is presumably similar to that of saprophytic<br />

ascomycetes (yeast, Neurospora). Ascomycotic model organisms could thus<br />

help to develop working models for ectomycorrhizal function (e.g., nitrogen<br />

uptake from soil and release at the <strong>plant</strong>/fungus interface; see Fig. 3) that<br />

could be investigated in turn in an ectomycorrhizal model system. In addition,<br />

differences, e.g., in carbon-dependent gene regulation for an ectomycorrhizal<br />

fungus (A. muscaria) and saprophytic ascomycetes (yeast, Neurospora)


386<br />

Uwe Nehls<br />

have been described. They might thus reveal adaptation processes that are<br />

necessary for ectomycorrhizal function.<br />

Acknowledgements I am indebted to Magret Ecke and Andrea Bock for excellent technical<br />

assistance and to Dr. Mika Tarkka and Dr. Rüdiger Hampp for critical reading of the<br />

manuscript. This work was supported by the Deutsche Forschungsgemeinschaft (DFG-<br />

Schwerpunkt Mykorrhiza).<br />

References and Selected Reading<br />

Abuzinadah RA, Read DJ (1986) The role of proteins in the nitrogen nutrition of ectomycorrhizal<br />

<strong>plant</strong>s. I. Utilization of peptides and proteins by ectomycorrhizal fungi.<br />

New Phytol 103:481–493<br />

Abuzinadah RA, Read DJ (1988) Amino acids as nitrogen sources for ectomycorrhizal<br />

fungi: utilisation of individual amino acids. Transact Br Mycol Soc 91:473–479<br />

Bajwa R, Abuarghub S, Read DJ (1985) The biology of mycorrhiza in the Ericaceae. X.<br />

The utilization of proteins and the production of proteolytic enzymes by the mycorrhizal<br />

endophyte and by mycorrhizal <strong>plant</strong>s. New Phytol 101:469–486<br />

Bending GD, Read DJ (1996) Nitrogen mobilization from protein-polyphenol complex<br />

by ericoid and ectomycorrhizal fungi. Soil Biol BioChem 28:1603–1612<br />

Berg MP, Kniese JP,Verhoef HA (1998) Dynamics and stratification of bacteria and fungi<br />

in the organic layers of a Scots pine forest soil. Biol Fertil Soils 26:313–322<br />

Berredjem A, Garnier A, Putra DP, Botton B (1998) Effect of nitrogen and carbon sources<br />

on growth and activities of NAD and NADP dependent isocitrate dehydrogenases of<br />

Laccaria bicolor. Mycol Res 4:427–434<br />

Cairney JWG, Burke RM (1996) Physiological heterogeneity within fungal mycelia: an<br />

important concept for a functional understanding of the ectomycorrhizal symbiosis.<br />

New Phytol 134:685–695<br />

Cao W, Crawford DL (1993) Carbon nutrition and hydrolytic and cellulolytic activities in<br />

the ectomycorrhizal fungus Pisolithus tinctorius. Can J Microbiol 39:529–535<br />

Celenza JL, Marshall-Carlson L, Carlson M (1988) The yeast SNF3 gene encodes a glucose<br />

transporter homologous to the mammalian protein. Proc Natl Acad Sci USA<br />

85:2130–2134<br />

Chakravarty P, Unestam T (1987) Differential influence of ectomycorrhizae on <strong>plant</strong><br />

growth and disease resistance of Pinus sylvestris seedlings. J Phytopathol 120:104–120<br />

Chalot M, Brun A (1998) Physiology of organic nitrogen acquisition by ectomycorrhizal<br />

fungi and ectomycorrhizas. FEMS Microbiol Rev 22:21–44<br />

Chalot M, Kytöviita MM, Brun A, Finlay RD, Söderström B (1995) Factors affecting<br />

amino acid uptake by the ectomycorrhizal fungus Paxillus involutus. Mycol Res<br />

99:1131–1138<br />

Chalot M, Brun A, Botton B, Soderstrom B (1996) Kinetics, energetics and specificity of a<br />

general amino acid transporter from the ectomycorrhizal fungus Paxillus involutus.<br />

Microbiol 142:1749–1756<br />

Chen XY, Hampp R (1993) Sugar uptake by protoplasts of the ectomycorrhizal fungus,<br />

Amanita muscaria (L. ex fr.) Hooker. New Phytol 125:601–608<br />

Coruzzi GM, Zhou L (2001) Carbon and nitrogen sensing and signaling in <strong>plant</strong>s: emerging<br />

‘matrix effects’. Curr Opin Plant Biol 4:247–253<br />

d’Enfert C (1997) Fungal spore germination: insights from the molecular genetics of<br />

Aspergillus nidulans and Neurospora crassa. Fungal Genet Biol 21:163–172


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 387<br />

El-Badaoui K, Botton B (1989) Production and characterization of exocellular proteases<br />

in ectomycorrhizal fungi. Annales des Sciences Foréstières 46:728–730<br />

Elbein A (1974) The metabolism of a,a-trehalose. Adv Carb Chem BioChem 30:227–256<br />

Felenbok B, Kelley JM (1996) Regulation of carbon metabolism in mycelial fungi. In:<br />

Brambl R, Marzluf GA (eds) The Mycota III. Biochemistry and molecular biology.<br />

Springer, Berlin Heidelberg New York, pp 369–380<br />

Finlay R, Ek H, Odham G, Söderström B (1988) Mycelial uptake, translocation and assimilation<br />

of nitrogen from 15 N labelled ammonium by Pinus sylvestris <strong>plant</strong>s infected<br />

with four different ectomycorrhizal fungi. New Phytol 110:59–66<br />

Finlay RD, Frostegärd Ä, Sonnerfeldt A-M (1992) Utilisation of organic and inorganic<br />

nitrogen sources by ectomycorrhizal fungi in pure culture and in symbiosis with<br />

Pinus contorta Dougl. ex Loud. New Phytol 120:105–111<br />

Fletcher M (1996) Bacterial adhesion: Molecular and ecological diversity. Wiley-Liss,<br />

New York<br />

France RC, Reid CPP (1983) Interactions of nitrogen and carbon in the physiology of<br />

ectomycorrhizas. Can J Bot 61:964–984<br />

France RC, Reid CPP (1984) Pure culture growth of ectomycorrhizal fungi on inorganic<br />

nitrogen sources. Microbiol Ecol 10:187–195<br />

Garbaye J (1991) Biological interactions in the mycorrhizosphere. Experientia 47:<br />

370–375<br />

Genet P, Prevost A, Pargney JC (2000) Seasonal variations of symbiotic ultrastructure<br />

and relationships of two natural ectomycorrhizae of beech (Fagus sylvatica/Lactarius<br />

blennius var. viridis and Fagus sylvatica/Lactarius subdulcis). Trees 14:465–474<br />

Gessler A, Schneider S, von Sengbusch D, Weber P, Hanemann U, Huber C, Rothe A,<br />

Kreutzer K, Rennenberg H (1998) Field and laboratory experiments on net uptake of<br />

nitrate and ammonium by the roots of spruce (Picea abies) and beech (Fagus sylvatica)<br />

trees. New Phytol 138:275–285<br />

Gonzalez R, Gavrias V, Gomez D, Scazzocchio C, Cubero B (1997) The integration of<br />

nitrogen and carbon catabolite repression in Aspergillus nidulans requires the GATA<br />

factor AreA and an additional positive-acting element, ADA. EMBO J 16:2937–2944<br />

Grenson M (1973) Specificity and regulation of the uptake and retention of amino acids<br />

and pyrimidines in yeast. In: Vanek Z, Hostalek Z, Culdin J (eds) Genetics of industrial<br />

microorganisms. Academica, Prague, pp 179–193<br />

Hampp R, Schaeffer C (1999) Mycorrhiza-carbohydrate and energy metabolism. In:<br />

Varma A, Hock B (eds) Mycorrhiza: structure, function, molecular biology and<br />

biotechnology. Springer, Berlin Heidelberg New York, pp 273–303<br />

Haselwandter K, Bobleter O, Read DJ (1990) Degradation of carbon-14 labelled lignin<br />

and dehydropolymer of coniferyl alcohol by ericoid and ectomycorrhizal fungi. Arch<br />

Microbiol 153:352–354<br />

Hoffmann E, Wallenda T, Schaeffer C, Hampp R (1997) Cyclic AMP, a possible regulator<br />

of glycolysis in the ectomycorrhizal fungus Amanita muscaria. New Phytol 137:351–<br />

356<br />

Javelle A, Chalot M, Soderstrom B, Botton B (1999) Ammonium and methylamine transport<br />

by the ectomycorrhizal fungus Paxillus involutus and ectomycorrhizas. FEMS<br />

Microbiol Ecol 30:355–366<br />

Javelle A, Rodriguez-Pastrana BR, Jacob C, Botton B, Brun A,Andre B, Marini AM, Chalot<br />

M (2001) Molecular characterization of two ammonium transporters from the ectomycorrhizal<br />

fungus Hebeloma cylindrosporum. FEBS Lett 505:393–398<br />

Jennings DJ (1995) The physiology of fungal nutrition. Cambridge University Press,<br />

Cambridge<br />

Johansson A, Le Quéré A, Ahrén D, Lundeberg J, Erlandsson R, Uhlèn M, Söderström B,<br />

Tulind A (2000) Transcript profiling during ectomycorrhizal development. In: Tran-


388<br />

Uwe Nehls<br />

script profiling during ectomycorrhizal development. Philipps University of Marburg,<br />

Marburg, pp 95<br />

Johnston M (1999) Feasting, fasting and fermenting: glucose sensing in yeast and other<br />

cells. TIGS 15:29–33<br />

Jordy MN, AzemarLorentz S, Brun A, Botton B, Pargney JC (1998) Cytolocalization of<br />

glycogen, starch, and other insoluble polysaccharides during ontogeny of Paxillus<br />

involutus–Betula pendula ectomycorrhizas. New Phytol 140:331–341<br />

Keeney DR (1980) Prediction of soil nitrogen availability in forest ecosystems: a literature<br />

review. For Sci 26:159–171<br />

Kowallik W, Thiemann M, Huang Y, Mutumba G, Beermann L, Broer D, Grotjohann N<br />

(1998) Complete sequence of glycolytic enzymes in the mycorrhizal basidiomycete,<br />

Suillus bovinus. Z Naturforsch 53:818–827<br />

Leake JR, Read DJ (1990) Proteinase activity in mycorrhizal fungi. I. The effect of extracellular<br />

pH on the production and activity of proteinase by ericoid endophytes from<br />

soil of contrasted pH. New Phytol 115:243–250<br />

Leake JR, Donnelly DP, Saunders EM, Boddy L, Read DJ (2001) Rates and quantities of<br />

carbon flux to ectomycorrhizal mycelium following 14C pulse labeling of Pinus<br />

sylvestris seedlings: effects of litter patches and interaction with a wood-decomposer<br />

fungus. Tree Physiol 21:71–82<br />

Lesage P, Yang X, Carlson M (1996) Yeast SNF1 protein kinase interacts with SIP4, a C 6<br />

zink cluster transcriptional activator: a new role for SNF1 in the glucose response.<br />

Mol Cell Biol 16:1921–1928<br />

Lewis DH, Harley JL (1965) Carbohydrate physiology of mycorrhizal roots of beech. I.<br />

Identity of endogenous sugars and utilization of exogenous sugars. New Phytol<br />

64:224–237<br />

Littke WR, Bledsoe CS, Edmonds RL (1984) Nitrogen uptake and growth in vitro by<br />

Hebeloma crustuliniforme and other Pacific Northwest mycorrhizal fungi. Can J Bot<br />

62:647–652<br />

Madi L, McBridge SA, Bailey LA, Ebbole DJ (1997) Rco-3, a gene involved in glucose<br />

transport and conidiation in Neurospora crassa. Genet 146:499–508<br />

Marschner H (1995) Mineral nutrition of <strong>plant</strong>s. 2nd edn. Academic Press, London<br />

Martin F, Canet D, Marchal JP (1985) 13 C nuclear magnetic resonance study of mannitol<br />

cycle and trehalose synthesis during glucose utilization by the ectomycorrhizal<br />

ascomycete Cenococcum geophilum. Plant Physiol 77:499–502<br />

Martin F, Ramstedt M, Söderhall K (1987) Carbon and nitrogen metabolism in ectomycorrhizal<br />

fungi and ectomycorrhizas. Biochimie 69:569–581<br />

Martin F, Ramstedt M, Söderhall K, Canet D (1988) Carbohydrate and amino acid metabolism<br />

in the ectomycorrhizal ascomycete Sphaerosporella brunnea during glucose<br />

utilization a 13 C NMR study. Plant Physiol 86:935–940<br />

Martin F, Chalot M, Brun A, Lorillou S, Botton B, Dell B (1992) Spatial distribution of<br />

nitrogen assimilation pathways in ectomycorrhizas. In: Read DJ, Lewis DH, Fitter A,<br />

Alexander I (eds) Mycorrhizas in ecosystems. CAB International, Wallingford, pp<br />

311–315<br />

Martin F, Boiffin VV, Pfeffer PE (1998) Carbohydrate and amino acid metabolism in the<br />

Eucalyptus globulus–Pisolithus tinctorius ectomycorrhiza during glucose utilization.<br />

Plant Physiol 118:627–635<br />

Marx DH (1969) The influence of ectotrophic ectomycorrhizal fungi on the resistance of<br />

pine roots to pathogenic infections. I.Antagonism of mycorrhizal fungi to pathogenic<br />

fungi and soil bacteria. Phytopathology 59:153–163<br />

Melin E, Nilsson H (1952) Transport of labelled nitrogen from an ammonium source to<br />

pine seedlings through mycorrhizal mycelium. Svensk Bot Tidskr 46:281–285<br />

Mellor R (1992) Is trehalose a symbiotic determinant in symbioses between higher<br />

<strong>plant</strong>s and microorganisms? Symbiosis 12:113–129


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 389<br />

Montanini B, Moretto N, Soragni E, Percudani R, Ottonello S (2002) A high-affinity<br />

ammonium transporter from the mycorrhizal ascomycete Tuber borchii. Fungal<br />

Genet Biol 36:22–34<br />

Müller J, Xie ZP, Staehelin C, Mellor RB, Boller T, Wiemken A (1994) Trehalose and trehalase<br />

in root nodules from various legumes. Physiolog Plant 90:86–92<br />

Nasholm T, Persson J (2001) Plant acquisition of organic nitrogen in boreal forests. Physiol<br />

Plant 111:419–426<br />

Nehls U, Wiese J, Guttenberger M, Hampp R (1998) Carbon allocation in ectomycorrhizas:<br />

identification and expression analysis of an Amanita muscaria monosaccharide<br />

transporter. Mol Plant Microbiol Interact 11:167–176<br />

Nehls U, Ecke M, Hampp R (1999a) Sugar- and nitrogen-dependent regulation of an<br />

Amanita muscaria phenylalanine ammonium lyase gene. J Bacteriol 181:1931–1933<br />

Nehls U, Kleber R, Wiese J, Hampp R (1999b) Isolation and characterization of a general<br />

amino acid permease from the ectomycorrhizal fungus Amanita muscaria.New Phytol<br />

144:343–349<br />

Nehls U, Bock A, Ecke M, Hampp R (2001a) Differential expression of hexose-regulated<br />

fungal genes within Amanita muscaria/Populus tremula x tremuloides ectomycorrhizas.<br />

New Phytol 150:583–589<br />

Nehls U, Bock A, Einig W, Hampp R (2001b) Excretion of two proteases by the ectomycorrhizal<br />

fungus Amanita muscaria. Plant Cell Environ 24:741–747<br />

Nehls U, Mikolajewski S, Magel E, Hampp R (2001 c) The role of carbohydrates in ectomycorrhizal<br />

functioning: gene expression and metabolic control. New Phytol<br />

150:533–541<br />

Norkrans B (1950) Studies in growth and cellolytic enzymes of Tricholoma.Symb Bot<br />

Upsal 11:1–126<br />

Özcan S, Johnston M (1999) Function and regulation of yeast hexose transporters.<br />

Microbiol Mol Biol Rev 63:554–569<br />

Özcan S, Dover J, Rosenwald AG, Woelfl S, Johnston M (1996) Two glucose transporters<br />

in Saccharomyces cerevisiae are glucose sensors that generate a signal for induction of<br />

gene expression. Proc Natl Acad Sci USA 93:12428–12432<br />

Palmer JG, Hacskaylo E (1970) Ectomycorrhizal fungi in pure culture. I. Growth on single<br />

carbon sources. Physiol Plant 23:1187–1197<br />

Plassard C, Scheromm P, Llamas H (1986) Nitrate assimilation by maritime pine and<br />

ectomycorrhizal fungi in pure culture. In: Gianinazzi-Pearson V, Gianinazzi S (eds)<br />

Physiological and genetical aspects of mycorrhizae. INRA, Paris, pp 383–388<br />

RadisBaptista G,Valdivia DNU,AbrahaoNeto J (1998) Fructose 2,6-bisphosphate biosynthesis<br />

and regulation of carbohydrate metabolism in Aspergillus oryzae. Can J Microbiol<br />

44:6–11<br />

Read DJ (1991) Mycorrhizas in ecosystems. Experientia 47:376–391<br />

Ronne H (1995) Glucose repression in fungi. Trends Genet 11:12–17<br />

Salzer P, Hager A (1991) Sucrose utilization of the ectomycorrhizal fungi Amanita muscaria<br />

and Hebeloma crustuliniforme depends on the cell wall-bound invertase activity<br />

of their host Picea abies. Bot Acta 104:439–445<br />

Sarjala T (1990) Effect of nitrate and ammonium concentration on nitrate reductase<br />

activity in five species of mycorrhizal fungi. Physiolog Plant 79:65–70<br />

Schaeffer C,Wallenda T, Guttenberger M, Hampp R (1995) Acid invertase in mycorrhizal<br />

and non-mycorrhizal roots of Norway spruce (Picea abies [L.] Karst.) seedlings. New<br />

Phytol 129:417–424<br />

Schaeffer C, Johann P, Nehls U, Hampp R (1996) Evidence for an up-regulation of the<br />

host and a down-regulation of the fungal phosphofructokinase activity in ectomycorrhizas<br />

of Norway spruce and fly agaric. New Phytol 134:697–702


390<br />

Uwe Nehls<br />

Scheller E (1996) Aminosäuregehalte von Ap- und Ah-Horizonten verschiedener Böden<br />

und deren Huminsäuren- und Fulvosäuren-Fraktion. Mitt Dtsch Bodenkd Ges<br />

81:201–204<br />

Scheromm PS, Plassard C, Salsac L (1990) Regulation of nitrate reductase in the ectomycorrhizal<br />

basidiomycete, Hebeloma cylindrosporum Romagn., cultured on nitrate or<br />

ammonium. New Phytol 114:441–448<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis. Academic Press, London<br />

Sophianopoulou V, Diallinas G (1995) Amino acid transporters of lower eukaryotes: regulation,<br />

structure and topogenesis. FEMS Microbiol Rev 16:53–75<br />

Spägele S (1992) Charakterisierung der intra- und extrazellulären Proteasenaktivitäten<br />

des Fliegenpilzes (Amanita muscaria [L. ex Fr.] Hooker). Eberhard-Karls-Universität,<br />

Tübingen<br />

Springael JY, Andre B (1998) Nitrogen-regulated ubiquitination of the Gapl permease of<br />

Saccharomyces cerevisiae. Mol Biol Cell 9:1253–1263<br />

Stülten C, Kong FX, Hampp R (1995) Isolation and regeneration of protoplasts from the<br />

ectomycorrhizal ascomycete Cenococcum geophilum Fr. Mycorrhiza 5:259–266<br />

Taber WA, Taber RA (1987) Carbon nutrition and respiration of Pisolithus tinctorius.<br />

Transact Brit Mycol Soc 89:13–26<br />

Tagu D, Martin F (1995) Expressed sequence tags of randomly selected cDNA clones<br />

from Eucalyptus globulus–Pisolithus tinctorius ectomycorrhiza. Mol Plant Microbiol<br />

Interact 8:781–783<br />

Tagu D, Lapeyrie F, Ditengou F, Lagrangem H, Laurent P, Missoum N, Nehls U, Martin F<br />

(2000) Molecular aspects of ectomycorrhiza development. In: Poldila G, Douds Jr DD<br />

(eds) Current advances in mycorrhizal research. Am Phytopathol Soc, pp 69–90<br />

Tazebay UH, Sophianopoulou V, Scazzocchio C, Diallinas G (1997) The gene encoding<br />

the major proline transporter of Aspergillus nidulans is upregulated during conidiospore<br />

germination and in response to proline induction and amino acid starvation.<br />

Mol Microbiol 24:105–117<br />

Ter Schure EG, Sillje HHW,Vermeulen EE, Kalhorn JW,Verkleij AJ, Boonstra J,Verrips CT<br />

(1998) Repression of nitrogen catabolic genes by ammonia and glutamine in nitrogen-limited<br />

continuous cultures of Saccharomyces cerevisiae. Microbiol Reading<br />

144:1451–1462<br />

Ter Schure EG, van Riel NA,Verrips CT (2000) The role of ammonia metabolism in nitrogen<br />

catabolite repression in Saccharomyces cerevisiae. FEMS Microbiol Rev 24:67–83<br />

Thevelein JM (1991) Fermentable sugars and intracellular acidification as specific activators<br />

of the RAS-adenylate cyclase signalling pathway in yeast: the relationship to<br />

nutrient-induced cell cycle control. Mol Microbiol 5:1302–1307<br />

Timonen S, Sen R (1998) Heterogeneity of fungal and <strong>plant</strong> enzyme expression in intact<br />

Scots pine-Suillus bovinus and -Paxillus involutus mycorrhizospheres developed in<br />

natural forest humus. New Phytol 138:355–366<br />

Trojanowski J, Haider K, Huttermann A (1984) Decomposition of 14 C-labelled lignin,<br />

holocellulose and lignocellulose by mycorrhizal fungi. Arch Microbiol 139:202–206<br />

Voiblet C, Duplessis S, Encelot N, Martin F (2001) Identification of symbiosis-regulated<br />

genes in Eucalyptus globulus–Pisolithus tinctorius ectomycorrhiza by differential<br />

hybridization of arrayed cDNAs. Plant J 25:181–191<br />

Wagner F, Gay G, Debaud JC (1989) Genetic variation of nitrate reductase activity in<br />

monokaryotic and dikaryotic populations of the ectomycorrhizal fungus, Hebeloma<br />

cylindrosporum Romagnesi. New Phytol 113:259–264<br />

Wallenda T (1996) Untersuchungen zur Physiologie der Pilzpartner von Ektomykorrhizen<br />

der Fichte (Picea abies [L.] Karst.). Eberhard-Karls-Universität, Tübingen<br />

Wallenda T, Read DJ (1999) Kinetics of amino acid uptake by ectomycorrhizal roots.<br />

Plant Cell Environ 22:179–187


21 Carbohydrates and Nitrogen: Nutrients and Signals in Ectomycorrhiza 391<br />

Wiese J, Kleber R, Hampp R, Nehls U (2000) Functional characterization of the Amanita<br />

muscaria monosaccharide transporter AmMst1. Plant Biol 2:1–5<br />

Wipf D, Benjdia M, Tegeder M, Frommer WB (2002) Characterization of a general amino<br />

acid permease from Hebeloma cylindrosporum. FEBS Lett 528:119–124<br />

Wipf D, Benjdia M, Rikirsch E, Zimmermann S, Tegeder M, Frommer WB (2003) An<br />

expression cDNA library for suppression cloning in yeast mutants, complementation<br />

of a yeast his4 mutant, and EST analysis from the symbiotic basidiomycete Hebeloma<br />

cylindrosporum. Genome 46(2):177–181<br />

Wisser G (2000) Isolation und Charakterisierung von Trehalasen aus Amanita muscaria<br />

[L. ex Fr.] Hooker, einem Ektomykorrhizapilz. PhD-thesis, Eberhard-Karls-Universitaet<br />

Tuebingen, Germany<br />

Wisser G, Guttenberger M, Hampp R, Nehls U (2000) Identification and characterization<br />

of an extracellular acid trehalase from the ectomycorrhizal fungus Amanita muscaria.<br />

New Phytol 146:169–175<br />

Zhu H (1990) Purification and characterization of an extracellular acid proteinase from<br />

the ectomycorrhizal fungus Hebeloma crustuliniforme. Appl Environ Microbiol<br />

56:837–843<br />

Zhu H, Dancik BP, Higginbotham KO (1994) Regulation of extracellular proteinase production<br />

in an ectomycorrhizal fungus Hebeloma crustuliniforme. Mycologia 86:227–<br />

234


22 Nitrogen Transport and Metabolism in<br />

Mycorrhizal Fungi and Mycorrhizas<br />

Arnaud Javelle, Michel Chalot, Annick Brun<br />

and Bernard Botton<br />

1 Introduction<br />

1.1 Ecological Significance of Ectomycorrhizas<br />

Unlike most other organisms, <strong>plant</strong>s and fungi are restricted to their habitats,<br />

creating potential problems when nutritional conditions become limited. To<br />

cope with nutrient deficiencies, they have developed a variety of adaptations<br />

that enable them to respond to their internal nutritional status as well as to<br />

the external availability of nutrients.A strategy for <strong>plant</strong>s is mycorrhizal association,<br />

in which expanding mycorrhizal mycelia that grow outward from the<br />

mantle into the surrounding soil is a very efficient nitrogen scavenger owing<br />

to (1) its capacity to explore a larger soil volume than roots alone (Smith and<br />

Read 1997), (2) its ability to provide access to nitrogenous reserves contained<br />

in organic horizons (Chalot and Brun 1998) and (3) its greater capacity for<br />

uptake of nitrogenous compounds (Javelle et al. 1999; Wallanda and Read<br />

1999).<br />

This interconnected network of hyphae (or specialized aggregates, i.e., rhizomorphs)<br />

forms a supracellular compartment for the transport of nutrients<br />

from sites of nutrient capture to sites of nutrient utilization and transfer. It<br />

has been estimated that the external mycelium makes, by far, the greatest contribution<br />

to the overall potential absorbing <strong>surface</strong> area of pine seedlings<br />

inoculated with Pisolithus tinctorius or Cenococcum geophilum (Rousseau et<br />

al. 1994). Fungal hyphae have a number of advantages compared with roots;<br />

(1) hyphae have a low ratio of biomass to absorptive <strong>surface</strong> area and can easily<br />

be regenerated (Harley 1989; Rousseau et al. 1994), (2) they have been<br />

shown to rapidly colonize nutrient-rich sites (Carleton and Read 1991; Bending<br />

and Read 1995) and (3) because of their small diameter, they can exploit<br />

small pores inaccessible to roots. The symbiotic association of higher <strong>plant</strong>s<br />

with mycorrhizal fungi is considered to have been responsible for the colonization<br />

of land by <strong>plant</strong>s (Taylor and Osborn 1995).<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


394<br />

A. Javelle et al.<br />

1.2 Nitrogen Uptake and Translocation by Ectomycorrhizas<br />

Nitrogen plays a critical role in <strong>plant</strong> and microorganism biochemistry, being<br />

needed for the synthesis of many compounds, including amino acids, purines,<br />

pyrimidines, some carbohydrates and lipids, enzyme cofactors and proteins,<br />

all of which are essential for growth processes. Ammonium and nitrate are<br />

believed to be the principal sources of nitrogen in forest soil. When the two<br />

compounds are supplied to <strong>plant</strong>s at similar concentrations, ammonium is<br />

generally taken up more rapidly than nitrate (Marschner et al. 1991; Kronzucker<br />

et al. 1996; Howitt and Udvardi 2000).Attention has also been paid to<br />

the utilization of organic nitrogen forms from more complex substrates (Smith<br />

and Read 1997; Perez-Moreno and Read 2000),and to the direct mobilization of<br />

nutrients from minerals (for a review, see Landeweert et al. 2001). The two<br />

processes involved in ammonium assimilation,namely transport and metabolism,<br />

have been studied in various ectomycorrhizal models. Increases in nitrogen<br />

content of ectomycorrhizal <strong>plant</strong>s,often connected with a growth increase,<br />

are well documented (Smith and Read 1997). Studies have demonstrated that<br />

the ectomycorrhizal partner plays an integral role in ammonium metabolism<br />

in trees (Chalot et al. 1991; Botton and Chalot 1995; Plassard et al. 1997).<br />

Nutrient uptake and transport by extraradical mycelium is suggested to be<br />

an important factor for improved nutrient acquisition. The contribution of<br />

extraradical mycelium to N nutrition of mycorrhizal Norway spruce was<br />

investigated. The addition of N to the hyphal compartment markedly<br />

increased dry weight, N concentration and N content in mycorrhizal <strong>plant</strong>s.<br />

Calculating the uptake, based on the difference in input and output of nutrients<br />

in solution, confirmed a hyphal contribution of 73 % to total N uptake in<br />

Picea abies seedlings under nitrogen and phosphorus starvation (Brandes et<br />

al. 1998). In further studies, Jentschke et al. (2001) have demonstrated in Picea<br />

abies/Paxillus involutus ectomycorrhizas that hyphal N uptake (NH 4 + +NO3 – )<br />

contributed 17 % to total N uptake in mycorrhizal seedlings. Moreover,<br />

ammonium is the major source of mineral nitrogen in forest soils (Marschner<br />

and Dell 1994), and consequently, ammonium assimilation by extraradical<br />

mycelium plays a crucial role for nitrogen transfer in ectomycorrhizal symbiosis.<br />

Melin and Nilsson (1952) showed that the mycelia phase of Suillus variegatus<br />

was capable of absorption and translocation to Pinus mycorrhizal<br />

seedlings of nitrogen from a labelled ammonium source. Disrupting the<br />

external mycelium from ectomycorrhizas greatly decreased [ 15 N]ammonium<br />

uptake by birch seedlings (Javelle et al. 1999).Ammonium is incorporated into<br />

a range of amino acids and these accumulate in fungal mycelium at considerable<br />

distances from <strong>plant</strong> roots (Finlay et al. 1988). Therefore, external hyphae<br />

can be considered as the absorbing structure of ectomycorrhizal roots. These<br />

results confirmed the function of extraradical mycelium in translocating N<br />

from sources to roots and that it can, therefore, be considered as a nutrient<br />

channel (Smith and Read 1997).


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 395<br />

2 Nitrate and Nitrite Transport<br />

2.1 Uptake Kinetics<br />

Nitrate uptake rates were estimated in a few ectomycorrhizal fungi and ectomycorrhizas.<br />

In the basidiomycete Rhizopogon roseolus, NO 3 – uptake measured<br />

after incubation of mycelia in 0.05 mM nitrate occurred at the same rate<br />

in the absence or presence of NO 3 – in the culture medium, suggesting that no<br />

inducible nitrate transporter exists in this species (Gobert and Plassard 2002).<br />

These results are in agreement with those of Jargeat (1999) who observed that<br />

the mRNA of a high-affinity transport system in the ectomycorrhizal basidiomycete<br />

Hebeloma cylindrosporum, was found in mycelia grown in N-free<br />

medium or in media containing low nitrate concentrations. Km estimates,<br />

around 12 mM in Rhizopogon roseolus (Gobert and Plassard 2002), and 67 mM<br />

in Hebeloma cylindrosporum (Plassard et al. 1994) are close to the Michaelis<br />

constants found in nonmycorrhizal fungi, with values of 23 mM in Aspergillus<br />

nidulans (Zhou et al. 2000) and 25 mM in Neurospora crassa (Blatt et al. 1997).<br />

In nonmycorrhizal Pinus pinaster roots, rates of NO 3 – uptake were<br />

enhanced by exposure to external nitrate, as usually found in higher <strong>plant</strong><br />

species. In the association Pinus pinaster/Rhizopogon roseolus, NO 3 – uptake<br />

was not modified by external nitrate, but was constantly higher than that<br />

measured in nonmycorrhizal roots (Gobert and Plassard 2002). According to<br />

these authors, the fungal uptake of nitrate may confer to the mycorrhiza a<br />

greater ability to use low and fluctuating concentrations of nitrate in the soil.<br />

However, in Fagus-Laccaria mycorrhizas, mycorrhization led to reduced rates<br />

of NO 3 – net uptake, this effect being caused by reduced influx, plus enhanced<br />

efflux of NO 3 – as compared with nonmycorrhizal beech roots (Kreuzwieser et<br />

al. 2000).<br />

2.2 Characterization of Nitrate and Nitrite Transporters<br />

Kinetically, two groups of nitrate transporters have been characterized: one<br />

with a high affinity, Km in the mM nitrate range, found in filamentous fungi,<br />

yeasts, algae and <strong>plant</strong>s (Crawford and Glass 1998; Forde 2000), and one low<br />

affinity group, Km in the mM nitrate range, found mainly in <strong>plant</strong>s, although<br />

there is indirect evidence of its presence in yeasts and algae (Machin et al.<br />

2000; Navarro et al. 2000).<br />

Aspergillus nidulans possesses two high-affinity nitrate transporters,<br />

encoded by the nrtA (formerly designated crnA) and the nrtB genes (Unkles<br />

et al. 1991; 2001). Whereas mutants expressing either gene grew normally on<br />

nitrate as sole nitrogen source, the double mutant was unable to grow even if<br />

the nitrate concentration was increased to 200 mM. This indicates that NRTA<br />

and NRTB are the only nitrate transporters in Aspergillus nidulans. Both


396<br />

A. Javelle et al.<br />

genes were regulated identically under an extensive range of conditions; nevertheless,<br />

the transporters revealed different Km and V max values for nitrate.<br />

Flux analysis of single gene mutants using 13 NO 3 – showed that Km values for<br />

the NRTA and NRTB proteins were about 100 and 10 mM, respectively, while<br />

V max values were approximately 600 and 100 nmol/mg DW/h, respectively<br />

(Unkles et al. (2001). This kinetic differentiation may provide the physiological<br />

plasticity to acquire sufficient nitrate despite highly variable external concentrations.<br />

In Hansenula polymorpha, the genomic DNA containing the nitrate reductase-(YNR1)<br />

and nitrite reductase-(YNI1) encoding genes, revealed an open<br />

reading frame of 1524 nucleotides (named YNT1, yeast nitrate transporter<br />

gene) encoding a putative protein of 508 amino acids with great similarity to<br />

the nitrate transporters from Aspergillus nidulans and Chlamydomonas reinhardtii<br />

(Perez et al. 1997). Disruption of the chromosomal YNT1 copy resulted<br />

in an incapacity to grow in nitrate and a significant reduction in the rate of<br />

nitrate uptake. The disrupted strain was still sensitive to chlorate and, in the<br />

presence of 0.1 mM nitrate, the expression of YNR1 and YNI1, as well as the<br />

activity of nitrate reductase and nitrite reductase, were significantly reduced<br />

compared to the wild type. Northern-blot analysis showed that YNT1 was<br />

expressed when the yeast was grown in nitrate and nitrite, but not in ammonium<br />

solution (Perez et al. 1997).<br />

In Hansenula polymorpha, the YNT1 gene encodes a high affinity nitrate<br />

transporter (Km 2–3 mM) which constitutes quantitatively the main nitrate<br />

transporter activity in the fungus. The existence of a second nitrate transporter<br />

has been inferred from different experimental pieces of evidence, but<br />

the gene has not yet been identified (Machin et al. 2000). The protein Ynt1 also<br />

transports nitrite with high affinity and belongs to the proposed NNP (nitrate<br />

nitrite porter) family involved in nitrate and nitrite transport (Forde 2000).<br />

This family, in turn belongs to the major facilitator superfamily (MFS), constituted<br />

by transmembrane proteins in which 12 membrane spanning helices<br />

connect cytosolic N-terminal and C-terminal domains (Pao et al. 1998). However,<br />

in Hansenula polymorpha, it is not clear whether nitrite enters through a<br />

specific transport system, or if it shares a nitrate transport.Ynt1 presents similarity<br />

in sequence with the Aspergillus nidulans nitrate transporter NRTA<br />

(CRNA) and the high affinity nitrate transporters in <strong>plant</strong>s (Siverio 2002).<br />

In the field of endomycorrhizas, PCR amplifications using tomato DNA<br />

and degenerate oligonucleotide primers allowed the identification of a new<br />

putative nitrate transporter, named NRT2 (Hildebrandt et al. 2002). Its<br />

sequence showed typical motifs of a high affinity nitrate transporter of the<br />

MFS. The formation of its mRNA was positively controlled by nitrate, and<br />

negatively by ammonia, but not by glutamine. In situ hybridization experiments<br />

showed that this transporter was mainly expressed in rhizodermal<br />

cells. In roots colonized by the arbuscular mycorrhizal fungus Glomus<br />

intraradices, transcript formation of NRT2 extended to the inner cortical cells


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 397<br />

where the fungal structures, arbuscules and vesicles, were concentrated.<br />

Northern analyses indicated that the expression of the transporter was higher<br />

in mycorrhized tomato roots than in noncolonized controls. In addition, mycorrhization<br />

caused a significant expression of a nitrate reductase gene of Glomus<br />

intraradices. According to the authors mentioned above, the results sug-<br />

AAT ALAT<br />

2-oxo Glu 2-oxo<br />

2-oxo<br />

NO 3 -<br />

Nrt2<br />

Asp oaa pyr Ala<br />

NR<br />

NO 3 -<br />

Amt1<br />

C metabolism<br />

Glu<br />

Glu<br />

GDH GS<br />

NIR<br />

NO 2 -<br />

NH 4 +<br />

Amt2<br />

NH 4 +<br />

2-oxo<br />

GOGAT<br />

Inhibition (?)<br />

-<br />

Amt3<br />

Gln<br />

Gap1<br />

Gln<br />

-<br />

transcription<br />

AMT1<br />

AMT2<br />

AMT3<br />

GDHA<br />

NAR1<br />

GLNA<br />

mRNA<br />

Fig. 1. A model describing the regulation of nitrogen transport and assimilation in<br />

Hebeloma cylindrosporum. This ectomycorrhizal fungus is able to use nitrate, ammonium<br />

and amino acids as nitrogen sources. Under low ammonium status, AMT1, AMT2,<br />

AMT3, GDHA and GLNA are transcribed, which results in elevated ammonium uptake<br />

and metabolism capacities. Under ammonium excess, AMT1, AMT2 and GDHA are efficiently<br />

repressed, which results in reduced ammonium assimilatory capacities. Under<br />

these conditions, AMT3 and GLNA would ensure the maintenance of a basal level of<br />

ammonium assimilation. AMT1 and AMT2 transcript levels are controlled through the<br />

effect of intracellular glutamine, whereas the GDHA and NAR1 mRNA level is controlled<br />

by ammonium (bold dotted lines). Ammonium uptake activity may be controlled by<br />

intracellular NH 4 + through a direct effect (dotted lines). 2-oxo 2-oxoglutarate, oaa<br />

oxaloacetate, pyr pyruvate, GOGAT glutamate synthase, Aat aspartate aminotransferase,<br />

Alat alanine aminotransferase, NR nitrate reductase, NIR nitrite reductase, Nrt2 nitrate<br />

transporter, GAP1 general amino acid transporter


398<br />

A. Javelle et al.<br />

gest that mycorrhization positively affects nitrate uptake from soil and nitrate<br />

allocation to the <strong>plant</strong> partner, probably mediated preferentially by the transporter.<br />

In addition, part of the nitrate taken up is very likely reduced by the<br />

fungal partner itself and may then be transferred, when in excess, as glutamine<br />

to the <strong>plant</strong>’s symbiotic partner.<br />

Nitrate transporters have not yet been fully characterized in ectomycorrhizal<br />

fungi. A gene has been isolated in Hebeloma cylindrosporum by Jargeat<br />

et al. (2000; Fig. 1), but the molecular mechanism of its regulation is unknown.<br />

However, in this fungus, Jargeat et al. (2003) has shown more recently that the<br />

nitrate transporter polypeptide is characterized by 12 transmembrane<br />

domains and presents both a long putative intracellular loop and a short Cterminal<br />

tail, two structural features which distinguish fungal high-affinity<br />

transporters from their <strong>plant</strong> homologues. In addition, in Hebeloma cylindrosporum,<br />

transcription of the nrt2 gene (as well as the gene encoding a<br />

nitrite reductase) was repressed by ammonium and stimulated, not only in<br />

the presence of nitrate, but also in the presence of organic nitrogen sources or<br />

under nitrogen deficiency (Jargeat et al. 2003).<br />

3 Ammonium Transport<br />

3.1 Physico-Chemical Properties of Ammonium: Active Uptake Versus<br />

Diffusion<br />

Using [ 14 C]methylammonium as an analogue of ammonium, the kinetics and<br />

the energetics of NH 4 + transport were studied in the ectomycorrhizal fungus<br />

Paxillus involutus (Javelle et al. 1999) and ammonium transporters were first<br />

cloned in Hebeloma cylindrosporum (AMT2 and AMT3; Javelle et al. 2001) and<br />

Tuber borchii (AMT1; Montanini et al. 2002). Although the process of ammonium<br />

uptake is often considered as a rate-limiting step in its acquisition<br />

(Jongbloed et al. 1991; Javelle et al. 1999) it has received relatively little attention<br />

(Burgstaller 1997).<br />

Ammoniac (NH 3) is a weak base (pK a of 9.25), with a dipole moment of<br />

1.47D. The neutral molecule, NH 3, dissolves much more rapidly in organic solvents<br />

than its ionic counterpart, NH 4 + . Consequently, the permeability of NH3<br />

across lipid bilayers is three orders of magnitude greater than that of NH 4 + .<br />

Whilst diffusion of NH 3 across the lipid portion of membranes is believed to<br />

be of biological significance, diffusion of NH 4 + is not. Reported permeability<br />

values for ammoniac, ranging from 2.6 mmol/s (Ritchie and Gibson 1987) to<br />

47 mmol/s (Yip and Kurtz 1995), were found in biomembranes. Therefore,<br />

previous investigations have supported the hypothesis that ammonia is transported<br />

as a small, uncharged and lipophilic compound across the plasma<br />

membrane, a process which does not require specific transporters. However,<br />

rates of diffusion do not seem to be sufficient to account for the requirements


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 399<br />

of <strong>plant</strong> growth (Burgstaller 1997). At a neutral pH typical of cell cytosol,<br />

approximately 99 % of ammonium is present as the cation NH 4 + . By definition,<br />

a decrease of one pH unit is accompanied by a tenfold increase of the<br />

ratio NH 4 + :NH3 . Therefore, in spite of the general acceptance that NH 3 can<br />

readily diffuse across natural membranes, it was postulated that ammonium<br />

uptake in cells could also be mediated by other mechanisms.<br />

3.2 Physiology of Ammonium Transport in Ectomycorrhizas<br />

The first evidence that a specific ammonium transport system acts in fungi<br />

came from the works of Hackette et al. (1970). They used the ammonium-analogue<br />

tracer [ 14 C]methylammonium and suggested that an ammonium transporter<br />

acts in the fungus Penicillium chrysogenum. The radioactive ammonium<br />

analogue [ 14 C]methylammonium has been widely used to assay uptake.<br />

Roon et al. (1975) measured an uptake in Saccharomyces cerevisiae which<br />

resulted in a 1000-fold accumulation. In a further study, Dubois and Grenson<br />

(1979) showed that the uptake of ammonium/methylammonium in S. cerevisiae<br />

is mediated by at least two functionally distinct systems, but this study<br />

was hampered by the lack of molecular characterization of the transport systems.<br />

The first ammonium transporter genes characterized were MEP1<br />

cloned in S. cerevisiae (Marini et al. 1994), and AMT1 cloned in Arabidopsis<br />

thaliana (Ninnemann et al. 1994). They belong to a multigenic family, the socalled<br />

Mep/Amt family.<br />

Ammonium mobilization by mycelium from soil sources is directly linked<br />

to hyphal uptake capacities. Using [ 14 C]methylamine, kinetics of ammonium/methylammonium<br />

transport in ectomycorrhizal fungi have been characterized<br />

(Jongbloed et al. 1991; Javelle et al. 1999). A saturable mediated<br />

uptake was obtained, which conformed to simple Michaelis-Menten kinetics,<br />

and was consistent with a carrier-mediated transport. Both pH dependence<br />

and inhibition by protonophores indicate that methylamine transport in P.<br />

involutus is dependent on the electrochemical H + -gradient (Javelle et al.<br />

1999). These results suggest that ammonium uptake is an active (energyrequiring)<br />

process. Comparing the ammonium uptake capacity of the two<br />

partners separately or in symbiosis, it was found that mycelia have much<br />

higher capacities for ammonium uptake than nonmycorrhizal roots and ectomycorrhizal<br />

fungi increase ammonium uptake capacities of their host roots<br />

(Plassard et al. 1997; Javelle et al. 1999).<br />

Nitrogen starvation increased methylamine transport in P. involutus<br />

(Javelle et al. 1999) and similarly, N-starved <strong>plant</strong>s usually showed a faster<br />

NH 4 + net uptake than N-fed <strong>plant</strong>s (Howitt and Udvardi 2000). However, these<br />

studies were hampered by the lack of molecular characterization of the transport<br />

systems involved and their regulation at the molecular level remains to<br />

be clarified.


400<br />

A. Javelle et al.<br />

3.3 Isolation of Ammonium Transporter Genes<br />

Molecular studies of ammonium transporters in ectomycorrhizal fungi are<br />

still scarce and concern only the ectomycorrhizal fungus Hebeloma cylindrosporum.<br />

Three ammonium transporters, HcAmt1, HcAmt2 and HcAmt3<br />

(Ammonium transporter) were cloned in H. cylindrosporum. Both Southern<br />

blot experiments and cDNA library screening indicate that H. cylindrosporum<br />

has only three ammonium transporters, like the yeast S. cerevisiae (Marini et<br />

al. 1997; Javelle et al. 2001; Javelle et al. 2003b). The hydropathy profiles of<br />

HcAmt1, HcAmt2 and HcAmt3 generated with the Kyte and Doolittle algorithm,<br />

consist of 11 hydrophobic domains of sufficient length to be considered<br />

as potential membrane-spanning domains.<br />

The function of HcAmts in ammonium transport was further characterized<br />

by yeast mutant complementation, as previously described for ammonium<br />

transporters from <strong>plant</strong>s and animals. S. cerevisiae possesses three<br />

ammonium transporters, namely Mep1, Mep2 and Mep3 (Methylammonium<br />

permease). The yeast strain 31019b, mep1D mep2D mep3D, was unable to<br />

grow on media containing less than 1 mM ammonium as sole nitrogen<br />

source (Marini et al. 1997). Functional expression of HcAmt1, HcAmt2 or<br />

HcAmt3 in this triple mutant resulted in complementation of growth defects<br />

in the presence of less than 1 mM ammonium as sole nitrogen source. Thus,<br />

HcAMTs cDNA encode functional NH 4 + transporters. Kinetic parameters<br />

were determined using [ 14 C]methylammonium as a tracer in the transformed<br />

yeast strain 31019b. Previous works with mycorrhizal fungi reported<br />

Km values in the range 110–180 mM when using methylamine as substrate<br />

(Javelle et al. 1999). However, such data could be the result of multiple transporter<br />

expressions. In H. cylindrosporum, as well as in other organisms<br />

(Marini et al. 1997; Gazzarrini et al. 1999; Howitt and Udvardi 2000), multiple<br />

Amt transporters with complementary affinities probably allow the fungus<br />

to maintain a steady ammonium uptake over a wide range of concentrations.<br />

Indeed, in forest soils the quality and quantity of nitrogen sources<br />

can vary considerably.<br />

3.4 Regulation of the Ammonium Transporters<br />

Expression levels of the three ammonium transporter (AMT1, AMT2, AMT3)<br />

genes were studied by Northern blot analysis under different nitrogen conditions.<br />

AMT1 and AMT2 are high affinity transporters (for example, Km:<br />

58 mM for methylammonium at pH 6.1 for AMT2), while AMT3 is a low affinity<br />

transporter (Km: 260 mM for methylammonium at pH 6.1; Javelle et al.<br />

2001). In response to exogenously supplied ammonium or Gln, AMT1 and<br />

AMT2 were down-regulated, while they were up-regulated upon nitrogen<br />

deprivation or in the presence of nitrate. This indicates that these genes are


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 401<br />

subjected to nitrogen repression in H. cylindrosporum (Fig. 2). AMT3 was<br />

poorly regulated at this level.<br />

Expression of AMT1 only in ammonium-limiting conditions is consistent<br />

with a role for the high-affinity ammonium transporter in scavenging low<br />

concentrations of ammonium. The low-affinity ammonium transporter Amt3<br />

would be required for growth in ammonium-sufficient conditions.<br />

In order to identify the effector(s) for nitrogen regulation in H. cylindrosporum,<br />

the correlation coefficient for the relationship between AMT1,<br />

AMT2, AMT3, transcript levels and N-compound amounts were calculated.<br />

This transcriptional control is driven by intracellular Gln. Indeed, an intracel-<br />

Fig. 2. AMT1,AMT2,AMT3,GDHA and GLNA mRNA levels in Hebeloma cylindrosporum.<br />

Fungal colonies were grown for 10 days on cellophane-covered agar medium containing<br />

3.78 mM ammonium as sole nitrogen source (T 0 ) and transferred to a N-free liquid medium<br />

for 12 h (–N).Some colonies were further transferred to a 0.1,1 or 10 mM ammonium-containing<br />

medium. Total RNA was extracted at 3, 6, 12 and 24 h from 100 mg of mycelium and<br />

20 mg/lane were separated on 1.5 % agarose-formaldehyde gel and hybridized to the<br />

[a- 32 P]dCTP labelled cDNA probes or 5.8S rRNA probe as loading control


402<br />

A. Javelle et al.<br />

lular Gln amount higher than 2 nmol/mg DW seems to be sufficient to promote<br />

AMT1 repression in H. cylindrosporum (Javelle et al. 2003b). Ammonium<br />

influx is inhibited by intracellular ammonium which agrees with other<br />

findings from A. bisporus (Kersten et al. 1999), and A. thaliana (Rawat et al.<br />

1999), but mechanisms responsible for this regulation remain unclear.<br />

3.5 Other Putative Functions of Ammonium Transporters<br />

In addition to their role in ammonium uptake and retrieval, ammonium<br />

transporters may have a third putative role. A diploid wild-type strain of the<br />

yeast S. cerevisiae undergoes a dimorphic transition to filamentous growth in<br />

response to nitrogen starvation. Mep2 is one of three related ammonium per-<br />

HcAmt3<br />

AAK82417<br />

ScMep3<br />

P53390<br />

AnMeaa<br />

AAL73117<br />

ScMep1<br />

P40260<br />

NcMep3<br />

Contig 3.17<br />

CaAmt2<br />

Contig 6.2476<br />

HcAmt1<br />

AY094982<br />

0.00<br />

0.05<br />

0.10<br />

0.15<br />

0.20<br />

ScMep2<br />

P41948<br />

HcAmt2<br />

AAK82416<br />

AnMepa<br />

AAL73118<br />

MvAmta<br />

AAD40955<br />

TbAmt1<br />

AAL11032<br />

UmMep1<br />

AAL08424<br />

NcMepa<br />

CAD21326.1<br />

Fig. 3. Phylogenetic relationships among fungal Mep/Amt proteins. Complete amino<br />

acid sequences derived from full-length cDNA predicted using TMHMM algorithm were<br />

aligned with Clustalw and the tree was constructed by the neighbor-joining method<br />

using Mega 2.1. p-distances were estimated between all pairs of sequences using the<br />

complete deletion option. Gene names and GenBank accession numbers are indicated.<br />

Proteins in bold belong to the high affinity ammonium transporter and sensor family<br />

(TC 2A 49 3 2), according to the TC classification. Organisms are as follows. An<br />

Aspergillus nidulans, Ca Candida albicans, Hc Hebeloma cylindrosporum, Mv Microbotryum<br />

violaceum, Nc Neurospora crassa, Sc Saccharomyces cerevisiae, Tb Tuber borchii,<br />

Um Ustilago maydis


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 403<br />

meases which plays a unique role as a nitrogen sensor in the transduction<br />

pathway of pseudohyphal differentiation in S. cerevisiae not shared with the<br />

related Mep1 and Mep3. Interestingly, in the ectomycorrhizal fungus H. cylindrosporum,<br />

two ammonium transporters (Amt1 and Amt2) are able to complement<br />

the pseudohyphal growth defect of a homozygotous mep2D yeast<br />

mutant, whereas the third ammonium transporter (Amt3) is unable to do so<br />

(Javelle et al. 2001, 2003b).According to the classification of the transport system<br />

available at http://www-biology.ucsd.edu/~msaier/transport/ (TC system),<br />

the HcAmts can be divided into two groups. HcAmt1, HcAmt2 and<br />

Mep2 belong to the high affinity ammonium transporter and sensor family<br />

(TC 2A 49 3 2), whereas HcAmt3 belongs to the low affinity ammonium transporter<br />

family (TC 2A 49 3 1; Fig. 3).<br />

We have recently hypothesized that high affinity ammonium transporters<br />

from mycorrhizal fungi sense the environment and induce via signal transduction<br />

cascades a switch of the fungal growth mode observed during mycorrhiza<br />

formation. Upon entering the root depletion zone, mycorrhizal fungi<br />

may receive a signal through this sensing mechanism which induces hyphal<br />

proliferation around roots, corresponding to the primary events in ectomycorrhiza<br />

formation (Javelle et al. 2003a).<br />

4 Amino Acid Transport<br />

4.1 Utilization of Amino Acids by Ectomycorrhizal Partners<br />

It has been well established that ectomycorrhizal fungi can use amino acids as<br />

nitrogen and carbon sources (Abuzinadah and Read 1988; Näsholm et al.<br />

1998). Using 14 C-labelled compounds, Wallenda and Read (1999) determined<br />

the kinetics of uptake of amino acids by excised ectomycorrhizal roots from<br />

beech, spruce, and pine. All mycorrhizal types took up amino acids via highaffinity<br />

transport systems with Km values ranging from 19 to 233 mM.A comparative<br />

analysis for the uptake of amino acids and the ammonium analogue<br />

methylammonium showed that ectomycorrhizal roots have similar or even<br />

higher affinities for the amino acids, indicating that absorption of these N<br />

organic forms can contribute significantly to total N uptake by ectomycorrhizal<br />

<strong>plant</strong>s.<br />

Transport of amino acids was investigated in the mycorrhizal fungi Paxillus<br />

involutus (Chalot et al. 1996), and Amanita muscaria (Nehls et al. 1999),<br />

which demonstrated their ability to take up a variety of amino acids. In the<br />

latter fungus, the uptake characteristics of the encoded transporter protein, as<br />

analysed by heterologous expression in yeast, identified the protein as a highaffinity,<br />

general amino acid permease (Km: 22 mM for histidine and up to<br />

100 mM for proline). The uptake of amino acids showed characteristic features<br />

of active transport.


404<br />

A. Javelle et al.<br />

In Paxillus involutus, the apparent Km derived from the Eadie-Hofstee<br />

plots ranged from 7 mM for alanine to 27 mM for glutamate. Maximal velocities,<br />

expressed as mmol (g dry weight) –1 min –1 , were between 0.24 for alanine<br />

and 0.71 for glutamine. In this fungus, the uptake of amino acids markedly<br />

depended on the pH and was optimal at pH 3.9–4.3 for glutamate and glutamine,<br />

and at pH 3.9–5.0 for alanine and aspartate.<br />

Both pH dependence and inhibition by protonophores, such as 2,4-dinitrophenol<br />

(DNP) and carbonyl cyanide m-chlorophenylhydrazone (CCCP), were<br />

consistent with a proton symport mechanism for amino acid uptake by Paxillus<br />

involutus. Competition studies indicated a broad substrate recognition by<br />

the uptake system, which resembles the general amino acid permease of yeast<br />

(Chalot et al. 1996, 2002).<br />

The impact of birch mycorrhization with Paxillus involutus led to a profound<br />

alteration of the metabolic fate of exogenously supplied amino acids<br />

(Blaudez et al. 2001). Inoculation increased [ 14 C]glutamate and [ 14 C]malate<br />

uptake capacities by up to 8 and 17 times, respectively, especially in the early<br />

stages of mycorrhiza formation. In addition, it was demonstrated that Gln was<br />

the major 14 C-sink in mycorrhizal roots and in the free-living fungus. In contrast,<br />

citrulline and insoluble compounds were the major 14 C compounds in<br />

nonmycorrhizal roots (Blaudez et al. 2001).<br />

In order to study how amino acid transport characteristics were affected by<br />

mycorrhization, Sokolovsky et al. (2002) used an electrophysiological<br />

approach in Calluna vulgaris associated or not with the ericoid fungus<br />

Hymenoscyphus ericae. Both the V max and Km parameters of amino acid<br />

uptake were affected by fungal colonization in a manner consistent with an<br />

increased availability of amino acid to the <strong>plant</strong>. Indeed, the transport capacity<br />

for asparagine, histidine, ornithine and lysine, in particular, was increased<br />

after colonization. Interestingly, a-aminobutyric acid led to a large depolarization<br />

only in colonized cells. This implies that mycorrhization triggers a<br />

capacity to transport a broader range of substrates, including amino acids<br />

that are not metabolized.<br />

4.2 Molecular Regulation of Amino Acid Transport<br />

In Amanita muscaria, only a low, constitutive expression of the amino acid<br />

transporter was detected in the presence of amino acids and ammonium,<br />

which are both sources of N for the fungus (Nehls et al. 1999). By contrast,<br />

under N starvation, or in the presence of nitrate or phenylalanine, not utilized<br />

by the fungus as N sources, expression of the gene was considerably<br />

enhanced. Therefore, in Amanita muscaria, as in S. cerevisiae or Aspergillus<br />

nidulans (Sophianopoulou and Diallinas 1995), gene expression of amino<br />

acid transporters is regulated at the transcriptional level by N repression. In<br />

addition to amino acid uptake for nutrition, the enhanced expression of the


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 405<br />

gene under conditions of N starvation, suggests that the transporter can also<br />

be involved in the prevention of amino acid loss by hyphal leakage in the<br />

absence of a suitable N source (Nehls et al. 1999).<br />

A gene named HcBap1 has recently been isolated from H. cylindrosporum<br />

by functional complementation of a yeast strain deficient in amino acid transporters<br />

(Wipf et al. 2002).<br />

5 Reduction of Nitrate to Nitrite and Ammonium<br />

5.1 Reduction of Nitrate to Nitrite<br />

Nitrate assimilation in fungi follows the same pathway as that described for<br />

yeasts and <strong>plant</strong>s. After transport into the cells, nitrate is converted to ammonium<br />

by two successive reductions catalysed respectively by nitrate reductase<br />

and nitrite reductase. Although nitrate is one of the most abundant nitrogen<br />

sources in nature, numerous fungi more readily use ammonium, especially<br />

ectomycorrhizal fungi which live predominantly in forest soils where a high<br />

organic material content maintains an acidic pH. Under these circumstances,<br />

nitrification is inhibited and ammonium is usually the main form of mineral<br />

nitrogen (Vitousek and Matson 1985). However, it has been shown that ectomycorrhizal<br />

fungi are also able to utilize NO 3 – which, for a few species, is capable<br />

of promoting better growth than ammonium (Scheromm et al. 1990;<br />

Anderson et al. 1999).<br />

The enzyme complex nitrate reductase which is a molybdoflavoprotein catalyzes<br />

the reduction of NO 3 – to NO2 – by reduced pyridine nucleotides. The<br />

enzyme of higher <strong>plant</strong>s has a high molecular weight, varying from 220 to<br />

600 kDa, depending on the organisms in which it occurs (Notton and Hewitt<br />

1978). In fungi, nitrate reductase has been extensively studied in Neurospora<br />

crassa where it is found as a 228-kDa homodimer (Garrett and Nason 1969)<br />

and in Aspergillus nidulans where the enzyme has a molecular mass of<br />

180 kDa (Minagawa and Yoshimoto 1982). In <strong>plant</strong>s and fungi, the polypeptide<br />

is located in the cytosolic soluble fraction, but is weakly bound to the<br />

plasmalemma and tonoplast in Neurospora crassa (Roldan et al. 1982).<br />

Nitrate reductase generally appears to be unstable and,due to the difficulties<br />

experienced in purifying the enzyme, information on its properties in mycorrhizal<br />

fungi is very scarce. However, nitrate reduction by partially purified<br />

enzyme preparations has been investigated in Hebeloma cylindrosporum by<br />

Plassard et al. (1984a). The Michaelis constants for nitrate, NADPH and FAD<br />

were found to be 152, 0.185, and 22.7 mM,respectively.In Pisolithus tinctorius,<br />

nitrate reductase exhibited less affinity for nitrate (Km: 328 mM) and for<br />

NADPH (Km: 49.6mM; Aouadj et al. 2000), but the enzyme was similar to those<br />

found in nonmycorrhizal fungi. Such values are in the same range as those<br />

found in higher <strong>plant</strong> tissues and suggest that ectomycorrhizal fungi have


406<br />

A. Javelle et al.<br />

capabilities of reducing NO 3 – similar to those of most higher <strong>plant</strong>s. However,<br />

nitrate reductase activity varies greatly between mycorrhizal species and isolates.For<br />

example,in Rhizopogon vulgaris,nitrate reductase was 32-fold higher<br />

in the S-251 isolate than in the S-219 isolate (Ho and Trappe 1987).<br />

In the ectomycorrhizal basidiomycete Suillus bovinus, nitrate reductase<br />

proved to be substrate-induced and activity could only be measured after<br />

exposure of the mycelia to exogenous nitrate (Grotjohann et al. 2000). Similar<br />

results were found in Scleroderma verrucosum (Prima Putra et al. 1999), and<br />

Pisolithus tinctorius (Aouadj et al. 2000), where both nitrate reductases were<br />

strongly induced in the presence of nitrate and repressed by ammonium.<br />

5.2 Reduction of Nitrite to Ammonium<br />

Nitrite reductase from the ectomycorrhizal basidiomycete Hebeloma cylindrosporum<br />

is specific for NADPH and was found to be very unstable (Plassard<br />

et al. 1984b). The saturation curve of the enzyme for NO 2 – was biphasic with<br />

two apparent Km values at 13 and 350 mM. This suggests that the enzyme of<br />

Hebeloma cylindrosporum has two types of binding sites for NO 2 – which could<br />

make the reaction continuously responsive to concentration changes over a<br />

wide range. Nitrite reductase activity measured in Hebeloma cylindrosporum<br />

was similar to the nitrate reductase activity, ranging from 10 to 30 mmol h –1 g –1<br />

fresh weight, which is considerably higher than the in vivo NO 3 – uptake capacity<br />

of the mycelium (Plassard et al. 1984b). Consequently, nitrite does not<br />

accumulate in the fungal cells, and this indicates that nitrite reductase is obviously<br />

not a limiting step of NO 3 – assimilation in this ectomycorrhizal fungus.<br />

5.3 Molecular Characterization of Nitrate Reductase and Nitrite<br />

Reductase<br />

Genes encoding proteins involved in nitrate assimilation are usually induced<br />

by nitrate and subjected to nitrogen catabolite repression. Cloning of two<br />

nitrate reductase (NR) genes has been carried out in the ectomycorrhizal fungus<br />

Hebeloma cylindrosporum (Jargeat et al. 2000). One of these genes (nar1)<br />

is transcribed and codes for a 908 amino acid polypeptide, while the other<br />

gene (nar2) for which no mRNA transcripts were detected, is considered to be<br />

an ancestral, nonfunctional duplication of nar1. It is well known that high<br />

nitrate reductase activities are found in mycelia of Hebeloma cylindrosporum<br />

cultivated in ammonium-containing media, sometimes higher than those<br />

exhibited in the presence of nitrate (Plassard et al. 1986). However, Northern<br />

analyses showed that nar1 in Hebeloma cylindrosporum was strongly<br />

repressed by ammonium, while low nitrogen concentrations or high levels of<br />

nitrate, urea, glycine or serine sustained a high level of transcription (Jargeat


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 407<br />

et al. 2000). The authors have put forward the hypothesis that the nitrate<br />

reductase enzyme of the fungus might be extremely stable in vivo and progressively<br />

accumulates in the cells growing on ammonium. In addition, such<br />

results indicate that in Hebeloma cylindrosporum, expression of the nitrate<br />

reductase gene is regulated primarily by the availability of ammonium, but<br />

not by the presence of nitrate in the medium. This regulation pattern clearly<br />

distinguishes this fungus from the other saprophytic and pathogenic species<br />

previously studied.<br />

Assimilatory nitrate reductase of higher <strong>plant</strong>s is subjected to a complex regulation<br />

of its expression and catalytic properties (Kaiser and Huber 2001).The<br />

NR protein is inactivated by phosphorylation combined with a link with a<br />

dimeric protein,which may cause a change in NR conformation that interrupts<br />

electron transport between the heme and the molybdenum-cofactor domains<br />

(Kaiser and Huber 2001).It is known that light as well as CO 2 and oxygen availability<br />

are the major external triggers for a rapid and reversible modulation of<br />

NR activity, and that sugars and/or sugar phosphates are the internal signals<br />

which regulate the protein kinase(s) and phosphatase. In ectomycorrhizal<br />

fungi, there is no evidence, so far, for a specific post-translational inactivation<br />

of the NR protein. In Hebeloma cylindrosporum, the main NR protein named<br />

NAR1,like all other fungal NR polypeptides,lacks the short motifs found in the<br />

N-terminal and hinge 1 domains of <strong>plant</strong> NRs,which are both necessary for the<br />

post-translational inactivation of these enzymes in response to changes in<br />

light or CO 2 status (Su et al. 1996; Jargeat et al. 2000).<br />

Indeed, in Neurospora crassa the structural genes that encode nitrogen<br />

catabolic enzymes are subject to nitrogen metabolite repression, mediated by<br />

the positive-acting NIT2 protein and by the negative-acting NMR protein (for<br />

“nitrogen metabolite repression”; Pan et al. 1997). NIT2, a globally acting factor,<br />

(or AREA in Aspergillus nidulans, or GLN3 in Saccharomyces cerevisiae) is<br />

a member of the GATA family of regulatory proteins and has a single<br />

Cys 2/Cys 2 zinc finger DNA-binding domain. Deletions or certain amino acid<br />

substitutions within this zinc finger and the carboxy-terminal tail resulted in<br />

a loss of nitrogen metabolite repression (Marzluf 1997). Those mutated forms<br />

of NIT2 that were insensitive to nitrogen repression had also lost one of the<br />

NIT2-NMR protein–protein interactions. These results provide compelling<br />

evidence that the specific NIT2–NMR interactions have a regulatory function<br />

and play a central role in establishing nitrogen metabolite repression (Pan et<br />

al. 1997).<br />

The different genes involved in nitrate assimilation, as well as putative<br />

nitrate transport systems, have been cloned from various saprophytic and<br />

pathogenic filamentous ascomycetes; all of these genes are single-copy genes<br />

and their transcription is subject to ammonium/glutamine repression and<br />

nitrate induction (Kinghorn and Unkles 1994). In the yeast Hansenula polymorpha,<br />

the genes YNT1, YNR1 and YNI1, encoding respectively nitrate transport,<br />

nitrate reductase and nitrite reductase (NiR), have been cloned, as well


408<br />

A. Javelle et al.<br />

as two other genes encoding transcriptional regulatory factors. Transcriptional<br />

regulation is the main regulatory mechanism that controls the levels of<br />

the enzymes involved in nitrate metabolism (Siverio 2002). The genetic and<br />

molecular bases of repression and induction have been studied in detail in<br />

Aspergillus nidulans and Neurospora crassa (Scazzocchio and Arst 1989; Caddick<br />

et al. 1994; Marzluf 1997).<br />

In both species, nitrate induction is mediated by a pathway-specific regulatory<br />

gene (nirA and nit-4 in, respectively Aspergillus nidulans and Neurospora<br />

crassa), whose product binds to the promoters of the nitrate pathway genes<br />

when NO 3 – is present in the culture medium. Similarly, derepression is mediated<br />

by a wide-domain regulatory gene (respectively areA and nit-2), which<br />

encodes a GATA DNA-binding protein. Both areA and nit-2 are responsible, at<br />

least in part, for the derepression, when ammonium is absent, of several other<br />

genes involved in the use of other nitrogen sources, such as several amino<br />

acids or proteins.<br />

In Neurospora crassa and Aspergillus nidulans, glutamine appears to be the<br />

critical metabolite which exerts nitrogen catabolite repression (Chang and<br />

Marzluf 1979; Premakumar et al. 1979). Ammonia leads to strong nitrogen<br />

repression in these fungi, but is not itself active, since it does not cause repression<br />

in mutants lacking glutamine synthetase (Premakumar et al. 1979). Intracellular<br />

glutamine, or possibly a metabolite derived from it, leads to repression,<br />

but the cellular location of the glutamine pool responsible for this<br />

control response, e.g., cytoplasmic or vacuolar, is unknown. An extremely<br />

important, but still unknown feature is the identity of the element or signal<br />

pathway system that senses the presence of repressing levels of glutamine. It is<br />

conceivable that the AREA, NIT2, GLN3, and similar global regulators themselves<br />

bind glutamine or that a complex such as a NIT2-NMR heterodimer<br />

recognizes the amino acid. However, it is also possible that an as yet unidentified<br />

factor detects glutamine and conveys the repression signal to the global<br />

activating proteins. Thus, an important goal for future research is the creative<br />

use of genetic and biochemical approaches to identify the signalling system<br />

that recognizes and processes environmental nitrogen cues.<br />

In the ectomycorrhizal fungus Hebeloma cylindrosporum, transcription of<br />

nar1 coding for the NR protein, was repressed in the presence of ammonium,<br />

suggesting that the organism might possess a gene homologous to nit-2 in<br />

Neurospora crassa. According to Jargeat et al. (2000), inspection of the<br />

sequences flanking the NR genes cloned from Hebeloma cylindrosporum<br />

revealed that they contain several GATA elements to which regulatory GATA<br />

proteins could bind.<br />

In Neurospora crassa, expression of structural genes which encode the<br />

nitrate assimilatory enzymes also has an absolute requirement for nitrate<br />

induction mediated by a pathway-specific factor, NIT4 (or NIRA in<br />

Aspergillus nidulans; Marzluf, 1997). The Neurospora crassa NIT4 protein is<br />

composed of 1090 amino acids and contains at its amino terminus a Cys 6 /Zn 2


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 409<br />

binuclear zinc cluster followed by a spacer region and a coiled-coil motif that<br />

mediates the formation of a homodimer, the form that is responsible for<br />

sequence-specific DNA binding.<br />

In Hebeloma cylindrosporum, supply of nitrate is not necessary for the<br />

transcription of the NR gene (Jeargeat et al. 2000), suggesting that in this fungus<br />

there is no transcription factor such as NIT4 capable of promoting transcription<br />

in the presence of nitrate. In agreement with this hypothesis, no<br />

motifs resembling the binding sites for NIT4 or NIRA were detected in the<br />

promoter regions of the genes cloned in the ectomycorrhizal fungus (Jeargeat<br />

et al. 2000).<br />

In the yeast Hansenula polymorpha the YNT1 gene encoding the nitrate<br />

transporter is clustered with the structural genes which encode nitrate reductase<br />

and nitrite reductase (Perez et al. 1997). Clustering of these three assimilation<br />

genes was previously reported in Aspergillus nidulans (Johnstone et al.<br />

1990), and more recently in the ectomycorrhizal fungus Hebeloma cylindrosporum<br />

(Jargeat, Gay, Debaud and Marmeisse, pers. comm.; gene accession<br />

number: AJ 238664), which might represent a cell strategy to make the regulation<br />

of this important pathway efficient.<br />

The role of arbuscular mycorrhizal fungi in assisting their host <strong>plant</strong> in<br />

nitrate assimilation was studied in the association Glomus intraradices/Zea<br />

mays by Kaldorf et al. (1998). With PCR technology, part of the gene coding<br />

for the nitrate reductase apoprotein from either the fungus or from the host<strong>plant</strong><br />

was specifically amplified and subsequently cloned and sequenced.<br />

Northern blot analysis with these probes indicated that the mRNA level of the<br />

maize gene was lower in roots and shoots of mycorrhizal <strong>plant</strong>s than in noncolonized<br />

controls, whereas the fungal gene was highly transcribed in roots of<br />

mycorrhizal <strong>plant</strong>s.<br />

In agreement with these data, the specific nitrate reductase activity of<br />

leaves was significantly lower in endomycorrhizal maize than in the controls.<br />

Nitrite formation catalyzed by nitrate reductase was mainly NADPH-dependent<br />

in roots of mycorrhizal <strong>plant</strong>s, but not in those of the controls, which is<br />

consistent with the fact that these enzymes of fungi preferentially utilize<br />

NADPH as reductant. In addition, it has been shown that the fungal nitrate<br />

reductase mRNA is detected in arbuscules, but not in vesicles by in situ RNA<br />

hybridization experiments (Kaldorf et al. 1998). There is obviously a differential<br />

formation of transcripts of a gene coding for the same function in both<br />

symbiotic partners.<br />

6 Assimilation of Ammonium<br />

Once inside the cell, NH 4 + can be incorporated into the key nitrogen donors<br />

Glu and Gln for biosynthetic reactions. Glutamate dehydrogenase (NADP-<br />

GDH, EC 1.4.1.4) catalyses the reductive amination of 2-oxoglutarate to form


410<br />

A. Javelle et al.<br />

Glu. Glutamine synthetase (GS, EC 6.3.1.2) incorporates ammonium into the<br />

carboxyl group of Glu to form Gln. In turn, the Glu and Gln formed serve as<br />

donors in transamination and amido nitrogen transfer reactions. Glu is an<br />

essential amino N donor for many transaminases and Gln amide nitrogen is<br />

used to synthesize many essential metabolites, such as nucleic acids, amino<br />

sugars, His, Tyr, Asn, and various cofactors. Both Glu and Gln are essential for<br />

protein synthesis. Glutamate synthase (GOGAT) is responsible for the reductive<br />

transfer of amide N to a-ketoglutarate for the generation of two molecules<br />

of glutamate, one of which is recycled for glutamine biosynthesis. The<br />

net result of the combined action of GS and GOGAT is the synthesis of glutamate<br />

from ammonium and a-ketoglutarate, frequently referred to as the<br />

GS/GOGAT cycle.<br />

6.1 Role and Properties of Glutamate Dehydrogenase<br />

Most of the ascomycete and basidiomycete fungi possess two glutamate dehydrogenases<br />

(GDH), each specific for one of the two cofactors. A catabolic role<br />

has been assigned to the NAD-specific enzyme (EC 1.4.1.2), whereas the<br />

NADP-specific enzyme (EC 1.4.1.4) has been involved in glutamate biosynthesis<br />

(Ferguson and Sims 1971). This was confirmed in the ectomycorrhizal<br />

fungus Laccaria laccata where both enzymes were purified and characterized<br />

(Brun et al. 1992; Botton and Chalot 1995; Garnier et al. 1997). Both enzymes<br />

revealed biphasic kinetics with two different Km values for glutamate, the<br />

NADP-GDH exhibiting a positive cooperativity, and the NAD-GDH a negative<br />

cooperativity. At all tested concentrations of glutamate, NAD-GDH showed a<br />

higher affinity for this amino acid than the NADP-specific enzyme. This was<br />

especially true at low glutamate concentrations where the affinity of NADP-<br />

GDH was very low (Km value: 100 mM), while the affinity of NAD-GDH was<br />

maximal (Km value: 0.24 mM). In addition, NADP-GDH was found to have a<br />

considerably higher affinity for ammonium than the NAD-dependent<br />

enzyme and was not calcium-dependent for its activity, contrary to what was<br />

found with the latter enzyme. The native NADP-GDHs purified from Cenococcum<br />

geophilum (Martin et al. 1983), and Laccaria bicolor (Ahmad and<br />

Hellebust 1991), revealed properties roughly similar to those of the Laccaria<br />

laccata NADP-GDH.<br />

Activities of glutamate dehydrogenase in conjunction with glutamine synthetase<br />

in the free-living Pezizella ericae, Cenococcum geophilum (Martin et<br />

al. 1983), and Laccaria laccata (Lorillou et al. 1996), were found to be high and<br />

sufficient to sustain high rates of nitrogen assimilation. In cultured Cenococcum<br />

geophilum,NH 4 + is assimilated via the glutamate dehydrogenase pathway<br />

and the glutamate formed is rapidly used to synthesize glutamine. Ammonium<br />

ion assimilation leads to the synthesis of large amounts of glutamine,<br />

alanine and arginine (Martin et al. 1987). These amino acids represent the


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 411<br />

bulk of the free amino acids found in mycelia of ectomycorrhizal fungi. It was<br />

suggested that polyphosphate, an impermeant macromolecule, traps the large<br />

pool of arginine in the vacuole (Martin 1985), and then reduces the osmotic<br />

pressure of the basic amino acid.<br />

The derepression of NADP-GDH specific activity has been observed on<br />

nitrate, on low ammonium concentrations, or on nitrogen-free media in Laccaria<br />

bicolor (Ahmad et al. 1990; Lorillou et al. 1996), and in a wide range of<br />

other fungal species such as Aspergillus nidulans, Neurospora crassa,<br />

Stropharia semiglobata (Pateman, 1969; Schwartz et al. 1991). The transfer of<br />

Laccaria bicolor from a NH 4 + -rich medium to either NO3 – or N-free media<br />

caused a rapid, several fold increase in enzyme concentration detected by<br />

immunological analysis (Lorillou et al. 1996). These results showed that the<br />

changes in NADP-GDH activity were not related to the activation of a constitutive<br />

inactive precursor of the enzyme, but to de novo accumulation of newly<br />

synthesized GDH. The latter claim was supported by in vivo 35 S-labelling<br />

experiments which showed that steady-state synthesis of the enzyme<br />

increased several fold in mycelia grown in the presence of nitrate or in nitrogen-deficient<br />

media (Lorillou et al. 1996).<br />

In the ectomycorrhizal basidiomycete Suillus bovinus, cultivated in the<br />

presence of ammonium, NADH-dependent glutamate dehydrogenase exhibited<br />

high aminating and low deaminating activities, suggesting that this<br />

enzyme might also be involved in ammonium assimilation (Grotjohann et al.<br />

2000).<br />

NADP-GDH was found to be located in the cytosol as determined by<br />

immunogold labelling carried out in Cenococcum geophilum (Chalot et al.<br />

1990) and Laccaria laccata (Brun et al. 1993).<br />

GDHA, the gene encoding the NADP-GDH has been cloned and characterized<br />

from various fungi (Table 1), including mycorrhizal fungi. In the<br />

ectomycorrhizal fungi Laccaria bicolor (Lorillou et al. 1996), and Tuber<br />

borchii (Vallorani et al. 2002), the increased activity of GDH was correlated<br />

with its increased synthesis, suggesting that an increased expression of<br />

mRNA encoding NADP-GDH occurs under derepressing growth conditions.<br />

Quantification of mRNA using a cDNA probe encoding the Laccaria bicolor<br />

NADP-GDH confirmed that the growth of mycelia on NO 3 – and N-free<br />

media, resulted in an increased accumulation of NADP-GDH transcripts<br />

(Lorillou et al. 1996). However, the two processes were studied independently<br />

in different ectomycorrhizal models and the data obtained until now give<br />

only a fragmentary view of ammonium assimilation and its regulation in<br />

ectomycorrhizal fungi.<br />

More recently, GDHA has been cloned and characterized from Hebeloma<br />

cylindrosporum and expression of the enzyme was studied in this fungus<br />

(Fig. 1; Javelle et al. 2003a). Transfer of the fungus from a 3 mM ammonium to<br />

a N-free medium resulted in a 12-fold increase in the GDH transcript level<br />

(Fig. 2), corresponding to a similar increase of enzyme activity. On the con-


412<br />

A. Javelle et al.<br />

Table 1. Relationships among fungal NADP-dependent GDH (E.C.1.4.1.4) and GS<br />

(E.C.6.3.1.2). Organism, GenBank accession number, sequence length (aa) and molecular<br />

weight (MW) are indicated. Sequence identity (ID) using H. cylindrosporum GDH or<br />

GS sequence as a reference (100 %) is indicated. A. bisporus, Agaricus bisporus; A. muscaria,<br />

Amanita muscaria; A. nidulans, Aspergillus nidulans; B. graminis, Blumeria<br />

graminis; F. neoformans, Filobasidiella neoformans; G. fujikuroi, Gibberella fujikuroi, G.<br />

cingulata, Glomerella cingulata; L. bicolor, Laccaria bicolor; N. crassa, Neurospora crassa;<br />

S. cerevisiae, Saccharomyces cerevisiae; S. pombe, Schizosaccharomyces pombe; S. commune,<br />

Schizophyllum commune; S. occidentalis, Schwanniomyces occidentalis; T. borchii,<br />

Tuber borchii<br />

Organism Accession no. aa MW ID<br />

NADP-dependent glutamate dehydrogenase<br />

N. crassa CAD21426 454 48.8 60.9<br />

A. nidulans S04904 459 49.6 69.2<br />

T. borchii AAG2878 457 50.1 56.9<br />

S. pombe T41492 451 48.8 55.6<br />

S. occidentalis S17907 459 49.8 57.6<br />

S. cerevisiae (GDH1) A25275 454 49.6 56.4<br />

S. cerevisiae (GDH3) AAC04972 457 49.6 56.7<br />

A. bisporus P54387 457 49.6 83.1<br />

L. bicolor AAA82936 450 48.5 84.9<br />

H. cylindrosporum<br />

Glutamine synthetase<br />

AAL06075 450 48.3 100<br />

A. bisporus O00088 354 39.5 90.7<br />

H. cylindrosporum AAK96111 354 39,2 100<br />

A. muscaria CAD22045 378 41.9 87.0<br />

S. commune AAF27660 348 38.3 84.2<br />

F. neoformans CAD10037 358 39.5 72.0<br />

S. cerevisiae NP015360 370 41.4 68.4<br />

S. pombe Q09179 359 40.0 63.8<br />

A. nidulans AAK70354 345 38.5 64.1<br />

G. cingulata Q12613 360 40.0 64.1<br />

G. fujikuroi CAC27836 353 39.4 63.4<br />

B. graminis AAK69535 487 54.1 12.1<br />

trary, feeding the mycelium with ammonium resulted in a rapid decrease of<br />

GDH transcripts, which correlated with a decline in GDH-specific activity.<br />

Addition of methionine sulfoximine (MSX), an inhibitor of the GS enzyme, to<br />

the ammonium-containing medium led to a depletion of glutamine and an<br />

accumulation of ammonium in the cells, while a significant decrease of GDH<br />

transcript occurred simultaneously (Javelle et al. 2003a). This result strongly<br />

suggests that in Hebeloma cylindrosporum, GDH repression is controlled by<br />

ammonium and not by glutamine, which is obviously different from what was<br />

found in Neurospora crassa (Chang and Marzluf 1979; Premakumar et al.<br />

1979), and very likely in Agaricus bisporus (Kersten et al. 1999), where gluta-


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 413<br />

mine or metabolites derived from this amino acid exerted nitrogen catabolite<br />

repression.<br />

In Pleurotus ostreatus, NADP-dependent glutamate dehydrogenase and<br />

glutamate synthase were not detected (Mikes et al. 1994). NAD-GDH was<br />

derepressed by ammonia and repressed by high concentrations of L-glutamate.<br />

This suggests that this enzyme obviously plays an active role in ammonium<br />

assimilation in Pleurotus ostreatus. However, a catabolic role of NAD-<br />

GDH in the deamination of L-glutamate, due to its very low Km for<br />

L-glutamate is not excluded (Mikes et al. 1994).<br />

6.2 Role and Properties of Glutamine Synthetase<br />

Glutamine synthetase (GS; EC 6.3.1.2) is the key enzyme involved in ammonium<br />

assimilation in <strong>plant</strong>s (Lea et al. 1990). GS catalyses the ATP-dependent<br />

condensation of NH 4 + with glutamate to produce glutamine. Plant GS is an<br />

octameric isozyme with a native molecular mass of approximately 320 or<br />

380 kDa depending on whether it is localized in the cytosol (GS 1 ) or in plastids/chloroplasts<br />

(GS 2; Lea et al.1990).<br />

The in vivo function of GS 2 has been elucidated using genetically modified<br />

barley <strong>plant</strong>s (Wallsgrove et al. 1987). The main role is assimilation of NH 4 +<br />

derived from nitrate reduction and photorespiration. The in vivo role of GS 1<br />

depends on the organ in which it is localized. In roots, GS 1 constitutes nearly<br />

all GS activity and the main role is assimilation of NH 4 + for translocation and<br />

biosynthesis (Lea et al. 1990).<br />

In gymnosperms, except in the nonconiferous gymnosperm Ginkgo, only<br />

cytosolic isoforms of GS (GS 1) have been identified (Suarez et al. 2002). The<br />

chloroplastic isoform (GS 2) has not yet been detected by using a number of<br />

different molecular approaches including separation of isoforms by ionexchange<br />

chromatography. This implies that in conifers, ammonium is assimilated<br />

in the cytosol and therefore, glutamine and glutamate biosynthesis<br />

occurs in separate compartments, the GOGAT enzyme being located within<br />

chloroplasts. Recent studies indicate the existence of a translocator in the<br />

chloroplast membranes of Pinus pinaster that may be responsible for the<br />

import of glutamine into the organelle, in antiport with glutamate (Suarez et<br />

al. 2002).<br />

It is generally assumed that GS activity in <strong>plant</strong>s is regulated at the transcriptional<br />

level, and many reports have focused on this aspect (Lam et al.<br />

1996; Oliveira et al. 1997). The dramatic light induction of mRNA for GS 2 is<br />

mediated in part by phytochrome and in part by light-induced changes in<br />

levels of sucrose (Oliveira and Coruzzi 1999), whereas the transcription of<br />

GS 1 in roots depends on the external nitrogen supply level ( Finnemann and<br />

Schjoerring 2000). Recent work suggests that GS 1 is not only regulated transcriptionally,<br />

but also post-translationally by reversible phosphorylation


414<br />

A. Javelle et al.<br />

catalysed by protein kinases and microcystin-sensitive serine/threonine protein<br />

phosphatase (Finneman and Schjoerring 2000). The more active form is<br />

phosphorylated, while the dephosphorylated enzyme is less active and is<br />

much more susceptible to degradation. Once phosphorylated, GS reaches its<br />

maximal activity through interaction with 14–3–3 proteins, a large group of<br />

binding proteins with multiple functions in all eukaryotes (Finneman and<br />

Schjoerring 2000). Such a post-translational modulation is similar to that<br />

found with nitrate reductase (Kaiser and Huber 2001). However, the activities<br />

of NR and GS 1 are oppositely affected by the reversible phosphorylation,<br />

as dephosphorylation activates NR, but deactivates GS 1. In addition, phosphorylated<br />

NR is an initial step in NR degradation, whereas phosphorylated<br />

GS 1 is more protected against degradation than dephosphorylated GS 1 .The<br />

phosphorylated status of GS 1 changes during light/dark transitions and<br />

depends in vitro on the ATP/AMP ratio. However, in leaves of Brassica napus,<br />

the phosphorylation level increased in darkness and decreased in light, suggesting<br />

that the enzyme plays a role in nitrogen remobilization (Finnemann<br />

and Schjoerring 2000).<br />

The enzyme was purified and studied from Douglas fir roots (Bedell et al.<br />

1995). The native enzyme had a molecular mass of 460 kDa and was composed<br />

of two different subunits of 54 and 64 kDa. The enzyme exhibited a negative<br />

cooperativity for ammonium with two Km values which were 11 and<br />

75 mM in the presence of ammonium concentrations lower and higher than<br />

1.3 mM, respectively (Bedell et al. 1995). This possibility for the enzyme to<br />

adjust its affinity to the level of ammonium is obviously a very efficient way to<br />

assimilate NH 4 + at different concentrations. However, the enzyme was not<br />

investigated after mycorrhization of the Douglas fir roots.<br />

In the fungus Pleurotus ostreatus, GS was derepressed by ammonium and<br />

L-glutamate, while repression of the enzyme was observed in the presence of<br />

L-glutamine (Mikes et al. 1994). This indicates a strong involvement of the<br />

enzyme in ammonium assimilation.<br />

GLNA, the gene encoding GS has been cloned and characterized from various<br />

fungi (Table 1), including mycorrhizal fungi. Moreover, GS has been purified<br />

from the ectomycorrhizal fungus Laccaria laccata (Brun et al. 1992). The<br />

native enzyme had a molecular weight of approximately 380 kDa and was<br />

composed of eight identical subunits of 42 kDa. The enzyme revealed a high<br />

affinity for NH 4 + (24 mM), contrasting with the low affinity of NADP-GDH for<br />

this cation (5 mM) in the same fungus. The GS enzyme also represented about<br />

3 % of the total soluble protein pool, which was considerably higher than<br />

NADP-GDH, which represented only 0.15 % (Brun et al. 1992).All these results<br />

strongly suggest that GS is likely to be the main route of ammonium assimilation<br />

in this fungus, especially at low NH 4 + concentrations.<br />

In ectomycorrhizal fungi, localization studies are more limited than in<br />

higher <strong>plant</strong>s. However, immunogold labelling of GS revealed a uniform distribution<br />

of the enzyme in the cytosol of Laccaria laccata cultivated in pure


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 415<br />

culture (Brun et al. 1993). In the association Douglas fir-Laccaria laccata,the<br />

fungal GS was uniformly detected over the entire section of the ectomycorrhizas<br />

where the fungal cells were present and no particular accumulation<br />

was detected in the mantle, or in the Hartig net fungal cells (Botton and<br />

Chalot 1995). The similar patterns of GS distribution observed in the free-living<br />

mycelia and in the ectomycorrhizal tissues suggest that the fungal enzyme<br />

plays an active role in the primary assimilation of ammonium in ectomycorrhizas.<br />

The expression level of the GS enzyme was studied by Javelle et al. (2003b)<br />

in the ectomycorrhizal fungus Hebeloma cylindrosporum, where a single<br />

mRNA of about 1.2 kb was detected. Transfer of the organism from ammonium-containing<br />

media to nitrogen-free media resulted in an increase of GS<br />

transcripts, correlating with an increase of GS activity. However, when the culture<br />

media were resupplemented with ammonium, up to the concentration of<br />

10 mM, GS transcripts remained almost unchanged or decreased very slowly,<br />

indicating that GS in this fungus is not highly regulated, although highly<br />

expressed (Javelle et al. 2003b; Fig. 2). Such a regulatory process at the transcriptional<br />

level has also been found in Agaricus bisporus (Kersten et al. 1997),<br />

while in Saccharomyces cerevisiae, the enzyme seems to be highly regulated at<br />

the post-transcriptional level (ter Schure et al. 1995).<br />

6.3 Role and Properties of Glutamate Synthase<br />

Three classes of glutamate synthases (GOGAT) have been defined, based on<br />

their amino acid sequences and the nature of the electron donor (Vanoni and<br />

Curti, 1999). (1) Bacterial NADPH-dependent GOGAT consists of two different<br />

subunits, the a-subunit of about 150 kDa and the b-subunit of about<br />

50 kDa; (2) Ferredoxin-dependent GOGAT found in photosynthetic cells<br />

(higher <strong>plant</strong>s, algae and cyanobacteria) is monomeric and shares considerable<br />

homology throughout its sequence with the a-subunit of bacterial<br />

GOGAT; (3) <strong>plant</strong>s (especially nonphotosynthetic cells) and fungi including<br />

yeasts, as well as lower animals contain a monomeric NAD(P)H-dependent<br />

GOGAT of about 200 kDa which results from the fusion of two fragments similar<br />

to the a and b-subunits of bacterial GOGAT.<br />

In <strong>plant</strong>s, both enzymes (NADH-GOGAT: EC 1.4.1.14. and ferredoxin (Fd)-<br />

GOGAT: EC 1.4.7.1) display different physico-chemical, immunological and<br />

regulatory properties and are encoded by separate genes (Ireland and Lea<br />

1999). Fd-GOGAT is an iron-sulphur monomeric flavoprotein, plastid-located<br />

and represents the predominant molecular form in photosynthetic tissues<br />

although its presence has also been reported in roots and nodules (Temple et<br />

al. 1998). In most <strong>plant</strong>s analysed, Fd-GOGAT is encoded by a single gene,<br />

however, in Arabidopsis, two genes have been characterized (Coschigano et al.<br />

1998). GLU1 is exclusively expressed in the leaf and is light-regulated, whereas


416<br />

A. Javelle et al.<br />

GLU2 is expressed in leaves and roots and is not regulated by light. The<br />

expression pattern of the genes and the physiological characterization of<br />

defective mutants support a role of GS 2 and Fd-GOGAT in the assimilation of<br />

ammonium derived from the reduction of nitrate and from photorespiration<br />

(Coschigano et al. 1998). NADH-GOGAT, also an iron-sulphur monomeric<br />

flavoprotein, is present at a low level in leaves, but is more abundant in nonphotosynthetic<br />

tissues such as roots and nodules, where it is located in<br />

nonchlorophyllous plastids (Temple et al. 1998). The structure of the alfalfa<br />

gene encoding NADH-GOGAT has been reported by these authors, and its<br />

expression is restricted to root nodules where it plays a significant role in the<br />

assimilation of ammonium derived from symbiotic N 2 fixation (Trepp et al.<br />

1999). The localization of GS 1 and NADH-GOGAT proteins in the root vascular<br />

bundles of rice, and very likely in many other <strong>plant</strong>s, supports the possibility<br />

of a co-ordinated function in the assimilation of ammonium in roots<br />

(Ishiyama et al. 1998).<br />

In fungi, NADH-GOGAT was purified and studied in Neurospora crassa<br />

where the enzyme was found as a single polypeptide of 200 kDa (Hummelt<br />

and Mora 1980) and in Saccharomyces cerevisiae where the enzyme is<br />

trimeric, composed of three identical 199-kDa subunits (Cogoni et al. 1995).<br />

In ectomycorrhizal fungi, very little is known about this enzyme. An<br />

NADH-dependent GOGAT was, however, detected in Laccaria bicolor by Vézina<br />

et al. (1989). In Pisolithus tinctorius, the kinetics of 15 N labelling and the<br />

effects of enzyme inhibitors have given evidence that ammonium assimilation<br />

occurs through the GS/GOGAT cycle (Kershaw and Stewart 1992). In<br />

agreement with these data, Botton and Dell (1994) failed to detect NADP-<br />

GDH in this fungus. In Scleroderma verrucosum, glutamine synthetase and<br />

NAD-glutamate synthase activities were clearly detected, while NADP-GDH<br />

was almost undetectable (Prima Putra et al. 1999). This is consistent with the<br />

view that ammonium assimilation occurs through the GS/GOGAT cycle in<br />

this fungus. In Cenococcum geophilum, a number of results based on the use<br />

of enzyme-specific inhibitors, enzyme assays and estimation of the amino<br />

acid pools are also consistent with the operation of the GS/GOGAT cycle (A.<br />

Khalid and B. Botton, unpublished results).<br />

The results obtained by Chalot et al. (1994a) with Paxillus involutus, also<br />

emphasize a GS/GOGAT cycle in this fungus. Indeed, feeding the fungus with<br />

[ 14 C]-glutamine resulted in a significant labelling of glutamate, while addition<br />

of azaserine, an inhibitor of the GOGAT enzyme, led to both an accumulation<br />

of 14 C-glutamine and a reduced pool of labelled glutamate. Interestingly, in<br />

these experiments, 14 C-aspartate and 14 C-alanine did not accumulate under<br />

azaserine treatment where 14 C-glutamine degradation was inhibited, thus<br />

indicating that aspartate and alanine synthesis depends on the carbon skeletons<br />

from glutamine (Chalot et al.1994a). In addition, feeding Paxillus involutus<br />

with 14 C-glutamate resulted in a significant accumulation of 14 C-glutamine<br />

under azaserine treatment, suggesting that the supplied glutamate is used for


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 417<br />

glutamine synthesis. These results are consistent with the existence of two<br />

pools of glutamate in the fungal cells, as previously demonstrated by<br />

[ 15 N]amino acid analysis in Cenococcum geophilum (Martin et al. 1988). It was<br />

thus suggested that newly absorbed glutamate, as well as glutamate synthetized<br />

by NADP-GDH are converted to glutamine, whereas glutamate produced<br />

by the GOGAT enzyme is utilized by the aminotransferases (Martin et<br />

al. 1988; Botton and Chalot 1995).<br />

The glutamine synthetase–glutamate synthase pathway was shown to be<br />

the main assimilatory route in beech ectomycorrhizas and glutamate dehydrogenase<br />

plays only a minor role, if any, in these tissues (Martin et al. 1986).<br />

Glutamine synthetase and glutamate synthase which share immunological<br />

similarities with higher <strong>plant</strong> enzymes were detected in beech ectomycorrhizas<br />

by means of Western immunoblotting, whereas a fungal glutamate<br />

dehydrogenase could not be detected (Martin, unpubl. results). The absorption<br />

of NH 4 + is associated with glutamine synthesis in beech ectomycorrhizas<br />

so that 60–80 % of the nitrogen absorbed is present as this amide after a few<br />

hours of absorption (Martin et al. 1986). In addition, there is a rapid and very<br />

high 15 N-labelling in alanine over the time course of the experiment performed<br />

with beech (Martin et al. 1986). These data, together with the measurement<br />

of high alanine aminotransferase activity in ectomycorrhizal fungi<br />

(Dell et al. 1989), suggest that glutamine and alanine might be the major forms<br />

of combined nitrogen exported to the root cells.<br />

7 Amino Acid Metabolism<br />

7.1 Utilization of Proteins by Ectomycorrhizal Fungi and<br />

Ectomycorrhizas<br />

As investigated primarily by Lundeberg (1970), it is generally accepted that<br />

most ectomycorrhizal fungal strains are unable to metabolize and use humusbound<br />

nitrogen. Several ectomycorrhizal and ericaceous fungi in pure culture<br />

are, however, able to grow in nutrient media containing proteins as the sole<br />

nitrogen source (Bajwa et al. 1985; Abuzinadah and Read 1986a), and this correlated<br />

with the production of exocellular proteinase activities (Botton and<br />

Chalot 1991; Leake and Read 1991). In the presence of exogenous proteins<br />

(casein, gelatin, albumin, soil proteins), Cenococcum geophilum was able to<br />

secrete active proteases into the nutrient medium, and ammonia strongly<br />

repressed the induction and secretion of these proteases (El-Badaoui and Botton<br />

1989). This capacity of the mycorrhizal fungus to use amino acids as<br />

nitrogen sources is retained in the symbiotic state. Melin and Nilsson (1953)<br />

have shown that 15 N from [ 15 N]glutamate is transferred to Pinus sylvestris<br />

roots and aerial parts through the mycelia of Suillus granulatus. The ability of<br />

several ectomycorrhizal fungi to assimilate proteins and to transfer its nitro-


418<br />

A. Javelle et al.<br />

gen to <strong>plant</strong>s of Pinus contorta was also clearly demonstrated (Abuzinadah<br />

and Read 1986a, b; Abuzinadah et al.1986). The use of nitrogen sources not<br />

available to nonmycorrhizal <strong>plant</strong>s contributes, therefore, to an increased<br />

uptake of nitrogen by infected roots.<br />

7.2 Amino Acids Used as Nitrogen and Carbon Sources<br />

Utilization of amino acids by ectomycorrhizal symbionts and ectomycorrhizas<br />

may have important implications, not only for their nitrogen metabolism,<br />

but also for the overall carbon economy of the <strong>plant</strong>. Axenic mycelia of<br />

the ectomycorrhizal basidiomycete Suillus bovinus have been grown in liquid<br />

media in the presence of glucose as the only carbohydrate source and under<br />

such conditions, they produced similar amounts of dry weight with ammonia,<br />

with nitrate or with alanine, 60–80 % more with glutamate or glutamine, but<br />

about 35 % less with urea as the only exogenous nitrogen source (Grotjohann<br />

et al. 2000).<br />

Recently, the fate of carbon derived from alanine, glutamate and glutamine<br />

was investigated in the ectomycorrhizal fungus Paxillus involutus (Chalot et<br />

al. 1994a, b). The result of the 14 C tracer experiments suggested that the carbon<br />

skeletons derived from newly absorbed glutamate were mainly used for<br />

the synthesis of glutamine. The accumulation of [ 14 C]glutamate and the<br />

marked decrease of [ 14 C]glutamine under MSX treatment were consistent<br />

with a rapid utilization of glutamate by the glutamine synthetase (GS)<br />

enzyme. The newly absorbed, as well as the newly synthesized [ 14 C]glutamine<br />

were degraded into [ 14 C]glutamate, suggesting the operation of the glutamate<br />

synthase (GOGAT) enzyme. This was confirmed by the striking accumulation<br />

of [ 14 C]glutamine when the fungus was cultivated in the presence of azaserine,<br />

an inhibitor of GOGAT. In addition, a strong inhibition of glutamine utilization<br />

by aminooxyacetate indicated that glutamine catabolism in Paxillus<br />

involutus might involve a transamination process as an alternative pathway to<br />

GOGAT for glutamine degradation (Chalot et al. 1994a).<br />

The use of 14 C-labelled amino acids also showed a direct involvement of<br />

glutamate and glutamine in the respiration pathways, these amino acids being<br />

obviously channelled through the tricarboxylic acid (TCA) cycle and oxidized<br />

to CO 2. Feeding the fungus with [ 14 C]alanine resulted in a rapid labelling of<br />

pyruvate, citrate, succinate, fumarate and CO 2. Further labelling was detected<br />

in glutamate, glutamine and aspartate. The presence of aminooxyacetate completely<br />

suppressed 14 CO 2 evolution and decreased the flow of carbon to the<br />

Krebs cycle intermediates and amino acids, suggesting that alanine aminotransferase<br />

plays a key role in metabolizing alanine in Paxillus involutus<br />

(Chalot et al. 1994b).<br />

It has been shown by measuring enzyme capacities and metabolite pools<br />

that mycorrhization causes a re-arrangement of the main metabolic pathways


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 419<br />

in the very early stages following contact between the two partners (Blaudez<br />

et al. 1998), which correlates with the observed structural changes (Brun et al.<br />

1995). The impact of inoculation with Paxillus involutus on the utilization of<br />

organic carbon compounds by birch roots was studied by feeding [ 14 C]glutamate<br />

or [ 14 C]malate to the partners of the symbiosis, separately or in association,<br />

and by monitoring the subsequent distribution of 14 C (Blaudez et al.<br />

2001). Inoculation increased [ 14 C]glutamate and [ 14 C]malate absorption<br />

capacities by up to 8 and 17 times, respectively. This heterotrophic carbon<br />

assimilation by mycorrhizal birch has been estimated using 14 C-labelled proteins<br />

(Abuzinadah and Read 1989). The authors calculated that 9 % of <strong>plant</strong> C<br />

may be derived from proteins. Moreover, our results demonstrated that inoculation<br />

strongly modified the fate of [ 14 C]glutamate and [ 14 C]malate. It was<br />

demonstrated that exogenously supplied glutamate and malate might serve as<br />

C skeletons for amino acid synthesis in mycorrhizal birch roots and in the<br />

free-living fungus. Glutamine was the major 14 C-sink in mycorrhizal roots<br />

and in the free-living P. involutus (Blaudez et al. 2001). In contrast, citrulline<br />

and insoluble compounds were the major 14 C sinks in nonmycorrhizal roots,<br />

whatever the 14 C source. Thus, it is obvious that mycorrhiza formation leads to<br />

a profound alteration of the metabolic fate of exogenously supplied C compounds.<br />

Translocation through the hyphal network and further transfer of nutrients<br />

from fungus to host root has also been discussed in detail (Smith and<br />

Read 1997), but the intimate anatomical connections between fungal and root<br />

cells presents considerable technical difficulties for unambiguous experimental<br />

investigations of nutrient transfer between fungus and host.<br />

8 Conclusion and Future Prospects<br />

After many decades of investigating the anatomical, physiological and biochemical<br />

features of ectomycorrhizas, recent years have brought new insights<br />

at the molecular level. Considerable knowledge has been gained over the last<br />

10 years on the molecular characteristics and molecular regulation of the<br />

transporters and the nitrogen-assimilating enzymes in higher <strong>plant</strong>s and<br />

fungi, as well as in ectomycorrhizas. This research has greatly contributed to<br />

our understanding of how organic and inorganic nitrogen is taken up by the<br />

cells and assimilated in the organisms. However, the available information is<br />

still limited and efforts should be made to increase basic research on nitrogen<br />

metabolism and to integrate new advances in biotechnology.<br />

A current focus in <strong>plant</strong> improvement is the modification of the expression<br />

of genes involved in metabolism. Recent studies have shown that important<br />

characteristics can be introduced in transgenic herbaceous <strong>plant</strong>s by the<br />

expression of heterologous GS isoenzymes. Thus, a higher capacity for photorespiration<br />

(Migge et al. 2000), and increase in tolerance to salt stress


420<br />

A. Javelle et al.<br />

(Hoshida et al. 2000), have been reported using engineered <strong>plant</strong>s which overexpress<br />

chloroplastic GS 2 . Furthermore, an increase in growth has been<br />

observed in leguminous <strong>plant</strong>s, which overexpress cytosolic GS 1 (Limami et<br />

al. 1999). The modification of N assimilation efficiency has recently been<br />

approached in trees by the overexpression of pine GS 1 in a hybrid poplar (Gallardo<br />

et al. 1999). Poplar is considered as a model in molecular investigations<br />

because of its small genome size, easy vegetative propagation and the possibility<br />

of in vitro culture, and its amenability to transformation via Agrobacterium<br />

tumefaciens (Gallardo et al. 1999).<br />

Considerable knowledge has been gained over the last decade on the molecular<br />

characteristics and molecular regulation of N-assimilating enzymes in<br />

woody <strong>plant</strong>s, including angiosperm and gymnosperm species. This research<br />

has greatly contributed to our understanding of how inorganic N is assimilated<br />

and utilized in trees. However, the available information is still limited<br />

and efforts should be made to increase basic research on N metabolism and to<br />

integrate new advances in biotechnology to improve growth and development<br />

of economically important woody species. Although all new studies will contribute<br />

to this goal, the concentration of efforts in model trees, such as poplar<br />

for angiosperms and pine for gymnosperms, is advisable. In future years, the<br />

availability of new molecular tools for biological studies of trees will permit<br />

characterization of new genes involved in N metabolism and determination<br />

of their specific physiological roles. Functional studies are now possible in<br />

woody <strong>plant</strong>s because routine transformation protocols via Agrobacterium<br />

are available for poplar and rapid progress has been reported in the last few<br />

years for conifers. The use of somatic embryogenic cell lines is critical for the<br />

generation of transgenic trees. For example, genomic technologies have<br />

recently been used to study the effect of a variety of N regimes on <strong>plant</strong><br />

metabolism (Wang et al. 2000). Results from this study indicate that changes<br />

in N supply influence not only expression of genes involved in N assimilation,<br />

but also those involved in other metabolic pathways. Similar studies of gene<br />

expression at the organ or tissue levels are now feasible in tree models with<br />

the existence of EST databases from poplar.<br />

Another promising line of research will be to study at the molecular level,<br />

the genetic basis of important traits, such as N use efficiency and growth efficiency<br />

in the presence of the mycorrhizal fungus. Genetic maps for poplar<br />

and pine have been established and now genes involved in N metabolism can<br />

be localized in the genome. The possible association of specific genes with<br />

quantitative trait loci (QTL) are currently being investigated in a number of<br />

laboratories. This will allow molecular characterization of gene clusters<br />

involved in traits of interest in forestry and tree management.<br />

Transformation of ectomycorrhizal fungi is more limited. Indeed, the<br />

assignment of functions to genes and their products has been limited to<br />

deduction based on sequence homologies, subcellular localization studies<br />

and expression in heterologous hosts, since transformation techniques for the


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 421<br />

vast majority of ectomycorrhizal basidiomycetes have not been readily available.<br />

Exceptions are Laccaria laccata (Barret et al. 1990), and Hebeloma cylindrosporum<br />

(Marmeisse et al. 1992), which have been transformed by the protoplast<br />

method, and Paxillus involutus (Bills et al. 1995), and Laccaria bicolor<br />

(Bills et al. 1999), which have been transformed by particle bombardment.<br />

Since the first report on successful genetic transfer from Agrobacterium tumefaciens<br />

to the yeast Saccharomyces cerevisiae (Bundock et al. 1995), a number<br />

of ascomycetous filamentous fungi were also shown to be amenable to this<br />

transformation system (Abuodeh et al. 2000; Chen et al. 2000).<br />

Our understanding of metabolite regulation of gene expression supports<br />

the notion that ammonium and N-assimilation products such as amino acids<br />

might act as signals whose levels are sensed as an indicator for a high internal<br />

N status. Along these lines, putative sensors of glutamate in <strong>plant</strong>s, glutamate<br />

receptor genes, have been identified in Arabidopsis (Lam et al. 1998). The<br />

emerging tools of genomics and bioinformatics should allow us, in the near<br />

future, to identify the interacting pathways that control gene expression in<br />

response to mycorrhization.<br />

References and Selected Reading<br />

Abuodeh RO, Orbach MJ, Mandel MA, Das A, Galgiani JN (2000) Genetic transformation<br />

of Coccidioides immitis facilitated by Agrobacterium tumefaciens. J Infect Dis<br />

181:2106–2110<br />

Abuzinadah RA, Read DJ (1986a) The role of proteins in the nitrogen nutrition of ectomycorrhizal<br />

<strong>plant</strong>s. I. Utilization of peptides and proteins by ectomycorrhizal fungi.<br />

New Phytol 103:481–493<br />

Abuzinadah RA, Read DJ (1986b) The role of proteins in the nitrogen nutrition of ectomycorrhizal<br />

<strong>plant</strong>s. III. Protein utilization by Betula pendula and Pinus mycorrhizal<br />

association with Hebeloma cylindrosporum. New Phytol 103:506–514<br />

Abuzinadah RA, Read DJ (1988) Amino acids as nitrogen sources for ectomycorrhizal<br />

fungi: utilization of individual amino acids. Trans Br Mycol Soc 91:473–479<br />

Abuzinadah RA, Read DJ (1989) Carbon transfer associated with assimilation of organic<br />

nitrogen sources by silver birch (Betula pendula Roth.). Trees 3:17–23<br />

Abuzinadah RA, Finlay RD, Read DJ (1986) The role of proteins in the nitrogen nutrition<br />

of ectomycorrhizal <strong>plant</strong>s. II. Utilization of proteins by mycorrhizal <strong>plant</strong>s of Pinus<br />

contorta. New Phytol 103:495–506<br />

Ahmad I, Hellebust JA (1991) Enzymology of nitrogen assimilation in mycorrhiza. In:<br />

Norris JR, Read DJ,Varma AK (eds) Methods in <strong>microbiology</strong> no 23, Academic Press,<br />

New York, pp 181–202<br />

Ahmad I, Carleton TJ, Malloch DW, Hellebust JA (1990) Nitrogen metabolism in the<br />

ectomycorrhizal fungus Laccaria bicolor (R. Mre) Orton. New Phytol 116:431–441<br />

Anderson IC, Chambers SM, Cairney JWG (1999) Intra- and interspecific variation in<br />

patterns of organic and inorganic nitrogen utilisation by three Australian Pisolithus<br />

species. Mycol Res 103:1579–1587<br />

Aouadj R, Es-Sgaouri A, Botton B (2000) Etude de la stabilité et de quelques propriétés de<br />

la nitrate réductase du champignon ectomycorrhizien Pisolithus tinctorius.Cryptog<br />

Mycol 21:187–202


422<br />

A. Javelle et al.<br />

Bajwa R, Abuarghub S, Read DJ (1985) The biology of mycorrhiza in the Ericaceae. X.<br />

The utilization of proteins and the production of proteolytic enzymes by the mycorrhizal<br />

endophyte and by mycorrhizal <strong>plant</strong>s. New Phytol 101:469–486<br />

Barret V, Dixon RK, Lemke PA (1990) Genetic transformation of a mycorrhizal fungus.<br />

Appl Microbiol Biotechnol 33:313–316<br />

Bedell J-P, Chalot M, Brun A, Botton B (1995) Purification and properties of glutamine<br />

synthetase from Douglas-fir roots. Physiol Plant 94:597–604<br />

Bending GD, Read DJ (1995) The structure and function of the vegetative mycelium of<br />

ectomycorrhizal <strong>plant</strong>s. V. Foraging behaviour and translocation of nutrients from<br />

exploited litter. New Phytol 130:401–409<br />

Bills SN, Richter DL, Podila GK (1995) Genetic transformation of the ectomycorrhizal<br />

fungus Paxillus involutus by particle bombardment. Mycol Res 99:557–561<br />

Bills SN, Podila GK, Hiremath ST (1999) Genetic engineering of the ectomycorrhizal fungus<br />

Laccaria bicolor for use as a biological control agent. Mycologia 91:237–242<br />

Blatt MR, Maurousset L, Meharg A (1997) High-affinity NO 3 – H + co-transport in the fungus<br />

Neurospora: induction and control by pH and membrane voltage. J Membr Biol<br />

160:59–76<br />

Blaudez D, Chalot M, Dizengremel P, Botton B (1998) Structure and function of the ectomycorrhizal<br />

association between Paxillus involutus and Betula pendula. II. Metabolic<br />

changes during mycorrhiza formation. New Phytol 138:543–552<br />

Blaudez D, Botton B, Dizengremel P, Chalot M (2001) The fate of [ 14 C]glutamate and<br />

[ 14 C]malate in birch roots is strongly modified under inoculation with Paxillus involutus.<br />

Plant Cell Environ 24:449–457<br />

Botton B, Chalot M (1991) Techniques for the studies of nitrogen metabolism in ectomycorrhiza.<br />

In: Norris JR, Read DJ,Varma AK (eds) Methods in <strong>microbiology</strong> no 23,Academic<br />

Press, New York, pp 204–244<br />

Botton B, Dell B (1994) Expression of glutamate dehydrogenase and aspartate aminotransferase<br />

in Eucalypt ectomycorrhizas. New Phytol 126:249–257<br />

Botton B, Chalot M (1995) Nitrogen assimilation: enzymology in ectomycorrhizas. In:<br />

Varma AK, Hock B (eds) Mycorrhiza, structure, function, molecular biology and biotechnology.<br />

Springer, Berlin Heidelberg New York, pp 325–363<br />

Brandes B, Godbold DL, Kuhn AJ, Jentschke G (1998) Nitrogen and phosphorus acquisition<br />

by the mycelium of the ectomycorrhizal fungus Paxillus involutus and its effect<br />

on host nutrition. New Phytol 140:735–743<br />

Brun A, Chalot M, Botton B, Martin F (1992) Purification and characterization of glutamine<br />

synthetase and NADP-glutamate dehydrogenase from the ectomycorrhizal fungus<br />

Laccaria laccata. Plant Physiol 99:938–944<br />

Brun A, Chalot M, Botton B (1993) Glutamate dehydrogenase and glutamine synthetase<br />

of the ectomycorrhizal fungus Laccaria laccata: occurrence and immunogold localization<br />

in the free living mycelium. Life Sci Adv Plant Physiol 12:53–60<br />

Brun A, Chalot M, Finlay RD, Söderström B (1995) Structure and function of the ectomycorrhizal<br />

association between Paxillus involutus (Batsch) Fr. and Betula pendula<br />

(Roth.). I. Dynamics of mycorrhiza formation. New Phytol 129:487–493<br />

Bundock P, den Dulk-Ras A, Beijersbergen A, Hoykaas PJJ (1995) Trans-kingdom T-DNA<br />

transfer from Agrobacterium tumefasciens to Saccharomyces cerevisiae. EMBO J<br />

14:3206–3214<br />

Burgstaller W (1997) Transport of small ions and molecules through the plasma membrane<br />

of filamentous fungi. Crit Rev Microbiol 23:1–46<br />

Caddick MX, Peters D, Platt A (1994) Nitrogen regulation in fungi.Antonie van Leeuwenhoek<br />

65:169–177<br />

Carleton TJ, Read DJ (1991) Ectomycorrhizas and nutrient transfer in conifer-feather<br />

moss ecosystems. Can J Bot 69:778–785


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 423<br />

Chalot M, Brun A (1998) Physiology of organic nitrogen acquisition by ectomycorrhizal<br />

fungi and ectomycorrhizas. FEMS Microbiol Rev 22:21–44<br />

Chalot M, Brun A, Botton B (1990) Occurrence and distribution of the fungal NADPdependent<br />

glutamate dehydrogenase in spruce and beech ectomycorrhizas. In:<br />

Werner D, Müller P (eds) Fast growing trees and nitrogen fixing trees. Gustav Fischer<br />

Verlag, Jena, Germany, pp 324–327<br />

Chalot M, Stewart GR, Brun A, Martin F, Botton B (1991) Ammonium assimilation by<br />

spruce-Hebeloma sp. ectomycorrhizas. New Phytol 119:541–550<br />

Chalot M, Brun A, Finlay RD, Söderström B (1994a) Metabolism of [ 14 C]glutamate and<br />

[ 14 C]glutamine by the ectomycorrhizal fungus Paxillus involutus. Microbiology<br />

140:1641–1649<br />

Chalot M, Brun A, Finlay RD, Söderström B (1994b) Respiration of [ 14 C]alanine by the<br />

ectomycorrhizal fungus Paxillus involutus. FEMS Microbiol Lett 121:87–92<br />

Chalot M, Brun A, Botton B, Söderström B (1996) Kinetics, energetics and specificity of<br />

the general amino acid transporter from the ectomycorrhizal fungus Paxillus involutus.<br />

Microbiology 142:1749–1756<br />

Chalot M, Javelle A, Blaudez D, Lambilliote R, Cooke R, Sentenac H, Wipf D, Botton B<br />

(2002) An update on nutrient transport processes in ectomycorrhizas. Plant Soil<br />

244:165–175<br />

Chang L, Marzluf GA (1979) Nitrogen regulation of uricase synthesis in Neurospora<br />

crassa. Mol Gen Genet 176:385–392<br />

Chen X, Stone M, Schlagnhaufer C, Romaine CP (2000) A fruiting body tissue method for<br />

efficient Agrobacterium-mediated transformation of Agaricus bisporus.Appl Environ<br />

Microbiol 66:4510–4513<br />

Cogoni C, Valenzuela L, Gonzales-Halphen D, Oliveira H, Macino G, Ballario P, Gonzalez<br />

A (1995) Saccharomyces cerevisiae has a single glutamate synthase gene coding for a<br />

<strong>plant</strong>-like high-molecular-weight polypeptide. J Bacteriol 177:792–798<br />

Coschigano KT, Melo-Oliveira R, Lim J (1998) Arabidopsis glts mutants and distinct Fdx-<br />

GOGAT genes: implications for photorespiration and primary nitrogen assimilation.<br />

Plant Cell 10:741–752<br />

Crawford NM, Glass AD (1998) Molecular and physiological aspects of nitrate uptake in<br />

<strong>plant</strong>s. Trends Plant Sci 3:389–395<br />

Dell B, Botton B, Martin F, Le Tacon F (1989) Glutamate dehydrogenases in ectomycorrhizas<br />

of spruce (Picea excelsa L.) and beech (Fagus sylvatica L.). New Phytol 111:<br />

683–692<br />

Dubois E, Grenson M (1979) Methylamine ammonia uptake systems in Saccharomyces<br />

cerevisiae. Multiplicity and regulation. Mol Gen Genet 175:67–76<br />

El-Badaoui K, Botton B. 1989. Production and characterization of exocellular proteases<br />

in ectomycorrhizal fungi. Ann Sci For 46:728–730<br />

Fergusson AR, Sims AP (1971) Inactivation in vivo of glutamine synthetase and NADspecific<br />

glutamate dehydrogenase: its role in the regulation of glutamine synthesis in<br />

yeasts. J Gen Microbiol 69:423–427<br />

Finlay RD, Ek H, Odham G, Söderström B (1988) Mycelial uptake, translocation and<br />

assimilation of nitrogen from 15 N-labelled ammonium by Pinus sylvestris <strong>plant</strong>s<br />

infected with four different ectomycorrhizal fungi. New Phytol 110:59–66<br />

Finnemann J, Schjoerring JK (2000) Post-translational regulation of cytosolic glutamine<br />

synthetase by reversible phosphorylation and 14–3–3 protein interaction. Plant J<br />

24:171–177<br />

Forde B (2000) Nitrate transporters in <strong>plant</strong>s: structure, function and regulation.<br />

Biochim Biophys Acta 1465:219–235<br />

Gallardo F, Fu J, Cantón FR, García-Gutiérrez A, Cánovas FM, Kirby EG (1999) Expression<br />

of a conifer glutamine synthetase gene in transgenic poplar. Planta 210:19–26


424<br />

A. Javelle et al.<br />

Garnier A, Berredjem A, Botton B (1997) Purification and characterization of the NADdependent<br />

glutamate dehydrogenase in the ectomycorrhizal fungus Laccaria bicolor<br />

(Maire) Orton. Fungal Genet Biol 22:168–176<br />

Garrett RH, Nason A (1969) Further purification and properties of Neurospora nitrate<br />

reductase. J Biol Chem 244:2870–2882<br />

Gazzarrini S, Lejay L, Gojon A, Ninnemann O, Frommer WB, von Wiren N (1999) Three<br />

functional transporters for constitutive, diurnally regulated, and starvation-induced<br />

uptake of ammonium into Arabidopsis roots. Plant Cell 11:937–947<br />

Gobert A, Plassard C (2002) Differential NO 3 – dependent patterns of NO3 – uptake in<br />

Pinus pinaster, Rhizopogon roseolus and their ectomycorrhizal association. New Phytol<br />

154:509–516<br />

Grotjohann N, Kowallik W, Huang Y, Schulte in den Baumen A (2000) Investigations into<br />

enzymes of nitrogen metabolism of the ectomycorrhizal basidiomycete, Suillus bovinus.<br />

Z Naturforsch C, 55:203–212<br />

Guan C, Ribeiro A, Akkermans ADL, Jing Y, van Kammen A, Bisseling T, Pawlowski K<br />

(1996) Nitrogen metabolism in actinorhizal nodules of Alnus glutinosa: expression of<br />

glutamine synthetase and acetylornithine transaminase. Plant Mol Biol 32:1177–1184<br />

Hackette SL, Skye GE, Burton C, Segel IH (1970) Characterization of an ammonium<br />

transport system in filamentous fungi with methylammonium- 14 C as the substrate. J<br />

Biol Chem 245:4241–4250<br />

Harley JL (1989) The significance of mycorrhizas. Mycol Res 92:129–139<br />

Hildebrandt U, Schmelzer E, Bothe H (2002) Expression of nitrate transporter genes in<br />

tomato colonized by an arbuscular mycorrhizal fungus. Physiol Plant 115:125–136<br />

Ho I, Trappe JM (1987) Enzymes and growth substances of Rhizopogon species in relation<br />

to mycorrhizal host and infrageneric taxonomy. Mycologia 79:553–558<br />

Hoshida H, Tanaka Y, Hibino T, Hayashi Y, Tanaka A, Takabe T, Takabe T (2000) Enhanced<br />

tolerance to salt stress in transgenic rice that overexpresses chloroplast glutamine<br />

synthetase. Plant Mol Biol 43:103–111<br />

Howitt SM, Udvardi MK (2000) Structure, function and regulation of ammonium transporters<br />

in <strong>plant</strong>s. Biochim Biophys Acta 1465:152–170<br />

Hummelt G, Mora J (1980) Regulation and function of glutamate synthase. Biochem Biophys<br />

Res Commun 96:1688–1694<br />

Ireland RJ, Lea PJ (1999) The enzymes of glutamine, glutamate, asparagine, and aspartate<br />

metabolism. In: Singh BK (ed) Plant amino acids, biochemistry and biotechnology.<br />

Marcel Dekker, New York, pp 49–109<br />

Ishiyama K, Hayakawa T, Yamaya T (1998) Expression of NADH-dependent glutamate<br />

synthase protein in the epidermis and exodermis of rice root in response to the supply<br />

of ammonium ions. Planta 204:288–294<br />

Jargeat P (1999) Caractérisation et manipulation génétique de la voie d’assimilation du<br />

nitrate du champignon symbiotique Hebeloma cylindrosporum. PhD Thesis, University<br />

Claude-Bernard, Lyon I, Lyon<br />

Jargeat P, Gay G, Debaud JC, Marmeisse R (2000) Transcription of a nitrate reductase<br />

gene isolated from the symbiotic basidiomycete fungus Hebeloma cylindrosporum<br />

does not require induction by nitrate. Mol Gen Genet 263:948–956<br />

Jargeat P, Rekangalt D, Verner MC, Gay G, Debaud JC, Marmeisse R, Fraissinet-Tacher L<br />

(2003) Characterisation and expression analysis of a nitrate transporter and nitrite<br />

reductase genes, two members of a gene cluster for nitrate assimilation from the symbiotic<br />

basidiomycete Hebeloma cylindrosporum. Curr Genet 43:199–205<br />

Javelle A, Chalot M, Söderström B, Botton B (1999) Ammonium and methylamine transport<br />

by the ectomycorrhizal fungus Paxillus involutus and ectomycorrhizas. FEMS<br />

Microbiol Ecol 30:355–366


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 425<br />

Javelle A, Rodriguez-Pastrana BR, Jacob C, Botton B, Brun A,André B, Marini AM, Chalot<br />

M (2001) Molecular characterization of two ammonium transporters from the ectomycorrhizal<br />

fungus Hebeloma cylindrosporum. FEBS Lett 505:393–398<br />

Javelle A, André B, Marini A, Chalot M (2003a) High affinity ammonium transporters<br />

and nitrogen sensing in mycorrhizas. Trends Microbiol 11:53–55<br />

Javelle A, Morel M, Rodriguez-Pastrana BR, Botton B, André B, Marini AM, Brun A,<br />

Chalot M (2003b) Molecular characterization, function and regulation of ammonium<br />

transporters (Amt) and ammonium-metabolizing enzymes (GS, NADP-GDH) in the<br />

ectomycorrhizal fungus Hebeloma cylindrosporum. Mol Microbiol 47:411–430<br />

Jentschke G, Brandes B, Kuhn AJ, Schröder WH, Godbold DL (2001) Interdependence of<br />

phosphorus, nitrogen, potassium and magnesium translocation by the ectomycorrhizal<br />

fungus Paxillus involutus. New Phytol 149:327–338<br />

Johnstone IL, McCabe PC, Greaves P, Gurr SJ, Cole GE, Brow MAD, Unkles SE, Clutterbuck<br />

AJ, Kinghorn JR, Innis MA (1990) Isolation and characterisation of the crnAniiA-niaD<br />

gene cluster for nitrate assimilation in Aspergillus nidulans. Gene<br />

90:181–192<br />

Jongbloed RH, Clement JMAM, Borst-Pauwels GWFH (1991) Kinetics of NH 4 + and K +<br />

uptake by ectomycorrhizal fungi. Effect of NH 4 + on K + uptake. Physiol Plant 83:427–<br />

432<br />

Kaiser WM, Huber SC (2001) Post-translational regulation of nitrate reductase: mechanism,<br />

physiological relevance and environmental triggers. J Exp Bot 363:1981–1989<br />

Kaldorf M, Schmelzer E, Bothe H (1998) Expression of maize and fungal nitrate reductase<br />

genes in arbuscular mycorrhiza. Mol Plant Microbe Interact 11:439–448<br />

Kershaw JC, Stewart GR (1992) Metabolism of 15 N-labelled ammonium by the ectomycorrhizal<br />

fungus Pisolithus tinctorius (Pers), Coker & Couch. Mycorrhiza 1:71–77<br />

Kersten MA, Muller Y, Op den Camp HJ,Vogels GD,Van Griensven LJ,Visser J, Schaap PJ<br />

(1997) Molecular characterization of the glnA gene encoding glutamine synthetase<br />

from the edible mushroom Agaricus bisporus. Biochim Biophys Acta 1428:260–272<br />

Kersten MA, Arninkhof MJ, Op den Camp HJ, Van Griensven LJ, van der Drift C (1999)<br />

Transport of amino acids and ammonium in mycelium of Agaricus bisporus. Biochim<br />

Biophys Acta 1428:260–272<br />

Kinghorn JR, Unkles SE (1994) Inorganic nitrogen assimilation: molecular aspects. In:<br />

Martinelli SD, Kinghorn JR (eds) Aspergillus: 50 years on. Elsevier, Amsterdam, pp<br />

181–194<br />

Kreuzwiezer J, Stulen I, Wiersema P,Vaalburg W, Rennenberg H (2000) Nitrate transport<br />

in Fagus-Laccaria mycorrhiza. Plant Soil 220:107–117<br />

Kronzucker HJ, Siddiqi MY, Glass ADM (1996) Kinetics of NH4 + uptake in spruce. Plant<br />

Physiol 110:773–779<br />

Lam HM, Coschigano KT, Oliveira IC, Melo-Oliveira R, Coruzzi GM (1996) The molecular-genetics<br />

of nitrogen assimilation into amino acids in higher <strong>plant</strong>s.Ann Rev Plant<br />

Physiol Plant Mol Biol 47:569–593<br />

Lam HM, Chiu J, Hsieh M, Meisel L, Oliveira I, Shin M, Coruzzi GM (1998) Glutamatereceptor<br />

genes in <strong>plant</strong>s. Nature 396:125–126<br />

Landeweert R, Hoffland E, Finlay RD, Kuyper TW, van Breemen N (2001) Linking <strong>plant</strong>s<br />

to rocks: ectomycorrhizal fungi mobilize nutrients from minerals. Trends Ecol Evol<br />

16:248–254<br />

Lea PJ, Blackwell RD, Chen FL, Hecht U (1990) Enzymes of nitrogen assimilation. In: Dey<br />

PM, Harborne JB (eds) Methods in <strong>plant</strong> biochemistry().Academic Press, London, pp<br />

257–276<br />

Leake JR, Read D (1991) Proteinase activity in mycorrhizal fungi. III. Effects of protein,<br />

protein hydrolysate, glucose and ammonium on production of extracellular proteinase<br />

by Hymenoscyphus ericae Read Korf and Kernan. New Phytol 117:309–318


426<br />

A. Javelle et al.<br />

Limami A, Phillipson B, Ameziane R, Pernollet N, Jiang Q, Roy R, Deleens E, Chaumont-<br />

Bonnet M, Gresshoff PM, Hirel B (1999) Does root glutamine synthetase control <strong>plant</strong><br />

biomass production in Lotus japonicus L.? Planta 209:495–502<br />

Lorillou S, Botton B, Martin F (1996) Nitrogen source regulates the biosynthesis of<br />

NADP-glutamate dehydrogenase in the ectomycorrhizal basidiomycete Laccaria<br />

bicolor. New Phytol 132:289–296<br />

Lundeberg G (1970) Utilization of various nitrogen sources, in particular bound soil<br />

nitrogen, by mycorrhizal fungi. Studia Forestalia Suecica 79:1–95<br />

Machin F, Perdomo G, Pérez MD, Brito N, Siverio JM (2000) Evidence for multiple nitrate<br />

uptake systems in Hansenula polymorpha. FEMS Microbiol Lett 194:171–174<br />

Marini AM, Vissers S, Urrestarazu A, André B (1994) Cloning and expression of the<br />

MEP1 gene encoding a transporter of ammonium in Saccharomyces cerevisiae.<br />

EMBO J 13:3456–3463<br />

Marini AM, Soussi-Boudekou S,Vissers S, André B (1997) A family of ammonium transporters<br />

in Saccharomyces cerevisiae. Mol Cell Biol 17:4282–4293<br />

Marschner H, Dell B (1994) Nutrient uptake in mycorrhizal symbiosis. Plant Soil<br />

159:89–102<br />

Marschner H, Haussling M, George E (1991) Ammonium and nitrate rates and rhizosphere<br />

pH in non-mycorrhizal roots of Norway spruce [Picea abies (L.) Karst]. Plant<br />

Soil 178:239–245<br />

Martin F (1985) 15 N-NMR studies of nitrogen assimilation and amino acid biosynthesis<br />

in the ectomycorrhizal fungus Cenococcum geophilum. FEBS Lett 182:350–354<br />

Martin F, Msatef Y, Botton B (1983) Nitrogen assimilation in mycorrhizas. I. Purification<br />

and properties of the nicotinamide adenine dinucleotide phosphate-specific glutamate<br />

dehydrogenase of the ectomycorrhizal fungus Cenococcum graniforme. New<br />

Phytol 93:415–422<br />

Martin F, Stewart GR, Genetet I, Le Tacon F (1986) Assimilation of 15 NH 4 by beech (Fagus<br />

sylvatica L.) ectomycorrhizas. New Phytol 102:85–94<br />

Martin F, Ramstedt M, Söderhäll K (1987) Carbon and nitrogen metabolism in ectomycorrhizal<br />

fungi and ectomycorrhizas. Biochimie 69:569–581<br />

Martin F, Stewart GR, Genetet I, Mourot B (1988) The involvement of glutamate dehydrogenase<br />

and glutamine synthetase in ammonia assimilation by the rapidly growing<br />

ectomycorrhizal ascomycete Cenococcum geophilum Fr. New Phytol 110:541–550<br />

Marmeisse R, Gay G, Debaud JC, Casselton LA (1992) Genetic transformation of the<br />

symbiotic basidiomycete fungus Hebeloma cylindrosporum. Curr Genet 22:41–45<br />

Marzluf GA (1997) Genetic regulation of nitrogen metabolism in the fungi. Microbiol<br />

Mol Biol Rev 61:17–32<br />

Melin E, Nilsson H (1952) Transport of labelled nitrogen from an ammonium source to<br />

pine seedling through mycorrhizal mycelium. Sven Bot Tidkr 46:281–285<br />

Melin E, Nilsson H (1953) Transfer of labelled nitrogen from glutamic acid to pine<br />

seedlings through the mycelium of Boletus variegatus (Sw.) Fr. Nature 171:134<br />

Migge A, Carrayol E, Hirel B, Becker TW (2000) Leaf-specific overexpression of plastidic<br />

glutamine synthetase stimulates the growth of transgenic tobacco seedlings. Planta<br />

210:252–260<br />

Mikes V, Zofall M, Chytil M, Fulnecek J, Schanel L (1994) Ammonia-assimilating<br />

enzymes in the basidiomycete fungus Pleurotus ostreatus. Microbiology 140:977–982<br />

Minagawa N, Yoshimoto A (1982) Purification and characterization of the assimilatory<br />

NADH-nitrate reductase of Aspergillus nidulans. J Biochem 91:761–774<br />

Montanini B, Moretto N, Soragni E, Percudani R, Ottonello S (2002) A high-affinity<br />

ammonium transporter from the mycorrhizal ascomycete Tuber borchii. Fungal<br />

Geneti Biol 36:22–34<br />

Näsholm T, Ekblad A, Nordin A, Giesler R, Högberg M, Högberg P (1998) Boreal forest<br />

<strong>plant</strong>s take up organic nitrogen. Nature 392:914–916


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 427<br />

Navarro MT, Guerra E, Fernandez E, Galvan A (2000) Nitrite reductase mutants as an<br />

approach to understanding nitrate assimilation in Chlamydomonas reinhardtii. Plant<br />

Physiol 122:283–290<br />

Nehls U, Kleber R, Wiese J, Hampp R (1999) Isolation and characterization of a general<br />

amino acid permease from the ectomycorrhizal fungus Amanita muscaria.New Phytol<br />

144:343–349<br />

Ninnemann O, Jauniaux JC, Frommer W (1994) Identification of a high affinity ammonium<br />

transporter from <strong>plant</strong>s. EMBO J 13:3464–3471<br />

Notton BA, Hewitt EJ (1978) Structure and properties of higher <strong>plant</strong> nitrate reductase,<br />

especially Spinacea oleracea. In: Hewitt EJ, Cuttings CV (eds) Nitrogen assimilation of<br />

<strong>plant</strong>s. Academic Press, New York, pp 227–244<br />

Oliveira IC, Coruzzi GM (1999) Carbon and amino acids reciprocally modulate the<br />

expression of glutamine synthetase in Arabidopsis. Plant Physiol 121:301–309<br />

Oliveira IC, Lam HM, Coschigano K, Melo-Oliveira R, Corruzi GM (1997) Moleculargenetic<br />

dissection of ammonium assimilation in Arabidopsis thaliana. Plant Physiol<br />

Biochem 35:185–198<br />

Pan H, Feng B, Marzluf GA (1997) Two distinct protein-protein interactions between the<br />

NIT2 and NMR regulatory proteins are required to establish nitrogen metabolite<br />

repression in Neurospora crassa. Mol Microbiol 26:721–729<br />

Pao SS, Paulsen IT, Saier MH (1998) Major facilitator superfamily. Microbiol Mol Biol Rev<br />

62:1–34<br />

Pateman JA (1969) Regulation of synthesis of glutamate dehydrogenase and glutamine<br />

synthetase in micro-organisms. Biochem J 115:769–775<br />

Perez MD, Gonzales C, Avila J, Brito N, Siverio JM (1997) The YNT1 gene encoding the<br />

nitrate transporter in the yeast Hansenula polymorpha is clustered with genes YNI1<br />

and YNR1 encoding nitrite reductase and nitrate reductase, and its disruption causes<br />

inability to grow in nitrate. Biochem J 321:397–403<br />

Perez-Moreno J, Read DJ (2000) Mobilization and transfer of nutrients from litter to tree<br />

seedlings via the vegetative mycelium of ectomycorrhizal <strong>plant</strong>s. New Phytol 145:<br />

301–309<br />

Plassard C, Mousain D, Salsac L (1984a) Mesure in vitro de l’activité nitrate réductase<br />

dans les thalles de Hebeloma cylindrosporum, champignon basidiomycète. Physiol<br />

Vég 22:67–74<br />

Plassard C, Mousain D, Salsac L (1984b) Mesure in vivo and in vitro de l’activité nitrite<br />

réductase dans les thalles de Hebeloma cylindrosporum, champignon basidiomycète.<br />

Physiol Vég 22:147–154<br />

Plassard C, Martin F, Mousain D, Salsac L (1986) Physiology of nitrogen assimilation by<br />

mycorrhiza. In: Gianinazzi S, Gianinazzi-Pearson V (eds) Les mycorhizes: physiologie<br />

et génétique. INRA, Paris, pp 111–120<br />

Plassard C, Barry D, Eltrop L, Mousain D (1994) Nitrate uptake in maritime pine (Pinus<br />

pinaster) and the ectomycorrhizal fungus Hebeloma cylindrosporum: effect of ectomycorrhizal<br />

symbiosis. Can J Bot 72:189–197<br />

Plassard C, Chalot M, Botton B, Martin F (1997) Le rôle des ectomycorhizes dans la nutrition<br />

azotée des arbres forestiers. Rev For Fr 49:82–98<br />

Premakumar RG, Sorger J, Gooden D (1979) Nitrogen metabolite repression of nitrate<br />

reductase in Neurospora crassa. J Bacteriol 137:1119–1126<br />

Prima Putra D, Berredjem A, Chalot M, Dell B, Botton B (1999) Growth characteristics,<br />

nitrogen uptake and enzyme activities of the nitrate-utilizing ectomycorrhizal fungus<br />

Scleroderma verrucosum. Mycol Res 103:997–1002<br />

Rawat SR, Silim SN, Kronzucler HJ, Siddiqi MY, Glass AD (1999) AtAMT1 gene expression<br />

and NH 4 + uptake in roots of Arabidopsis thaliana: evidence for regulation by<br />

root glutamine levels. Plant J 19:143–52


428<br />

A. Javelle et al.<br />

Ritchie RJ, Gibson J (1987) Permeability of ammonia and amines in Rhodobacter<br />

sphaeroides and Bacillus firmus. Arch Biochem Biophys 258:332–341<br />

Roldan JM,Verbelen J, Waren LB, Tokuiyasu K (1982) Intracellular localization of nitrate<br />

reductase in Neurospora crassa. Plant Physiol 70:872–874<br />

Roon RJ, Even HL, Dunlop P, Larimore FL (1975) Methylamine and ammonia transport<br />

in Saccharomyces cerevisiae. J Bacteriol 122:502–509<br />

Rousseau JVD, Sylvia DM, Fox AJ (1994) Contribution of ectomycorrhiza to the potential<br />

nutrient-absorbing <strong>surface</strong> of pine. New Phytol 128:639–644<br />

Scazzocchio C, Arst HN Jr (1989) Regulation of nitrate assimilation in Aspergillus nidulans.<br />

In: Wray JL, Kinghorn JR (eds) Molecular and genetic aspects of nitrate assimilation.<br />

Oxford University Press, Oxford, pp 314–363<br />

Scheromm P., Plassard C., Salsac L (1990) Effect of nitrate and ammonium nutrition on<br />

the metabolism of the ectomycorrhizal fungus Hebeloma cylindrosporum.New Phytol<br />

114:227–234<br />

Schwartz T, Kusnan MB, Fock HP (1991) The involvement of glutamate dehydrogenase<br />

and glutamine synthetase/glutamate synthase in ammonia assimilation by the basidiomycete<br />

fungus Stropharia semiglobata. J Gen Microbiol 137:2253–2258<br />

Siverio JM (2002) Assimilation of nitrate by yeasts. FEMS Microbiol Rev 26:277–284<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis. Academic Press, London, 521 pp<br />

Sokolovski SG, Meharg AA, Maathuis FJM (2002) Calluna vulgaris root cells show<br />

increased capacity for amino acid uptake when colonized with the mycorrhizal fungus<br />

Hymenoscyphus ericae. New Phytol 155:525–530<br />

Sophianopoulou V, Diallinas G (1995) Amino acid transporters of lower eucaryotes: regulation,<br />

structure and topogenesis. FEMS Microbiol Rev 16:53–75<br />

Su W, Huber SC, Crawford NM. 1996. Identification in vitro of a post-translational regulatory<br />

site in the hinge 1 region of Arabidopsis nitrate reductase. Plant Cell 8, 519–527<br />

Suarez MF, Avila C, Gallardo F, Canton FR, Garcia-Gutiérrez A, Claros MG, Canovas FM<br />

(2002) Molecular and enzymatic analysis of ammonium assimilation in woody<br />

<strong>plant</strong>s. J Exp Bot 370:891–904<br />

Taylor TM, Osborn JM (1995) The importance of fungi in shaping the paleoecosystem.<br />

Rev Palaeobot Palynol 90:249–262<br />

Temple SJ, Vance CP, Gantt JS (1998) Glutamate synthase and nitrogen assimilation.<br />

Trends Plant Sci 3:51–56<br />

ter Schure EG, Sillje HH, Raeven LJ, Boonstra J, Verkleij AJ, Verrips CT (1995) Nitrogenregulated<br />

transcription and enzyme activities in continuous cultures of Saccharomyces<br />

cerevisiae. Microbiology 141:1101–1108<br />

Trepp GB, Plank DW, Gantt S,Vance CP (1999) NADH-glutamate synthase in alfalfa root<br />

nodules. Immuno-cytochemical localization. Plant Physiol 119:829–837<br />

Unkles SE, Hawker C, Grieve EI, Campbell EI, Montague P, Kinghorn JR (1991) crnA<br />

encodes a nitrate transporter in Aspergillus nidulans. Proc Natl Acad Sci USA<br />

88:204–208<br />

Unkles SE, Zhou D, Siddiqi MY, Kinghorn JR, Glass ADM (2001) Apparent genetic redundancy<br />

facilitates ecological plasticity for nitrate transport. EMBO J 20:6246–6255<br />

Vallorani L, Polidori E, Sacconi C, Agostini D, Pierleoni R, Piccoli G, Zepa S, Stocchi V<br />

(2002) Biochemical and molecular characterization of NADP-glutamate dehydrogenase<br />

from the ectomycorrhizal fungus Tuber borchii. New Phytol 154:779–790<br />

Vanoni MA, Curti B (1999) Glutamate synthase: a complex iron-sulfur flavoprotein. Cell<br />

Mol Life Sci 55:617–638<br />

Vézina LP, Margolis HA, McAfee BJ, Delanay S(1989) Changes in the activity of enzymes<br />

involved with primary nitrogen metabolism due to ectomycorrhizal symbiosis on<br />

jack pine seedlings. Physiol Plant 75:55–62<br />

Vitousek PM ., Matson PA (1985) Causes of delayed nitrate production in two Indiana<br />

forests. Forest Sci 31:122–131


22 Nitrogen Transport and Metabolism in Mycorrhizal Fungi and Mycorrhizas 429<br />

Wallenda T, Read DJ (1999) Kinetics of amino acid uptake by ectomycorrhizal roots.<br />

Plant, Cell Environ 22:179–187<br />

Wallsgrove RM, Turner JC, Hall NP, Kendall AC, Bright SWJ (1987) Barley mutants lacking<br />

chloroplast glutamine synthetase. Biochemical and genetic analysis. Plant Physiol<br />

83:155–158<br />

Wang R, Guegler K, LaBrie ST, Crawford N (2000) Genomic analysis of a nutrient<br />

response in Arabidopsis reveals diverse expression patterns and novel metabolic and<br />

potential regulatory genes induced. Plant Cell 12:1491–1509<br />

Wipf D, Benjdia M, Tegeder M, Frommer WB (2002) Characterization of a general amino<br />

acid permease from Hebeloma cylindrosporum. FEBS Lett 528:119–24<br />

Yip KP, Kurtz I (1995) NH 3 permeability of principal cells and intercalated cells measured<br />

by confocal fluorescence imaging. Am J Physiol 269:545–50<br />

Zhou JJ, Trueman LJ, Boorer KJ, Theodoulou FL, Fordes BG, Miller AJ (2000) A high<br />

affinity fungal nitrate carrier with two transport mechanisms. J Biol Chem<br />

275:39894–39899


23 Visualisation of Rhizosphere Interactions of<br />

Pseudomonas and Bacillus Biocontrol Strains<br />

Thomas F.C. Chin-A-Woeng, Anastasia L. Lagopodi,<br />

Ine H.M. Mulders, Guido V. Bloemberg and Ben J.J. Lugtenberg<br />

1 Introduction<br />

This chapter provides hands-on protocols as well as theoretical background<br />

information for the selection of Pseudomonas and Bacillus strains from the<br />

rhizosphere antagonistic to phytopathogens. These strains can be evaluated<br />

in a bioassay for their beneficial properties. The strains can be marked with a<br />

reporter gene after selection and used to study cellular and molecular interactions<br />

between one or more beneficial strains and a soil-borne phytopathogen<br />

in the rhizosphere of a host <strong>plant</strong>.Autofluorescent proteins can be<br />

used for the non-invasive study of rhizosphere interactions using epifluorescence<br />

and confocal laser scanning microscopy (CLSM). Autofluorescent proteins<br />

have become an outstanding and convenient tool for studying rhizosphere<br />

and other in situ environmental interactions and have allowed<br />

microbiologists to visualise the spatial distribution of various microorganisms.<br />

Intricate molecular mechanisms and relationships in the rhizosphere<br />

can now be studied. Methods to mark rhizosphere bacteria as well as fungi are<br />

provided.<br />

2 Tomato Foot and Root Rot and the Need for Biological<br />

Control<br />

Tomato (Esculentum lycopersicum) foot and root rot caused by the fungus<br />

Fusarium oxysporum Schlechtend.:Fr. f. sp. radicis-lycopersici W.R. Jarvis and<br />

Shoemaker (F.o.r.l.) is a disease which causes considerable losses to tomato<br />

crops. The disease differs from fusarium wilt caused by Fusarium oxysporum<br />

Schlechtend.:Fr. f. sp. lycopersici (Sacc.) W.C. Snyder & H.N. Hans. Plants with<br />

Fusarium foot and root rot show yellowing along the margin of the oldest<br />

leaves, followed by necrosis. Dry brown lesions develop in the cortex of the tap<br />

or main lateral roots. Necrotic lesions may also develop on the <strong>surface</strong> of the<br />

stem from the soil line to 10–30 cm above it. Infected <strong>plant</strong>s may be stunted<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


432<br />

Thomas F.C. Chin-A-Woeng et al.<br />

and wilted. Cool soil temperatures favour the disease. The fungus lives over<br />

winter and survives for many years in the soil as chlamydospores. Long distance<br />

spread is caused by trans<strong>plant</strong>s and by soil on farm machinery. Spores<br />

are air-borne in greenhouses. The disease causes losses in tomato cropping in<br />

agricultural fields, glasshouses, and hydroponic growth. The fungus forms a<br />

problem for hydroponic tomato growth in glasshouses in the Netherlands. In<br />

the southwest of Florida it is one of the most important tomato diseases and<br />

it is emerging at new locations in the United States. Until now only partially<br />

resistant varieties have been identified and pre<strong>plant</strong> fumigation with, e.g.<br />

methylbromide, which is a management practice often used for many soilborne<br />

diseases, does not completely control the fungus. This practice is also<br />

deprecated in view of sustainable agricultural practices. Hence, an efficient<br />

way to control the disease is important.<br />

An alternative to chemical control of <strong>plant</strong> diseases is the use of bacteria<br />

(biocontrol). They have the potential to displace or antagonise phytopathogenic<br />

or deleterious microorganisms in the rhizosphere. Biocontrol bacteria<br />

also produce chemicals, but these are degradable and only produced in low<br />

amounts at targeted locations. The latter approach fits well in the worldwide<br />

strategy to grow healthier <strong>plant</strong>s in a sustainable way and, therefore produce<br />

high quality food.<br />

To use biocontrol strains efficiently, the molecular interactions between<br />

<strong>plant</strong>, biocontrol agent, pathogen and their environment need to be understood.<br />

Genetic engineering is an important tool in helping us to define the<br />

molecular basis of pathogenicity and is also useful in helping us to identify<br />

the mechanisms in the action of biocontrol strains. Molecular genetic modification<br />

of microorganisms requires the development of plasmid-mediated<br />

transformation systems that include: (1) introduction of exogenous DNA into<br />

recipient cells, (2) expression of transformed genes, and (3) stable maintenance<br />

and replication of the inserted DNA leading to expression of the<br />

desired phenotypic trait. In this chapter, a practical approach to the analysis<br />

of biocontrol strains including the isolation, testing, and tagging of these<br />

strains, and transformation systems for pathogenic fungi to express reporter<br />

genes to track and visualise them in the rhizosphere, are discussed in relation<br />

to the pathogenic fungus Fusarium oxysporum f. sp. radicis-lycopersici.<br />

3 Selection of Antagonistic Strains<br />

3.1 Selection of Antagonistic Pseudomonas and Bacillus sp.<br />

Pseudomonas and Bacillus species constitute, together with Streptomyces<br />

species, a substantial fraction of the bacterial community isolated from the<br />

rhizosphere. Their presence is sometimes correlated with disease suppression.<br />

These beneficial bacteria can be exploited as biological pesticides to be


23 Visualisation of Rhizosphere Interactions 433<br />

used either as an alternative to, or in combination with chemicals to reduce<br />

the dose of these chemicals. Pseudomonas and Bacillus spp. are often abundantly<br />

present in the rhizosphere and surrounding soil of many crop <strong>plant</strong>s.<br />

Many of these species produce secondary metabolites that inhibit growth of,<br />

or kill, soil-borne phytopathogens. These antagonistic bacteria can either be<br />

isolated from the rhizosphere or from the soil in which <strong>plant</strong>s have been<br />

grown.<br />

In the following isolation procedure, tomato <strong>plant</strong>s harvested at the end of<br />

the growing season were picked randomly. Plant roots (0.3–0.4 g fresh weight)<br />

were vigorously shaken in phosphate buffer saline (PBS) for 1 h to detach the<br />

rhizosphere bacteria from the roots. The resulting bacterial suspensions from<br />

individual root systems were diluted and plated on one tenth strength tryptic<br />

soy agar (TSA) supplemented with the fungicide cycloheximide (50 mg/ml).<br />

The use of a nutrient-poor medium was reported to yield the highest numbers<br />

of isolates.After an incubation period of 2–7 days at 28 °C, a large variety<br />

of colonies with different morphologies were observed.<br />

The number of fluorescent pseudomonads found in the rhizosphere is very<br />

often variable. In some studies they were reported to be a dominant group,<br />

whereas other studies report that their numbers did not exceed 1 % of the<br />

total rhizosphere population isolated. The variations may be due to differences<br />

in <strong>plant</strong> species or cultivars, soil type, age of the <strong>plant</strong> roots, or the isolation<br />

method. Recently, it was also found that the percentage of antagonistic<br />

pseudomonads from a maize rhizosphere grown without chemical pesticides<br />

in Totontepec, Oaxaca State, Mexico, was 20 times higher than that from a rhizosphere<br />

grown in a commercial tomato field treated with chemicals in<br />

Andalusia, Spain (van den Broek et al., unpubl. data). No single medium is<br />

definitely suited for an unbiased selection of all culturable rhizosphere bacteria.<br />

Pseudomonas isolation (PI) agar can be used to specifically favour the<br />

growth of pseudomonads. One should also keep in mind that only the culturable<br />

part of the rhizosphere population will be obtained.<br />

Putative Bacillus strains are isolated by heating root samples at 80 °C for<br />

10 min prior to washing the bacteria from the roots. The bacterial solution is<br />

plated on Luria-Bertani (LB) agar plates supplemented with cycloheximide<br />

(50 mg/ml) and incubated for 2–5 days at 28 °C. Colonies with a Bacillus-like<br />

morphology are then compared to Bacillus-type strains. To determine<br />

whether one is dealing with Gram-positive or Gram-negative organisms, a<br />

first identification of colonies can be performed by determining the ability to<br />

form mucoid threads after pulling a toothpick out of a bacterial suspension in<br />

3 % KOH, which is indicative for Gram-negative organisms. A definite determination<br />

requires a standard Gram stain. Further characterisation methods<br />

include the use of Biolog, which is based on the ability of a strain to oxidise<br />

particular carbon sources, amplified ribosomal DNA restriction analysis<br />

(ARDRA), or PCR amplification of 16S ribosomal DNA fragments with specific<br />

primers followed by nucleotide sequencing and homology studies. In the


434<br />

Thomas F.C. Chin-A-Woeng et al.<br />

Biolog method, data sets derived from the carbon source utilisation patterns<br />

can be analysed with an appropriate software program (depending on the<br />

Gram character of the strain) and compared to known patterns of species<br />

present in commercially available databases. The latter two methods are<br />

based on specific sequences conserved between closely related species in the<br />

ribosomal rRNA gene fragments encompassing the 16S rDNA, the 16S–23S<br />

spacer region, and part of the 23S rDNA.<br />

3.2 In Vitro Antifungal Activity Test<br />

A simple in vitro assay to determine the activity against fungi can be performed<br />

by growing single bacterial colonies on agar medium in the presence<br />

of the fungus. The fungus is stab-inoculated in the centre of a Petri dish and<br />

bacterial strains are spot-inoculated at 2–2.5 cm distance from the fungus.<br />

The bacteria and fungus are allowed to grow concentrically and the formation<br />

of an inhibitory zone around the bacterial colony is an indication that the<br />

strain secretes a diffusible compound which inhibits growth of the fungus.<br />

A large scale identification of antifungal activity in growth supernatants of<br />

bacterial cultures can be performed in 96-well microtiter plates in the presence<br />

of F.o.r.l. The assay allows the convenient screening of a large number of<br />

strains in a reproducible and quantitative way. Strains to be tested are grown<br />

in a 96-well microtiter plate in a volume of 200 ml. After growth, the cells are<br />

sedimented by centrifugation at 5000 rpm for 10 min and the culture supernatants<br />

are passed through a 0.45 or 0.22-mm pore size filter.A volume of 75 ml<br />

supernatant is mixed with an equal volume of an agarose-spore suspension<br />

(2x malt extract broth, 1.3x10 4 spores/ml, 1.5 % (w/v) agarose). The final concentration<br />

of the spores in the wells is 1000 spores/well. The wells are sealed<br />

either with 75 ml paraffin oil (filter-sterile), with an oxygen-permeable plate<br />

seal, or with a piece of Saran wrap. Germination and mycelium growth is followed<br />

by measuring optical density (OD 620) of the wells using a microtiter<br />

plate reader (every hour) for approximately 72 h during growth at 28 °C.<br />

When an automated stack reader is used, many plates can be screened simultaneously<br />

in this way.<br />

4 In Vivo Biocontrol Assays<br />

4.1 Fusarium oxysporum–Tomato Biocontrol Assay in a Potting Soil<br />

System<br />

Biocontrol of Pseudomonas and Bacillus rhizosphere isolates can be tested in<br />

a bioassay in which tomato seedlings grown from seeds coated with biocontrol<br />

bacteria are grown in potting soil infected with F.o.r.l. spores. Spores are


23 Visualisation of Rhizosphere Interactions 435<br />

obtained from liquid cultures and mixed with the soil prior to <strong>plant</strong>ing the<br />

seeds.<br />

To isolate spores, F.o.r.l. is stab-inoculated onto potato dextrose agar<br />

medium and grown at 24 °C until the fungal mycelium covers the entire plate<br />

after a few days. One third of a PDA agar plate with F.o.r.l. is minced and used<br />

for inoculation of 200 ml Czapek-Dox medium in a 1-l Erlenmeyer flask. The<br />

fungus is grown for 2–3 days at room temperature under shaking at 110 rpm.<br />

Fungal mycelium and spore growth should be clearly visible at this stage. The<br />

F.o.r.l. inoculum is passed through Miracloth (Calbiochem-Novabiochem<br />

Corporation, La Jolla, CA, USA) or glass wool to remove the mycelium. The<br />

spore concentration is determined with a haemocytometer (with a depth of<br />

100 mm).<br />

The spore suspension is diluted in water to 1x10 6 spores/ml and added to<br />

potting soil to a final concentration of 6x10 6 spores/kg of soil. Spores are thoroughly<br />

mixed through the potting soil and the pots are filled with the infected<br />

soil. Seeds are sown in 8 plots of 12 pots, one seed per pot at a depth of 1–2 cm.<br />

Plants are watered from below to prevent disturbance of the root colonisation<br />

process.<br />

Bacterial strains are coated onto the tomato seeds in a simple procedure<br />

using methylcellulose. Pseudomonas strains are grown overnight in 3 ml<br />

King’s medium B at 28 °C. Bacilli are grown in 3 ml tryptic soy broth (TSB) for<br />

3 days at 28 °C. The overnight culture of bacteria is washed with PBS to<br />

remove the growth medium and diluted to a concentration of 2x10 9 CFU/ml.<br />

For bacilli the concentration is adjusted to 2x10 7 CFU/ml. Then equal volumes<br />

of the bacterial suspension and a 2.0 % (w/v) methylcellulose solution are<br />

mixed (methylcellulose is dissolved in water by vigorous stirring or by using<br />

a blender). Seeds are dipped into the mixture and dried in a sterile air stream<br />

on a filter paper. The coated seeds can be sown directly or kept at 4 °C for 1 or<br />

2 days. The number of bacteria recovered from tomato seeds after coating is<br />

approximately 10 4 CFU/seed. Seedling germination is determined 1 week<br />

after sowing. Between 2–3 weeks after sowing, depending upon the disease<br />

pressure, the percentage of diseased <strong>plant</strong>s is determined.A percentage of diseased<br />

<strong>plant</strong>s of approximately 60 % is preferred to perform statistical analyses.<br />

4.2 Gnotobiotic Fusarium oxysporum–Pythium ultimum and<br />

Rhizoctonia solani–Tomato Bioassays<br />

The gnotobiotic system used for this bioassay has been extensively used to<br />

study root colonisation. Briefly, tomato seeds are <strong>surface</strong>-sterilised in a 5 %<br />

household sodium hypochlorite solution for 3 min, followed by four thorough<br />

rinses with 20 ml sterile water for 2 h. Incubation of sterilised tomato seeds on<br />

KB medium, in our hands, shows that this method consistently yields seeds


436<br />

Thomas F.C. Chin-A-Woeng et al.<br />

free of contamination.After incubation for 24 h on agar-solidified <strong>plant</strong> nutrient<br />

solution (PNS) medium at 4 °C, seeds are allowed to germinate at 28 °C.<br />

Seedlings are inoculated 2 days later. A F.o.r.l. spore suspension, prepared as<br />

described previously, is added to the <strong>plant</strong> nutrient solution to a final concentration<br />

of 5x10 2 spores/ml, which is than mixed through the sterile sand to<br />

10 % (v/w) PNS.<br />

Rhizoctonia solani was grown on 2 % water agar for 5 days. Discs of approximately<br />

4 mm in diameter were cut from the edge of a growing colony and<br />

blended in PNS. P. ultimum was grown for 3–4 weeks in clarified V8 medium<br />

or hemp seed extract in water for 1–2 weeks. Oospores were collected free of<br />

the mycelium by washing them three times with sterile water and blending in<br />

0.1 M sucrose. The blended culture was incubated for 2 h on a shaker<br />

(130 rpm) at 28 °C, sedimented, and resuspended in 1 M sucrose. To kill the<br />

mycelium fragments, the suspension was incubated at –20 °C for 12 h. The culture<br />

was washed, layered over 1 M sucrose and centrifuged at 2300 rpm.<br />

Oospores were added in a final concentration of 5–25 oospores/g of sand.<br />

Germinated tomato seeds were incubated in a bacterial suspension with a<br />

concentration of 10 7 CFU/ml (Pseudomonas) or 10 9 CFU/ml (Bacillus) for<br />

10 min, after which the germinated seeds were <strong>plant</strong>ed in the sand at a depth<br />

of approximately 5 mm. Seed inoculation is preferred above inoculation from<br />

soil since commercial biocontrol of tomato pathogens is also based on seed<br />

coating while the pathogen is already present in the soil. This form of inoculation<br />

also results in more reproducible experimental data. Plants were grown<br />

in a growth chamber or a greenhouse for 7 days and the disease index was<br />

determined by scoring the <strong>plant</strong>s according a fixed disease index (Table 1).<br />

The data can be analysed statistically using a standard c 2 analysis.<br />

To confirm the presence of the fungus on <strong>plant</strong>s, suspected diseased root<br />

parts can be placed in 0.005 % household bleach for 30 s, thoroughly rinsed<br />

with sterile water, and placed on a rich (LC or PDA) medium. Plates are<br />

inspected for fungal growth after incubation at 28 °C for 2 days.<br />

Table 1. Example of Pythium ultimum disease indices<br />

Disease symptoms Disease index<br />

No visible symptoms 0<br />

Small brown spots on the main root and/or the crown 1<br />

Brown spots on the central root and extensive discoloration of crown 2<br />

Damping-off 3<br />

Dead 4


5 Microscope Analysis of Infection and Biocontrol<br />

5.1 Marking Fungi with Autofluorescent Proteins<br />

5.1.1 Transformation of Pathogenic Fungi<br />

5.1.1.1 Growth of Fungal Mycelium<br />

Protoplasts are usually used for transformations of fungi. The removal of the<br />

cell wall is achieved by treating mycelia or germlings in the presence of lytic<br />

enzymes. The osmotic balance of protoplasts in a suspension is usually maintained<br />

using sugars such as sucrose and sorbitol and salts such as magnesium<br />

chloride, potassium chloride, and ammonium sulphate.<br />

The following polyethylene/CaCl 2-mediated transformation procedure has<br />

been successfully applied to mycelium of F.o.r.l. Growing mycelium is prepared<br />

by inoculation of 100 ml potato dextrose broth in a 300-ml Erlenmeyer<br />

flask with a 5x4 mm size inoculum of mycelium. Depending on the particular<br />

F.o.r.l. strain, the fungus is grown between 2 and 5 days at 28 °C and 160 rpm.<br />

For example, F. oxysporum Fo47 is grown for 5 days, F. oxysporum f. sp. radicis-lycopersici<br />

ZUM2407 (IPO-DLO, Wageningen, The Netherlands) is grown<br />

for 2 days. Subsequently, the culture is passed through two layers of Miracloth<br />

and the filtrate is collected and sedimented by centrifugation at 5000 rpm for<br />

10 min. The supernatant is immediately discarded and washed three times<br />

with 50 ml of sterile water and sedimented. The characteristic purple upper<br />

layer is discarded and the pellet is resuspended in 2–5 ml of sterile water. The<br />

spore concentration is determined with a haemocytometer. From this spore<br />

suspension, a number of 5x10 8 conidia is inoculated into 40 ml potato dextrose<br />

broth and grown at 25 °C and 300 rpm for approximately 18 h or until<br />

the length of the germ tubes is at least ten times the size of a spore. The overall<br />

percentage of germinated spores should be higher than 95 %.<br />

5.1.1.2 Preparation of Protoplasts<br />

23 Visualisation of Rhizosphere Interactions 437<br />

Germlings to be converted into protoplasts are sedimented by centrifugation<br />

at 2000 rpm for 10 min, after which the supernatant is carefully removed and<br />

the pellet resuspended in 25 ml magnesium sulphate solution (1.2 M MgSO 4 ,<br />

50 mM sodium citrate, pH 5.8). The suspension is then passed through three<br />

layers of Miracloth. The mycelium trapped in the Miracloth is washed twice<br />

with magnesium sulphate solution and then transferred to a new tube with a<br />

cotton swab. The mycelium is then incubated in a protoplasting mix (10 mg/l<br />

Lysing Enzyme (Sigma L-2265, Sigma Chemicals Co., St. Louis, MO, USA),<br />

15 mg/ml Driselase (Sigma Chemicals Co., St. Louis, MO, USA) in magnesium<br />

sulphate solution). The enzyme solution should be centrifuged to remove any<br />

solid particles prior to use. The mixture is incubated for 24 h at 30 °C on a<br />

shaker (65 rpm). The conversion of cells into protoplasts can be followed by


438<br />

Thomas F.C. Chin-A-Woeng et al.<br />

phase contrast microscopy and, when the protoplastation nears completion,<br />

the protoplasts are collected on three layers of Miracloth, transferred to a new<br />

tube and washed with a sterile cold sorbitol solution (1 M sorbitol, 50 mM<br />

CaCl 2 , 10 mM Tris-HCl, pH 7.4). The protoplasts are sedimented by centrifugation<br />

at 850xg (2100 rpm) at 4 °C and the number of protoplasts is determined<br />

with a haemocytometer.<br />

5.1.1.3 Transformation of Protoplasts<br />

Protoplast are transformed by addition of up to 15 mg of DNA to 200 ml of protoplast<br />

suspension and incubated on ice for 15 min, or stored at 4 °C.A volume<br />

of 1.0 ml PEG solution (60 % (w/v) polyethylene glycol 6000, 50 mM CaCl 2,<br />

10 mM Tris-HCl, pH 7.4) is slowly added while shaking gently. The mixture is<br />

incubated on ice for 30 min after which the protoplasts are washed with a<br />

magnesium sulphate/potato dextrose broth solution at 4 °C. The protoplasts<br />

are sedimented by centrifugation at 2500 rpm at 4 °C for 10 min and resuspended<br />

in the remaining fluid after discarding the supernatant. The protoplasts<br />

are incubated 30 min at room temperature and portions (50–1200 ml)<br />

are plated onto selective media containing 0.8 M sucrose, 10 mM Tris-HCl pH<br />

7.4, 100 mg/ml hygromycin, and 1.5 % (w/v) agar. Plates are incubated for 2 or<br />

3 days at the appropriate growth temperature.<br />

5.2 Marking Rhizosphere Bacteria with Autofluorescent Proteins<br />

The green fluorescent protein (GFP) of the jellyfish Aequorea victoria has<br />

been rapidly and successfully adopted as an important marker for investigating<br />

processes in the rhizosphere. GFP is a 27-kDa polypeptide which converts<br />

the blue chemiluminescence of the Ca 2+ -sensitive photoprotein<br />

aequorin into green light. The active chromophore is a tripeptide, the formation<br />

of which is oxygen-dependent and occurs gradually after translation<br />

by undergoing an autocatalytic reaction. GFP emits bright green light<br />

(l max =510 nm) when excited with ultraviolet (UV) or blue light<br />

(l max=395 nm) in vivo and in vitro.<br />

GFP allows the non-destructive localisation and monitoring of individual<br />

cells on the root <strong>surface</strong> and does not require, unlike other biomarkers, exogenously<br />

added substrates, energy sources, or cofactors other than molecular<br />

oxygen. GFP fluorescence is stable, species-independent, requires no processing<br />

by the cells and fixing or staining is not necessary so artefacts cannot be<br />

introduced. However, if required, GFP allows fixation since it is unaffected by<br />

paraformaldehyde treatment. It is also stable under many other denaturing<br />

conditions such as the presence of denaturants or proteolytic enzymes, high<br />

temperatures (65 °C), and pH levels (6–12). Expression can be easily detected<br />

using epifluorescence or confocal laser scanning microscopy. Other optical


23 Visualisation of Rhizosphere Interactions 439<br />

methods that can be used to detect GFP-marked bacteria include the use of<br />

charge couple device (CCD) microscopy and cell sorting by fluorescent-activated<br />

cell sorters (FACS), which allows the sampling and identification of subpopulations<br />

of bacteria in a non-destructive way at the single cell level. Autofluorescently<br />

labelled colonies on agar plates can be detected under a<br />

hand-held UV-lamp or a low-resolution binocular microscope equipped with<br />

a UV lamp.<br />

Since gfp is eukaryotic in origin, optimised constructs for the expression of<br />

gfp in bacteria have been constructed and successfully applied. This was<br />

achieved by expression of gfp under the control of strong constitutive promoters<br />

or using red-shifted and UV-optimised mutant derivates. These GFP<br />

variants provide an increased fluorescent signal intensity in bacteria, faster<br />

rates of oxidative chromophore formation, resistance to photobleaching and<br />

excitation maximums better suited to conventional detection instruments.<br />

GFPuv emits bright green light (maximum at 509 nm) when exposed to UV or<br />

blue light (395 or 470 nm). Mutant proteins GFPmut2 and GFPmut3 have<br />

emission maximums of 507 and 511 nm when excited by blue light (481 and<br />

501 nm, respectively).<br />

Stable plasmid vectors (multicopy) and transposon vectors (single copy)<br />

for marking with fluorescent proteins are available for use in Gram-negative<br />

as well as Gram-positive bacteria. They can be used for tagging bacteria with<br />

a biomarker, construction of fusion proteins, assaying gene activity, or promoter<br />

probing. Plasmids pGB5, carrying gfp driven by a tac promoter, was<br />

shown to be 100 % stably maintained in Pseudomonas in the tomato rhizosphere<br />

and resulted in constitutive expression in Pseudomonas without addition<br />

of an inducer. Dandie et al. (2001) constructed transposon-based tagging<br />

vectors using a gfp marker gene under control of either constitutive or<br />

inducible promoters.<br />

Plasmids pFPV1 and pFPV2 direct high levels of gfp expression in E. coli,<br />

Salmonella typhimurium, and Yersinia pseudotuberculosis and in different<br />

mycobacterial species. The high levels of gfp expression were achieved by<br />

expression under control of the lacZpo and hsp60 heat-shock promoters,<br />

respectively. They have been used to visualise the infection process of mammalian<br />

cells by the three species. Transposon plasmid Tn5GFP1 was successfully<br />

used to follow Pseudomonas putida cells during water transport<br />

through a sand matrix. To study the colonisation pattern of P. chlororaphis<br />

MA342 on barley seeds, the strain was tagged using a plasmid pUTgfp2X<br />

harbouring gfp.<br />

For many applications, such as the analysis of chromosomal genes under<br />

physiological (monocopy) conditions using transcriptional fusions, stable<br />

integration of the reporter, or reduction of the risk of transfer of the genetic<br />

marker to other microorganisms, it is necessary to integrate the gfp transcriptional<br />

fusion into the chromosome of target bacteria by site-specific<br />

recombination or by random insertion, e.g. by means of transposons. A gfp


440<br />

Thomas F.C. Chin-A-Woeng et al.<br />

cloning cassette vector, pGreenTIR, was designed specifically for use in the<br />

construction of prokaryotic transcriptional fusions. The cassette confers sufficient<br />

fluorescence to recipient cells to be used in low copy-number plasmids<br />

with promoters conferring low levels of transcription in E. coli and<br />

Pseudomonas. The bacterial transposon Tn7 inserts at a high frequency into a<br />

specific intergenic site attTn7 on the chromosome in a number of Gram-negative<br />

bacteria. Tn7-based systems allow stable single-copy insertion of marker<br />

genes and insertion of transcriptional fusions in a single copy on the chromosome<br />

for gene expression studies at a neutral, intergenic site. Koch et al.<br />

developed a panel of flexible mini-Tn7 delivery vectors, including cloning<br />

vectors with an increased number of unique cloning sites, the lack of which<br />

has limited the use of Tn7 systems so far. A Tn10-based transposon was successfully<br />

used for fluorescence tagging of marine bacteria.<br />

Based on mini-Tn5 transposon derivatives, a gfp containing promoterprobe<br />

mini transposon was constructed for use in Pseudomonas species.<br />

Another set of vectors containing a mutated gfp gene was constructed for use<br />

with Gram-negative bacteria other than E. coli. pTn3gfp can be used for making<br />

random promoter probe gfp insertions into cloned DNA in E. coli for subsequent<br />

introduction into host strains. pUTmini-Tn5gfp can be used for making<br />

random promoter probe insertions directly into host strains. Plasmids<br />

p519gfp and p519nfp are broad host range mobilisable plasmids with gfp<br />

expressed from a lac and an npt2 promoter, respectively.<br />

Fluorescent markers can also be used to study viability and metabolic<br />

activity of bacteria in the rhizosphere. Normander et al. used gfpmut3b (Ser-<br />

64 Gly) to visualise the effect of indigenous populations on the distribution<br />

and activity of inoculated P. fluorescens DR54-BN14 in the barley rhizosphere.<br />

Using gfp-marked strains, they demonstrated that microcolonies of<br />

the inoculant strain were closely associated with cells of indigenous populations<br />

and that the majority of the cells have properties similar to those of<br />

starved cells.<br />

Mutagenesis and protein engineering of the original GFP from the jellyfish<br />

Aequorea has yielded variants with different excitation and emission<br />

spectra that can be used for dual colour imaging. Many engineered variants<br />

also appear to be improved in other aspects such as photostability, codon<br />

usage, and thermosensitivity. The first dual colour imaging of bacteria in a<br />

mixed population of E. coli cells was achieved by selective excitation of<br />

wild-type GFP and mutant derivatives with a red-shift in the excitation spectrum.<br />

Fluorescent proteins can also be successfully combined with the use of<br />

other biomarkers such as luciferase. To monitor cell numbers and metabolic<br />

activity of specific bacterial populations in liquid cultures and soil samples, a<br />

dual gfp-luxAB under control of the psbA promoter was integrated into the<br />

chromosomes of E. coli DH5a and P. fluorescens SBW25. Since luciferase output<br />

from luxAB-tagged bacteria decreases during starvation, lux expression


23 Visualisation of Rhizosphere Interactions 441<br />

was used as a marker for metabolic activity, while the much more stable gfp<br />

expression was used as an indicator for biomass. Alternatively, unstable variants<br />

of autofluorescent proteins with shorter half-lives can be used.<br />

Variants, fluorescent in colours ranging from blue to yellow, namely blue<br />

fluorescent protein (BFP), yellow fluorescent protein (YFP), and cyan fluorescent<br />

protein (CFP), and optimised counterparts of EGFP and EBFP were created<br />

by mutagenesis. By labelling microorganisms differently, these variants<br />

can be used to track multiple microorganisms simultaneously. The major<br />

problem with using GFP variants to label strains for simultaneous detection is<br />

the complicated separation of the spectral overlap of the different GFP-isoforms.<br />

Recently, red fluorescent protein (drFP583 or DsRed), isolated from the<br />

tropical Indo Pacific reef coral Discosoma sp., has been cloned. With an emission<br />

maximum at 583 nm, DsRed is suitable for almost crossover-free dual<br />

colour labelling in combination with EGFP (emission 509 nm) upon simultaneous<br />

excitation. Similarly, combination of cells tagged with ECFP and EGFP<br />

or a mixture of cells labelled with ECFP and EYFP allows them to be clearly<br />

distinguished from each other in the tomato rhizosphere. In addition, DsRed<br />

can be combined with any other autofluorescent protein since the emission<br />

spectrum of DsRed does not overlap that of the others. Using different colours<br />

of fluorescent proteins, up to three labels (e.g. EGFP, ECFP and DsRed) can be<br />

simultaneously traced in the rhizosphere. These variants have also been used<br />

to visualise interactions of a DsRed-labelled biocontrol bacterium P. chlororaphis<br />

PCL1391 with gfp-labelled F.o.r.l. strain in the tomato rhizosphere<br />

(Lagopodi et al., unpubl. data). Bacteria were dually labelled merely to localise<br />

them in the rhizosphere.<br />

The gfp genes can also be used as reporters for gene expression in the rhizosphere<br />

or for genes involved in quorum sensing. The estimated half-life of<br />

wild-type GFP is estimated to be at least 1 day. Since fluorescent proteins are<br />

extremely stable, they cannot be used for transient (real time) gene expression<br />

studies. Less stable variants have been constructed that can be used for<br />

analysis of transient gene expression in bacteria and, hence, promoter activity<br />

in the rhizosphere. Unstable variants of fluorescent proteins can be produced<br />

by addition of C-terminal degradation domains to the protein that are targets<br />

of natural protein degradation systems in cells. One such system exploits the<br />

action of intracellular tail-specific protein via the ssrA-mediated peptide<br />

degradation of prematurely terminated polypeptides at the C-terminal end.<br />

Homologues of ssrA have been identified in both Gram-negative and Grampositive<br />

bacteria. Gfpmut3 derivatives carrying these degradation domains<br />

have half-lives between 40 min and 2 h, while the estimated half-life of wild<br />

gfpmut3 is estimated to be at least 1 day.<br />

GFP can also be used and expressed in Gram-positive species such as Bacillus<br />

spp. pAD213 was constructed as a promoter-trap plasmid for Bacillus<br />

cereus. It allows screening of large libraries for identifying regulatory<br />

sequences and screening using flow cytometry and cell sorting. Plasmid vec-


442<br />

Thomas F.C. Chin-A-Woeng et al.<br />

tors have been described that enable routine production of GFP,YFP and CFP<br />

fusions in Gram-positive bacteria.<br />

One disadvantage of the use of fluorescent proteins is the maturation time<br />

of the protein, particularly that of DsRed. Although EGFP requires ~ 4 h for<br />

efficient microscopic visualisation, visualisation of DsRed requires longer<br />

periods. This delay is not due to inefficient expression of the DsRed protein<br />

since the protein can be detected in high quantities very soon, but it is rather<br />

due to an extended maturation time of the protein (20–48 h). DsRed is in fact<br />

brighter than first reported, but the fluorescence matures very slowly and the<br />

protein naturally forms a tetramer. More rapidly maturing and soluble variants<br />

of DsRed have been generated by mutagenesis (Brooke and Glick 2002).<br />

Furthermore, E. coli cells expressing DsRed protein are in general smaller<br />

than cells expressing EGFP or untransformed bacteria, indicating that DsRed<br />

might have a toxic effect. Another problem with the use of fluorescent proteins<br />

is the variability of expression in different bacterial species. GFP<br />

expressed from the same constructs is two to ten times higher expressed in E.<br />

coli than in pseudomonads. Interference by other fluorescent particles, bacteria,<br />

or root autofluorescence may also introduce artefacts or complicate the<br />

observations.<br />

5.3 Confocal Laser Scanning Microscopy of Rhizosphere Interactions<br />

The advent of fluorescent proteins offers a broad range of applications to<br />

track bacteria and study gene expression in the rhizosphere. By labelling different<br />

strains with different flavours of fluorescent proteins such as green, red,<br />

blue, or yellow fluorescent protein, multiple bacterial strains and their interactions<br />

with pathogens can be tracked simultaneously in the rhizosphere.<br />

To express gfpin F.o.r.l., pGFDGFP on which the sgfp gene is cloned between<br />

the A. nidulans gpdA promoter and the trpC terminator sequences was transformed<br />

to F.o.r.l.. The fungus was transformed by the previously described<br />

polyethyleneglycol/CaCl 2 -mediated transformation of protoplasts in the presence<br />

of pAN7–1, which allows selection for hygromycin B resistance<br />

(100 mg/ml). The level of gfp expression was high in the mycelium, micro- and<br />

macroconidia, and chlamydospores. The labelled isolates were equally pathogenic<br />

to tomato as the wild type. The marked fungus was introduced into the<br />

gnotobiotic sand system by mixing spores with sand. First, the interactions<br />

between fungal pathogens and the tomato root were studied. CLSM observations<br />

show that after 2 days the main root is surrounded by hyphae, which are<br />

interwoven with the root hairs. The contact between hyphae and the root was<br />

initiated at or via the root hairs.After 3 days,spot attachments of hyphae to the<br />

root <strong>surface</strong> are observed, predominantly at the crown and hyphae grow along<br />

the junctions of the epidermal cells after attachment. The first infection events<br />

take place 4 days after inoculation, as observed by penetration of epidermal


cells by hyphae. No penetration structures are observed except for swollen<br />

hyphae at the penetration site.Five days after <strong>plant</strong>ing,at which the first disease<br />

symptoms can be observed, a tight network of hyphae has grown around the<br />

root <strong>surface</strong> and epidermal cells are intercellularly colonised by hyphae. After<br />

complete destruction of the root system, the fungus forms macroconidia and<br />

starts colonising the cotyledons.<br />

After introduction of biocontrol bacteria to the test system, observations<br />

show that in the F.o.r.l. -tomato biocontrol system Pseudomonas bacteria not<br />

only colonise the tomato root <strong>surface</strong>, but also fungal hyphae (Bolwerk and<br />

Lagopodi, unpublished). These are indications that biocontrol bacteria not<br />

only protect the roots against fungi by niche exclusion and production of<br />

antibiotics, but that they actively attack the pathogen. Still, there is much to be<br />

discovered from these rhizosphere studies. The use of autofluorescent proteins<br />

has shown to be a promising way of visualising and understanding the<br />

interactions taking place in the rhizosphere between Pseudomonas and Bacillus<br />

biocontrol strains and fungal pathogens.<br />

6 Conclusions<br />

The whole procedure of isolation, screening for antifungal activity, and determining<br />

disease suppression in bioassays allows fast isolation of potential biocontrol<br />

strains. The gnotobiotic test system has proven to be a valuable test<br />

system to study interactions between biocontrol bacteria, phytopathogen, and<br />

host <strong>plant</strong>. Combined with the use of autofluorescent proteins, it provides us<br />

with an extraordinary opportunity to study the intricate cellular and molecular<br />

interactions that the key players use to mediate their actions in the rhizosphere.<br />

References and Selected Reading<br />

23 Visualisation of Rhizosphere Interactions 443<br />

Alabouvette C (1986) Fusarium wilt suppressive soils from the Chateaurenard region:<br />

reviews of a 10 year study. Agronomie 6:273–284<br />

Alexeyev MF, Shokolenko IN, Croughan TP (1995) New mini-Tn5 derivatives for insertion<br />

mutagenesis and genetic engineering in gram-negative bacteria. Can J Microbiol<br />

41:1053–1055<br />

Andersen JB, Sternberg C, Poulsen LK, Bjorn SP, Givskov M, Molin S (1998) New unstable<br />

variants of green fluorescent protein for studies of transient gene expression in<br />

bacteria. Appl Environ Microbiol 64:2240–2246<br />

Anderson M, Pollitt CE, Roberts IS, Eastgate JA (1998) Identification and characterization<br />

of the Erwinia amylovora rpoS gene: RpoS is not involved in induction of fireblight<br />

disease symptoms. J Bacteriol 180:6789–6792<br />

Baird GS, Zacharias DA, Tsien RY (2000) Biochemistry, mutagenesis, and oligomerization<br />

of DsRed, a red fluorescent protein from coral. Proc Natl Acad Sci USA<br />

97:11984–11989


444<br />

Thomas F.C. Chin-A-Woeng et al.<br />

Ballance DJ, Buxton FP, Turner G (1983) Transformation of Aspergillus nidulans by the<br />

orotidine-5¢-phosphate decarboxylase gene of Neurospora crassa. Biochem Biophys<br />

Res Commun 112:284–289<br />

Beckman CH (1987) The nature of wilt diseases of <strong>plant</strong>s. The American Phytopathological<br />

Society Press, Saint Paul, MN<br />

Bloemberg GV, O’Toole GA, Lugtenberg BJJ, Kolter R (1997) Green fluorescent protein as<br />

a marker for Pseudomonas spp. Appl Environ Microbiol 63:4543–4551<br />

Bloemberg GV,Wijfjes AH, Lamers GE, Stuurman N, Lugtenberg BJ (2000) Simultaneous<br />

imaging of Pseudomonas fluorescens WCS365 populations expressing three different<br />

autofluorescent proteins in the rhizosphere: new perspectives for studying microbial<br />

communities. Mol Plant-Microbe Interact 13:1170–1176<br />

Bochner BR (1989) Sleuthing out bacterial identities. Nature 339:157–158<br />

Brooke JB, Glick BS (2002) Rapidly maturing variants of the Discosoma red fluorescent<br />

protein (DsRed). Nat Biotechnol 20:83–87<br />

Brown JW, Hunt DA, Pace NR (1990) Nucleotide sequence of the 10Sa RNA gene of the<br />

beta-purple eubacterium Alcaligenes eutrophus. Nucleic Acids Res 18:2820<br />

Burlage RS, Yang ZK, Mehlhorn T (1996) A transposon for green fluorescent protein<br />

transcriptional fusions: application for bacterial transport experiments. Gene<br />

173:53–58<br />

Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC (1994) Green fluorescent protein<br />

as a marker for gene expression. Science 263:802–805<br />

Chin-A-Woeng TFC, Bloemberg GV, van der Bij AJ, van der Drift KMGM, Schripsema J,<br />

Kroon B, Scheffer RJ, Keel C, Bakker PAHM, Tichy HV, de Bruijn FJ, Thomas-Oates JE,<br />

Lugtenberg BJJ (1998) Biocontrol by phenazine-1-carboxamide-producing Pseudomonas<br />

chlororaphis PCL1391 of tomato root rot caused by Fusarium oxysporum f. sp.<br />

radicis-lycopersici. Mol Plant-Microbe Interact 11:1069–1077<br />

Chiu WL, Niwa Y, Zeng W, Hirano T, Kobayashi H, Sheen J (1996) Engineered GFP as a<br />

vital reporter in <strong>plant</strong>s. Curr Biol 6:325–330<br />

Cody CW, Prasher DC, Westler WM, Prendergast FG, Ward WW (1993) Chemical structure<br />

of the hexapeptide chromophore of the Aequorea green-fluorescent protein. Biochemistry<br />

32:1212–1218<br />

Cormack BP, Valdivia RH, Falkow S (1996) FACS-optimized mutants of the green fluorescent<br />

protein (GFP). Gene 173:33–38<br />

Cowan SE, Gilbert E, Khlebnikov A, Keasling JD (2000) Dual labeling with green fluorescent<br />

proteins for confocal microscopy. Appl Environ Microbiol 66:413–418<br />

Craig NL (1989) Transposon Tn7. In: Berg DE, Howe MM (eds) Mobile DNA. American<br />

Society for Microbiology, Washington, pp 211–225<br />

Crameri A, Whitehorn EA, Tate E, Stemmer WP (1996) Improved green fluorescent protein<br />

by molecular evolution using DNA shuffling. Nat Biotechnol 14:315–319<br />

Cuppers HGAM, Oomes S, Brul S (1997) A model for the combined effects of temperature<br />

and salt concentration on growth rate of food spoilage molds. Appl Environ<br />

Microbiol 63:3764–3769<br />

Dandie CE, Thomas SM, McClure NC (2001) Comparison of a range of green fluorescent<br />

protein-tagging vectors for monitoring a microbial inoculant in soil. Lett Appl Microbiol<br />

32:26–30<br />

Daubner SC, Astorga AM, Leisman GB, Baldwin TO (1987) Yellow light emission of Vibrio<br />

fischeri strain Y-1: purification and characterization of the energy-accepting yellow<br />

fluorescent protein. Proc Natl Acad Sci USA 84:8912–8916<br />

de Lorenzo V, Timmis KN (1994) Analysis and construction of stable phenotypes in<br />

gram-negative bacteria with Tn5- and Tn10-derived minitransposons. Methods<br />

Enzymol 235:386–405


23 Visualisation of Rhizosphere Interactions 445<br />

de Lorenzo V, Herrero M, Jakubzik U, Timmis KN (1990) Mini-Tn5 transposon derivatives<br />

for insertion mutagenesis, promotor probing, and chromosomal insertion of<br />

cloned DNA in Gram-negative Eubacteria. J Bacteriol 172:6568–6572<br />

Dekkers LC, Mulders IH, Phoelich CC, Chin AWT, Wijfjes AH, Lugtenberg BJ (2000) The<br />

sss colonization gene of the tomato-Fusarium oxysporum f. sp. radicis-lycopersici biocontrol<br />

strain Pseudomonas fluorescens WCS365 can improve root colonization of<br />

other wild-type Pseudomonas spp. bacteria. Mol Plant Microbe Interact 13:1177–1183<br />

Dunn AK, Handelsman J (1999) A vector for promoter trapping in Bacillus cereus.Gene<br />

226:297–305<br />

Emmert EAB, Handelsman J (1999) Biocontrol of <strong>plant</strong> disease: a (Gram-) positive perspective.<br />

FEMS Microbiol Lett 171:1–9<br />

Feucht A, Lewis PJ (2001) Improved plasmid vectors for the production of multiple fluorescent<br />

protein fusions in Bacillus subtilis. Gene 264:289–297<br />

Geels FP, Schippers G (1983) Selection of antagonistic fluorescent Pseudomonas spp. and<br />

their root colonization and persistence following treatment of seed potatoes. Phytopath<br />

Z 108:193–206<br />

Glandorf DCM, Sluis I, Anderson AJ, Bakker PAHM, Schippers B (1994) Agglutination,<br />

adherence, and root colonization by fluorescent pseudomonads.Appl Environ Microbiol<br />

60:1726–1733<br />

Gutterson N (1990) Microbial fungicides: recent approaches to elucidating mechanisms.<br />

Crit Rev Biotechnol 10:69–91<br />

Haidinger W, Szostak MP, Beisker W, Lubitz W (2001) Green fluorescent protein (GFP)dependent<br />

separation of bacterial ghosts from intact cells by FACS. Cytometry<br />

44:106–112<br />

Halebian S, Harris B, Finegold SM, Rolfe RD (1981) Rapid method that aids in distinguishing<br />

gram-positive from gram-negative anaerobic bacteria. J Clin Microbiol<br />

13:444–448<br />

Handelsman J, Raffel SJ, Mester EH, Wunderlich L, Grau CR (1999) Biological control of<br />

damping-off of alfalfa seedlings with Bacillus cereus UW85. Appl Environ Microbiol<br />

56:713–718<br />

Heim R, Prasher DC, Tsien RY (1994) Wavelength mutations and posttranslational<br />

autoxidation of green fluorescent protein. Proc Natl Acad Sci USA 91:12501–12504<br />

Hoffland E, Findenegg GR, Nelemans JA (1989) Solubilization of rock phosphate by rape.<br />

Plant Soil 113:161–165<br />

Inouye S, Tsuji FI (1994) Evidence for redox forms of the Aequorea green fluorescent<br />

protein. FEBS Lett 351:211–214<br />

Johnstone IL, Hughes SG, Clutterbuck AJ (1985) Cloning an Aspergillus nidulans developmental<br />

gene by transformation. EMBO J 4:1307–1311<br />

Jakobs S, Subramaniam V, Schonle A, Jovin TM, Hell SW (2000) EFGP and DsRed expressing<br />

cultures of Escherichia coli imaged by confocal, two-photon and fluorescence<br />

lifetime microscopy. FEBS Lett 479:131–135<br />

Karatani H,Wilson T, Hastings JW (1992) A blue fluorescent protein from a yellow-emitting<br />

luminous bacterium. Photochem Photobiol 55:293–299<br />

Keiler KC,Waller PR, Sauer RT (1996) Role of a peptide tagging system in degradation of<br />

proteins synthesized from damaged messenger RNA. Science 271:990–993<br />

King EO, Ward MK, Raney DE (1954) Two simple media for the demonstration of<br />

pyocyanin and fluorescein. J Lab Clin Med 44:301–307<br />

Kistler HC, Benny UK (1988) Genetic transformation of the fungal wilt pathogen, Fusarium<br />

oxysporum. Curr Genet 13:145–149<br />

Koch B, Jensen LE, Nybroe O (2001) A panel of Tn7-based vectors for insertion of the gfp<br />

marker gene or for delivery of cloned DNA into Gram-negative bacteria at a neutral<br />

chromosomal site. J Microbiol Methods 45:187–195


446<br />

Thomas F.C. Chin-A-Woeng et al.<br />

Kremer RJ, Begonia MFT, Stanley L, Lanham ET (1990) Characterization of rhizobacteria<br />

associated with weed seedlings. Appl Environ Microbiol 56:1649–1655<br />

Lagopodi AL, Ram AFJ, Lamers GEM, Punt PJ, van den Hondel CAMJJ, Lugtenberg BJJ,<br />

Bloemberg GV (2002) Novel aspects of tomato root colonization and infection by<br />

Fusarium oxysporum f. sp. radicis-lycopersici revealed by confocal laser scanning<br />

microscopic analysis using the green fluorescent protein as a marker. Mol Plant-<br />

Microbe Interact 15:172–179<br />

Lambert B, Leyns F, Rooyen L, Gossele F, Papon Y, Swings J (1987) Rhizobacteria of maize<br />

and their antifungal activities. Appl Environ Microbiol 53:1866–1871<br />

Lambert B, Meire P, Joos H, Lens P, Swings J (1990) Fast-growing, aerobic, heterotrophic<br />

bacteria from the rhizosphere of young sugar beet <strong>plant</strong>s. Appl Environ Microbiol<br />

56:3375–3381<br />

Lewis PJ, Errington J (1996) Use of green fluorescent protein for detection of cell-specific<br />

gene expression and subcellular protein localization during sporulation in Bacillus<br />

subtilis. Microbiology 142(Pt 4):733–740<br />

Lewis PJ, Marston AL (1999) GFP vectors for controlled expression and dual labelling of<br />

protein fusions in Bacillus subtilis. Gene 227:101–110<br />

Macheroux P, Schmidt KU, Steinerstauch P, Ghisla S, Colepicolo P, Buntic R, Hastings JW<br />

(1987) Purification of the yellow fluorescent protein from Vibrio fischeri and identity<br />

of the flavin chromophore. Biochem Biophys Res Commun 146:101–106<br />

Matthysse AG, Stretton S, Dandie C, McClure NC, Goodman AE (1996) Construction of<br />

GFP vectors for use in gram-negative bacteria other than Escherichia coli. FEMS<br />

Microbiol Lett 145:87–94<br />

Matz MV, Fradkov AF, Labas YA, Savitsky AP, Zaraisky AG, Markelov ML, Lukyanov SA<br />

(1999) Fluorescent proteins from nonbioluminescent Anthozoa species. Nat Biotechnol<br />

17:969–973<br />

Mess JJ,Wit R, Testerink CS, de Groot F, Haring MA, Cornelissen BJC (1999) Loss of avirulence<br />

and reduced pathogenicity of a gamma-irradiated mutant of Fusarium oxysporum<br />

f. sp. lycopersici. Phytopathology 89:1131–1137<br />

Miller DM III, Desai NS, Hardin DC, Piston DW, Patterson GH, Fleenor J, Xu S, Fire A<br />

(1999) Two-color GFP expression system for C. elegans. BioTechniques 26:914–916<br />

Miller HJ, Henken G, van Veen JA (1989) Variation and composition of bacterial populations<br />

in the rhizospheres of maize, wheat, and grass cultivars. Can J Microbiol<br />

35:656–660<br />

Miller HJ, Liljeroth E, Henken G, van Veen JA (1990) Fluctuations in the fluorescent<br />

pseudomonad and actinomycete populations of the rhizosphere and rhizoplane during<br />

growth of spring wheat. Can J Microbiol 36:254–258<br />

Miller WG, Lindow SE (1997) An improved GFP cloning cassette designed for prokaryotic<br />

transcriptional fusions. Gene 191:149–153<br />

Normander B, Hendriksen NB, Nybroe O (1999) Green fluorescent protein-marked<br />

Pseudomonas fluorescens: localization, viability, and activity in the natural barley rhizosphere.<br />

Appl Environ Microbiol 65:4646–4651<br />

Oakley BR, Rinehart JE, Mitchell BL, Oakley CE, Carmona C, Gray GL, May GS (1987)<br />

Cloning, mapping and molecular analysis of the pyrG (orotidine-5¢- phosphate decarboxylase)<br />

gene of Aspergillus nidulans. Gene 61:385–399<br />

Osmani SA, May GS, Morris NR (1987) Regulation of the mRNA levels of nimA,a gene<br />

required for the G2-M transition in Aspergillus nidulans. J Cell Biol 104:1495–1504<br />

Pusey PL (1999) Use of Bacillus subtilis and related organisms as biofungicides. Pestic<br />

Sci 27:133–140<br />

Pusey PL, Wilson CL (1984) Postharvest biological control of stone fruit brown rot by<br />

Bacillus subtilis. Plant Dis 68:753–756<br />

Scher FM, Baker R (1980) Mechanism of biological control in a fusarium-suppressive<br />

soil. Phytopathology 72:1567–1573


23 Visualisation of Rhizosphere Interactions 447<br />

Silo-suh LA, Lethbridge BJ, Raffel SJ, He H, Clardy J, Handelsman J (1994) Biological<br />

activities of two fungistatic antibiotics produced by Bacillus cereus UW85. Appl Environ<br />

Microbiol 60:2023–2030<br />

Simons M, van der Bij AJ, Brand J, de Weger LA, Wijffelman CA, Lugtenberg BJJ (1996)<br />

Gnotobiotic system for studying rhizosphere colonization by <strong>plant</strong> growth-promoting<br />

Pseudomonas bacteria. Mol Plant-Microbe Interact 9:600–607<br />

Steidle A, Sigl K, Schuhegger R, Ihring A, Schmid M, Gantner S, Stoffels M, Riedel K,<br />

Givskov M, Hartmann A, Langebartels C, Eberl L (2001) Visualization of N-acylhomoserine<br />

lactone-mediated cell-cell communication between bacteria colonizing the<br />

tomato rhizosphere. Appl Environ Microbiol 67:5761–5770<br />

Stretton S, Techkarnjanaruk S, McLennan AM, Goodman AE (1998) Use of green fluorescent<br />

protein to tag and investigate gene expression in marine bacteria. Appl Environ<br />

Microbiol 64:2554–2559<br />

Stutz EW, Defago G, Kern H (1986) Naturally occurring fluorescent pseudomonads<br />

involved in the suppression of black root rot of tobacco. Phytopathology 76:181–185<br />

Stuurman N, Bras CP, Schlaman HR, Wijfjes AH, Bloemberg G, Spaink HP (2000) Use of<br />

green fluorescent protein color variants expressed on stable broad-host-range vectors<br />

to visualize rhizobia interacting with <strong>plant</strong>s. Mol Plant Microbe Interact 13:1163–1169<br />

Suarez A, Guttler A, Stratz M, Staendner LH, Timmis KN, Guzman CA (1997) Green fluorescent<br />

protein-based reporter systems for genetic analysis of bacteria including<br />

monocopy applications. Gene 196:69–74<br />

Tilburn J, Scazzocchio C, Taylor GG, Zabicky-Zissman JH, Lockington RA, Davies RW<br />

(1983) Transformation by integration in Aspergillus nidulans. Gene 26:205–221<br />

Tombolini R, Unge A, Davey ME, deBruijn FJ, Jansson JK (1997) Flow cytometric and<br />

microscopic analysis of GFP-tagged Pseudomonas fluorescens bacteria. FEMS Microbiol<br />

Ecol 22:17–28<br />

Tombolini R, van der Gaag DJ, Gerhardson B, Jansson JK (1999) Colonization pattern of<br />

the biocontrol strain Pseudomonas chlororaphis MA 342 on barley seeds visualized by<br />

using green fluorescent protein. Appl Environ Microbiol 65:3674–3680<br />

Tyagi JS, Kinger AK (1992) Identification of the 10Sa RNA structural gene of Mycobacterium<br />

tuberculosis. Nucleic Acids Res 20:138<br />

Unge A, Tombolini R, Davey ME, de Bruijn FJ, Jansson JK (1998) GFP as a marker gene.<br />

In: Akkermans AD, van Elsas JD, de Bruijn FJ (eds) Molecular microbial ecology manual.<br />

Kluwer, Dordrecht, pp 1–16<br />

Unge A, Tombolini R, Molbak L, Jansson JK (1999) Simultaneous monitoring of cell<br />

number and metabolic activity of specific bacterial populations with a dual gfpluxAB<br />

marker system. Appl Environ Microbiol 65:813–821<br />

Ushida C, Himeno H, Watanabe T, Muto A (1994) tRNA-like structures in 10Sa RNAs of<br />

Mycoplasma capricolum and Bacillus subtilis. Nucleic Acids Res 22:3392–3396<br />

Valdivia RH, Falkow S (1996) Bacterial genetics by flow cytometry: rapid isolation of Salmonella<br />

typhimurium acid-inducible promoters by differential fluorescence induction.<br />

Mol Microbiol 22:367–378<br />

Valdivia RH, Hromockyj AE, Monack D, Ramakrishnan L, Falkow S (1996) Applications<br />

for green fluorescent protein (GFP) in the study of host–pathogen interactions. Gene<br />

173:47–52<br />

Vaneechoutte M, Boerlin P, Tichy HV, Bannerman E, Jager B, Bille J (1998) Comparison of<br />

PCR-based DNA fingerprinting techniques for the identification of Listeria species<br />

and their use for atypical Listeria isolates. Int J Syst Bacteriol 48:127–139<br />

Ward DM (1989) Molecular probes for analysis of microbial communities. In: Characklis<br />

WG, Wilderer PA (eds) Structure and function of biofilms. Wiley, New York, pp<br />

145–155


448<br />

Thomas F.C. Chin-A-Woeng et al.<br />

Waterhouse RN, Buhariwalla H, Bourn D, Rattray EAS, Glover LA (1996) CCD detection<br />

of lux-marked Pseudomonas syringae pv. phaseolicola forms associated with Chinesecabbage<br />

and the resulting disease protection against Xanthomonas campestris. Lett<br />

Appl Microbiol 22:262–266<br />

Weller DM, Cook RJ (1983) Suppression of take-all of wheat by seed treatments with fluorescent<br />

pseudomonads. Phytopathology 73:463–469<br />

Weller DM, Zhang BX, Cook RJ (1985) Application of a rapid screening test for selection<br />

of bacteria suppressive to take-all of wheat. Plant Dis 69:710–713<br />

Williams JG, Kubelik AR, Livak KJ, Rafalski JA, Tingey SV (1990) DNA polymorphisms<br />

amplified by arbitrary primers are useful as genetic markers. Nucleic Acids Res<br />

18:6531–6535<br />

Yang TT, Kain SR, Kitts P, Kondepudi A,Yang MM,Youvan DC (1996a) Dual color microscopic<br />

imagery of cells expressing the green fluorescent protein and a red-shifted<br />

variant. Gene 173:19–23<br />

Yang TT, Cheng L, Kain SR (1996b) Optimized codon usage and chromophore mutations<br />

provide enhanced sensitivity with the green fluorescent protein. Nucleic Acids Res<br />

24:4592–4593<br />

Yang TT, Sinai P, Green G, Kitts PA, Chen YT, Lybarger L, Chervenak R, Patterson GH, Piston<br />

DW, Kain SR (1998) Improved fluorescence and dual color detection with<br />

enhanced blue and green variants of the green fluorescent protein. J Biol Chem<br />

273:8212–8216<br />

Yelton MM, Hamer JE, Timberlake WE (1984) Transformation of Aspergillus nidulans by<br />

using a trpC plasmid. Proc Natl Acad Sci USA 81:1470–1474


24 Microbial Community Analysis in the<br />

Rhizosphere by in Situ and ex Situ Application of<br />

Molecular Probing, Biomarker and Cultivation<br />

Techniques<br />

Anton Hartmann, Rüdiger Pukall, Michael Rothballer,<br />

Stephan Gantner, Sigrun Metz, Michael Schloter<br />

and Bernhard Mogge<br />

1 Introduction<br />

It is well known that the bacterial diversity in soil habitats is much greater<br />

compared to the artificial cultivation techniques (Torsvik et al. 1996;<br />

Chatzinotas et al. 1998). It is generally accepted that only a combination of<br />

methods including cultivation and several cultivation-independent techniques<br />

is able to provide a more representative picture of the microbial diversity<br />

in environmental habitats (Wagner et al. 1993; Liesack et al. 1997). This is<br />

also true for the <strong>plant</strong>/soil compartment, although the degree of culturability<br />

is thought to be higher on the root <strong>surface</strong>. Supposedly, rhizosphere microbes<br />

respond to the presence of easily consumable substrates on the root <strong>surface</strong><br />

with fast growth rates, which is indicative for r-strategy; successful colonization<br />

of the rhizosphere is the final result of this behavior.<br />

In-depth characterization of bacterial communities residing in environmental<br />

habitats has been greatly stimulated by the application of molecular<br />

phylogenetic tools, such as 16S ribosomal RNA-directed oligonucleotide<br />

probes derived from extensive 16S rDNA sequence analysis. These phylogenetic<br />

probes can be successfully applied in diverse microbial habitats using<br />

the fluorescence in situ hybridization (FISH) technique (Giovannoni et al.<br />

1988; Amann et al. 1995; Tas and Lindström 2001). In addition, the application<br />

of the immunofluorescence techniques to detect specific subpopulations of<br />

enzymes and of fluorescence marker-tagged bacteria or reporter constructs<br />

enables a highly resolving population and functional analysis (Hartmann et<br />

al. 1997; Unge et al. 1999). Phylogenetic in situ studies of the population structure<br />

can thus be supplemented with functional or phenotypic in situ investigation<br />

approaches.<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


450<br />

Anton Hartmann et al.<br />

The rhizosphere is defined as the soil compartment which is greatly influenced<br />

by <strong>plant</strong> roots (Campbell and Greaves 1990a). The rhizosphere microbial<br />

community is shaped by the effect of root exudates (Brimecomb et al.<br />

2001). Several methodological approaches are available to study the rhizosphere<br />

carbon flow and the microbial population dynamics induced by rootborn<br />

carbon sources (Morgan and Whipps 2001). In addition, multiple communicative<br />

links exist between the rhizosphere microflora and the roots on<br />

the basis of highly specific organic signals (Werner 2001). It is appropriate to<br />

distinguish the root itself (with the endorhizosphere and the root <strong>surface</strong>, the<br />

rhizoplane) from the soil compartment surrounding the root (bulk soil and<br />

ectorhizosphere). In the following sections, two experimental approaches to<br />

investigate root-associated bacterial communities are presented. Figure 1<br />

provides a flow diagram of the separation of the rhizosphere compartments<br />

and the various in situ and ex situ methods applied. On one hand, population<br />

and functional studies can be conducted directly in the rhizoplane (in situ) by<br />

combining specific fluorescence probing with confocal laser scanning microscopy<br />

yielding detailed information about the localization and small scale<br />

distribution of bacterial cells and their activities on the root <strong>surface</strong> (Sect. 2).<br />

On the other hand, the separated rhizosphere compartments and the bacteria<br />

extracted from these different compartments allow a variety of subsequent ex<br />

situ-studies (Sect. 3). Studies, such as cultivation of bacteria on plates and<br />

microscopic counting of bacteria on filters after FISH analysis provide quan-<br />

Plants<br />

Roots with<br />

adhering soil<br />

Shaking,<br />

washing<br />

Root free soil<br />

(Compartment I)<br />

Ectorhizosphere soil<br />

(Compartment II)<br />

Roots: Rhizoplane<br />

and endorhizosphere<br />

(Compartment III)<br />

ISS ESS<br />

Fixation Extraction<br />

In situ-studies (ISS):<br />

FISH, Immunolabeling, monitoring<br />

of fluorescence tagged bacteria and<br />

constructs<br />

ISS ESS<br />

Ex situ-studies (ESS):<br />

DNA-extraction, PCR-amplification of<br />

phylogenetic marker regions / TGGE<br />

PLFA-biomarker,CSLP-techniques<br />

Fig. 1. Flow diagram of separation of rhizosphere compartments and overview of in situ<br />

and ex situ analyses using molecular probing, biomarker and cultivation techniques


titative data about the community composition. In addition, the bacterial<br />

diversity can be investigated using PCR-amplification of phylogenetic marker<br />

genes combined with subsequent electrophoretic fingerprint analysis or<br />

cloning and sequencing studies. These approaches can be supplemented by a<br />

general microbial structural and functional diversity analysis using community<br />

phospholipid fatty acid and substrate utilization pattern analysis, respectively.<br />

2 In Situ Studies of Microbial Communities Using Specific<br />

Fluorescence Labeling and Confocal Laser Scanning<br />

Microscopy<br />

A detailed understanding of the ecology of bacterial populations requires in<br />

situ information about the localization of the colonization sites at specific<br />

areas on root <strong>surface</strong>s and also about neighboring populations. Therefore,<br />

true in situ studies need to be performed and these must include an identification<br />

of the bacteria on a phylogenetic level and also information about their<br />

in situ activity. Since soil and <strong>plant</strong> <strong>surface</strong>s are very complex in microstructure<br />

and optical appearance, special microscopic techniques have to be<br />

applied. Confocal laser scanning microscopy enables us to circumvent to a<br />

great degree disturbing autofluorescence from out-of-focus-planes by performing<br />

optical sections (xy and xz scans) through the sample (Hartmann et<br />

al. 1998). It has been demonstrated that CSLM studies combined with the<br />

application of specific fluorescent probes considerably improve microbial<br />

ecology studies in the rhizosphere (Schloter et al. 1993; Aßmus et al. 1995).<br />

The confocal pinhole cuts off all out-of-focus fluorescence to reach the amplifiers.<br />

The application of several lasers with different excitation wavelengths in<br />

combination with differently fluoro-labeled probes allow the simultaneous<br />

analysis of different populations and/or activities (Amann et al. 1995; Stoffels<br />

et al. 2001). If possible, nested approaches with overlapping probe specificities<br />

should be used to improve the fidelity of the in situ identification, e.g., by fluorescence<br />

in situ hybridization. In addition, the use of the green fluorescent<br />

protein (GFP) as a structural and functional autofluorescence marker has<br />

successfully lightened up the biology and ecology of diverse biota, including<br />

bacteria, fungi, protozoa and <strong>plant</strong>s (Lorang et al. 2001).<br />

2.1 Fluorescence in Situ Hybridization<br />

24 Microbial Community Analysis in the Rhizosphere 451<br />

Root samples are fixed overnight at 4 °C in 3 % paraformaldehyde containing<br />

PBS (phosphate-buffered saline, composed of 0.13 M NaCl, 7 mM Na 2HPO 4<br />

and 3 mM NaH 2 PO 4 [pH 7.2]). Root pieces are washed in PBS, mixed with<br />

0.3 % agarose, dropped onto glass slides and dried at room temperature.


452<br />

Table 1. Phylogenetic oligonucleotide probes for fluorescence in situ hybridization (FISH) and dot blot hybridization<br />

Probe Probe sequence (5¢–3¢) Target site, rRNA position a Specificity Reference<br />

Anton Hartmann et al.<br />

EUB338 GCTGCCTCCCGTAGGAGT 16S rRNA, 338–355 Bacteria Amann et al. (1990)<br />

EUB788b CTACCAGGGTATCTAATCC 16S rRNA, 785–803 Bacteria Lee et al. (1993)<br />

EUB927b ACCGCTTGTGCGGGCCC 16S rRNA, 927–942 Bacteria Giovannoni et al. (1988)<br />

EUB1055b CACGAGCTGACGACAGCCAT 16S rRNA, 1055–1074 Bacteria Lee et al. (1993)<br />

EUB1088b GCTCGTTGCGGGACTTAACC 16S rRNA, 1088–1107 Bacteria Lee et al. (1993)<br />

ALF1b CGTTCG(C/T)TCTGAGCCAG 16S rRNA, 19–35 Alpha subclass of Proteobacteria Manz et al. (1992)<br />

BET42ac GCCTTCCCACTTCGTTT 23S rRNA, 1027–1043 Beta subclass of Proteobacteria Manz et al. (1992)<br />

CF319a TGGTCCGTGTCTCAGTAC 16 SrRNA, 319–336 Cytophaga-Flavobacterium cluster Manz et al. (1996)<br />

GAM42ac GCCTTCCCACATCGTTT 23S rRNA, 1027–1043 Gamma subclass of Proteobacteria Manz et al. (1992)<br />

Rhi1247 TCGCTGCCCACTGTC 16S rRNA, 1247–1261 Rhizobium, Ochrobactrum Ludwig et al. (1998)<br />

GPd TCATCATGCCCCTTATG 16S rRNA, 1199–1215 Gram-positive bacteria Rheims et al. (1996)<br />

HMd CCCTGAGTTATTCCGAAC 16S rRNA, 142–159 Hyphomicrobium, methylotrophs Tsien et al. (1990)<br />

PLAd GGC(GA)TGGATTAGGCATGC 16S rRNA, 41–58 Planctomycetaceae Liesack and Stackebrandt<br />

(1992)<br />

PS-MGd CCTTCCTCCCAACTT 16S rRNA, 440–454 Pseudomonas aeruginosa Braun-Howland et al.<br />

(1993)<br />

a E. coli numbering, Brosius et al. (1981)<br />

b Used in combination with probe EUB338 and three other domain-specific probes for quantification of bacterial cells on filters (EUB-MIX)<br />

c Used with an equimolar amount of unlabeled competitor oligonucleotide GAM42a or BET42a, respectively<br />

d Used for dot blot hybridization only


24 Microbial Community Analysis in the Rhizosphere 453<br />

These glass slides are immersed in 50, 80 and 96 % ethanol for 3 min each and<br />

stored at room temperature. Oligonucleotide probes (Table 1) labeled with<br />

Cy3, Cy5 or 5(6)-carboxyfluorescein-N-hydroxysuccinimide ester (FLUOS) at<br />

the 5¢¢-end are used. The oligonucleotides are stored in distilled water at a<br />

concentration of 50 ng/ml (Amann et al. 1990).<br />

FISH was performed as described in detail, e.g., by Wagner et al. (1993) at<br />

46 °C for 90 min in hybridization buffer (20 mM Tris-HCl, pH 7.2, 0.01 % SDS<br />

and 5 mM EDTA) containing 0.9 M NaCl and formamide at the percentages<br />

shown in Table 1. Hybridization was followed by a stringent washing step at<br />

48 °C for 15 min. The washing buffer was removed by rinsing the slides with<br />

distilled water. Counterstaining with DAPI and mounting in Citifluor AF1<br />

(Citifluor Ltd., London, UK) was performed as described previously (Aßmus<br />

et al. 1995).<br />

The microscopic in situ analysis can be performed using an LSM 410 or<br />

LSM 510 inverted confocal laser scanning microscope (Zeiss, Jena, Germany),<br />

equipped with two lasers (Ar-ion UV; Ar-ion visible; HeNe) supplying excitation<br />

wavelengths at 365, 488, 543 and 633 nm, respectively. Sequentially<br />

recorded images are assigned to the respective fluorescence color and then<br />

merged into a true color display. All image combining and processing is performed<br />

with the standard software provided by Zeiss.<br />

Using the general cell/DNA staining with DAPI and FISH with probes specific<br />

for the domain bacteria and group-specific probes (Table 1),bacteria can simultaneously<br />

be localized and identified at the rhizoplane.In addition to the groupspecific<br />

probes, in situ binding genus- and species-specific oligonucleotide<br />

probes are available for a number of root-associated and symbiotic bacteria<br />

(Ludwig et al. 1998; Hartmann et al. 2000). Figure 2A shows the localization of<br />

Azospirillum brasilense in the wheat rhizosphere by FISH (combination of two<br />

differently labeled oligonucleotide probes Eub338-Cy3 and Abras1420-Cy5)<br />

and CSLM. The application software “orthogonal view” of the LSM 510 (Zeiss,<br />

Germany) allows the display of optical cuts through the sample in xz and yz sections<br />

(Fig.2B).The localization within the tissue is clearly visible.<br />

2.2 Immunofluorescence Labeling Combined with Fluorescence in Situ<br />

Hybridization<br />

The combination of FISH, which allows a phylogenetic identification of bacteria<br />

from the phylum down to the species level, with immunological<br />

approaches extends the in situ identification to the individual strain level, if<br />

strain-specific antisera or special monoclonal antibodies are applied. Antibodies<br />

directed against bacterial <strong>surface</strong> antigens can be created by using,<br />

e.g., UV-inactivated bacteria as antigens (Schloter et al. 1995; Hartmann et al.<br />

1997). In addition, antibodies can also be created to identify specific enzymes,<br />

e.g., denitrifying enzymes (Bothe et al. 2000) and thus add a phenotypic or


454<br />

Anton Hartmann et al.<br />

B<br />

D


24 Microbial Community Analysis in the Rhizosphere 455<br />

expression level approach to organismic identification.As a basic protocol for<br />

combining FISH with immunofluorescence labeling, the procedure in Aßmus<br />

et al. (1997) can be used with some modifications in specific cases.<br />

After fixation of the sample and FISH analysis (see Sect. 2.1), the immunolabeling<br />

is performed with solutions containing 0.9 M NaCl. The presence of<br />

NaCl is necessary for the stability of the rRNA-oligonucleotide probe complex<br />

(Metz 2002). In addition, all incubation steps are performed at 4 °C. As usual,<br />

the immunolabeling procedure starts with a 1-h incubation of the slides carrying<br />

the samples with 3 % BSA (Frac. V) in 1/10 PBS+0.9 M NaCl to block<br />

unspecific binding of the antibody. After rinsing in washing solution (0.5 %<br />

BSA, 0.5 % Tween 80, 1/10 PBS, 0.9 M NaCl), the slides are incubated for 2.5 h<br />

at 4 °C with the specific antibody to be applied. After two washing steps, the<br />

second antibody (e.g., antimouse-FLUOS-Fab-Fragment) is applied at 4 °C for<br />

1.5 h. After washing, the slides are mounted in Citifluor AF1 (Citifluor Ltd.,<br />

London, UK). It has to be noted that not all monoclonal antibodies or polyclonal<br />

antisera are applicable to this protocol, because the antigen–antibody<br />

complex may not be stable at 0.9 M NaCl. Alternatively, the original protocol<br />

of Aßmus et al. (1997) can be applied, using the antibody treatment first and<br />

the fixation and FISH analysis second. Using this approach, strain-specific<br />

monoclonal antibodies against a specific Azospirillum brasilense strain were<br />

applied in situ together with the FISH analysis (Aßmus et al. 1997). Thus, the<br />

Fig. 2. In situ identification of bacteria in the rhizosphere using fluorescence-labeling<br />

techniques and CLSM. A Rhizosphere of wheat (Brazilian cultivar PF839197) inoculated<br />

with Azospirillum brasilense strain Sp245 (rgb-laser scanning image). Roots of inoculated,<br />

soil-grown wheat <strong>plant</strong>s were harvested 4 weeks after inoculation. After thorough<br />

washing in PBS, the root was cut manually, fixation by heat was performed for 30 min at<br />

70 °C and fixation in 3 % paraformaldehyde was done for 2 h at room temperature. Fluorescence<br />

in situ hybridization (FISH) was performed using 45 % formamide and the<br />

probes Eub338Mix-Cy3 and Abras1420-Cy5. A. brasilense Sp245 cells appear violet,<br />

because they bind two probes (red and blue emission color code) simultaneously. Plant<br />

cell walls have a different emission light, giving a green color code. B Same picture as A,<br />

but in the “orthogonal view”, providing insight into optical sections of the sample; zscan<br />

density: 21 mm. C In situ localization of GFP-labeled Serratia liquefaciens MG44 on<br />

root hairs of tomato <strong>plant</strong>s. Using 488-nm excitation wavelength, the GFP-labeled bacteria<br />

are clearly visible in the bright field picture. D Laser scanning microscopic picture of<br />

the same sample as C, but here two excitation wavelengths (488 and 560 nm) were used<br />

simultaneously, making the RFP-labeled Pseudomonas putida IsoF also visible. E Laser<br />

scanning microscopic picture of bacteria extracted from roots of Medicago sativa,inoculated<br />

with Sinorhizobium meliloti L33. The bacteria were treated as described and<br />

finally concentrated on polycarbonate filters. The fluorescence-labeled probes<br />

EuB338Mix-FLUOS and Rhi1247-TRITC were used in FISH analysis. Active bacteria<br />

with high ribosome content were labeled green (green arrow), while Rhizobia – obviously<br />

bacteroids released from nodules – appear yellow (yellow arrow), binding both<br />

probes simultaneously


456<br />

Anton Hartmann et al.<br />

root <strong>surface</strong> colonization by a particular bacterial strain could be investigated<br />

in a background of other members of this species, identified by using rRNAtargeting<br />

probes and FISH.<br />

2.3 Application of Fluorescence Tagging and Reporter Constructs<br />

The fate of particular bacterial inocula in the rhizosphere can also be monitored<br />

using molecular-tagged bacteria. In addition to the use of the visually<br />

detectable lux- and gus-markers (Lux: luciferase, Gus: b-glucuronidase), the<br />

exploitation of the green fluorescent protein (GFP) from the jellyfish<br />

Aequorea victoria has brought further progress into the field. GFP is a protein<br />

that contains a fluorescent cyclic tripeptide sequence. It requires only molecular<br />

oxygen for fluorescence, which means that GFP will fluoresce in virtually<br />

any aerobic organism (Lorang et al. 2001). Therefore, GFP-labeled bacteria<br />

can be observed by CLSM or by regular fluorescence microscopy. Figure 2C, D<br />

shows a localization of GFP-labeled Serratia liquefaciens MG44 in the rhizoplane<br />

of tomato. Furthermore, the application of DsRed from Discosoma sp.<br />

provides a red fluorescing molecular marker (Christensen et al. 1999; Tolker-<br />

Nielsen et al. 2000). In addition, a mutated form of GFP (ASV) with a short<br />

half-life enables real-time in situ expression studies (Andersen et al. 1998;<br />

Ramos et al. 2000).<br />

The application of GFP-labeling in expression studies using promotor-gfp<br />

fusions and GFP fusion proteins has revolutionized the in situ activity studies,<br />

because of the relative ease of recording the fluorescence microscopically.<br />

The bacteria carrying the gene constructs either on a plasmid or integrated<br />

into the chromosome are applied to sense or report conditions in the microhabitat<br />

they have been introduced. As in the case of simple tagging of organisms,<br />

not only lux- and gus-reporter (Kragelund et al. 1997) were used, but<br />

also constructs using the ice-nucleation gene (Loper and Henkels 1997), or<br />

the ferrichrom iron receptor (FhuA; Stubner et al. 1994). These constructs<br />

allowed the in situ sensing of N-, P- and C-starvation response (Kragelund et<br />

al. 1997; Koch et al. 2001), expression of nitrogen fixation genes (Egener et al.<br />

1999), presence of oxygen (Hojberg et al. 1999), availability of iron (Loper and<br />

Henkels 1997) general activity and cell number (Unge et al. 1999), genotoxic<br />

effects (Stubner et al. 1994) or the presence of quorum-sensing signal molecules<br />

of the N-acylhomoserine lactone type (Steidle et al. 2001). Figure 2C<br />

provides an example of in situ localization of GFP-labeled Serratia liquefaciens<br />

MG44 on root hairs in the rhizosphere of tomato as a bright field picture<br />

with 488-nm excitation wavelength, while Fig. 2D shows the same sample as<br />

CLSM-picture with two excitation wavelengths (560 and 488 nm) making the<br />

RFP-labeled Pseudomonas putida isoF also visible.<br />

In some of these studies, bacterial cells with reporter constructs need to<br />

be extracted from the habitat for analysis (Koch et al. 2001). Although these


24 Microbial Community Analysis in the Rhizosphere 457<br />

reporter cells monitor in situ conditions, the tests are performed ex situ. For<br />

this purpose, a separation of the bacteria from the soil was accomplished by<br />

applying formaldehyde (1 %)-fixed extracts to density gradient centrifugation<br />

with Nycodenz (Nycomed Pharma, Oslo, Norway) with a density of<br />

1.3 g/ml. After a centrifugation step (10,000xg, 30 min, 4 °C) the bacteria on<br />

the top of the Nycodenz layer were used for further analysis (Unge et al.<br />

1999).<br />

Monitoring of in situ bacterial growth activity in the <strong>plant</strong> rhizosphere is<br />

suggested by Ramos et al. (2001) using ribosome content and synthesis rate<br />

measurements.<br />

3 Ex Situ Studies of Microbial Communities After<br />

Separation of Rhizosphere Compartments<br />

For the desorption of bacteria from <strong>surface</strong>s, Campbell and Greaves (1990b)<br />

recommended the use of a stomacher. Sodium cholate and the ion exchange<br />

resin beads Dowex A1 or Chelex 100 were recommended for the treatment of<br />

soil particles or root pieces by Macdonald (1986) or Hopkins et al. (1991),<br />

respectively, to obtain the bacteria adsorbed. Herron and Wellington (1990)<br />

developed a method to extract streptomycete spores from soil particles and<br />

used polyethylene glycol (PEG) 6000 for reducing hydrophobic interactions.<br />

Each extraction protocol for root-associated bacteria has to be optimized for<br />

the system under investigation with the appropriate controls to prove its success.<br />

Mogge et al. (2000) described a standardized protocol for the differentiation<br />

of the rhizosphere compartments ectorhizosphere and rhizoplane/<br />

endorhizosphere and the extraction of the adsorbed bacteria from the rhizoplane<br />

of Medicago sativa europae. This procedure used the recommendations<br />

by Macdonald (1986) and Herron and Wellington (1990) in a modified form.<br />

FISH in combination with CLSM was applied for the proof of desorption efficiency<br />

in root <strong>surface</strong> studies.<br />

3.1 Recovery of Bacteria from Bulk Soil, Ecto- and Endorhizosphere<br />

Roots are carefully separated from the soil using sterile tweezers. The soil<br />

should be rather dry at the time of harvest to facilitate the separation of roots<br />

from the adhering soil. All steps are conducted with sterile solutions on ice.<br />

Bulk soil (compartment I) and root-attached soil particles which have been<br />

collected by shaking the roots (ectorhizosphere: compartment II) are suspended<br />

1:9 (w/v) in 0.01 M phosphate buffer (Na 2HPO 4/KH 2PO 4, pH 7.4) and<br />

dispersed for 1 min at the highest speed in a Stomacher 80 (Seward Medical,<br />

UK). To extract rhizoplane and endorhizosphere bacteria (compartment III),<br />

1 g (fresh weight) of roots that have been cleaned from adhering soil particles


458<br />

Anton Hartmann et al.<br />

(see above) and washed in phosphate buffer is suspended in 20 ml 0.1 %<br />

sodium-cholate buffer (Macdonald 1986). The suspension is treated in a<br />

Stomacher 80 at the highest speed for 4 min to disrupt polymers. After transfer<br />

into Erlenmeyer flasks, 0.5 g of polyethylene glycol 6000 (Sigma, Deisenhofen)<br />

and 0.4 g of cation change polystyrene beads (chelex 100: Sigma,<br />

Deisenhofen) are added and the suspension is stirred at 50 rpm/min for 1 h at<br />

4 °C. The stomacher/stirring procedure is repeated three times, whereby the<br />

roots are transferred to “fresh” 0.1 % sodium cholate buffer with PEG 6000<br />

and chelex 100 after each extraction step (compartment IIIa-c). Finally,<br />

aliquots of the obtained suspensions are combined. Root and soil particles are<br />

removed by filtration through gauze (40-mm mesh width) and subsequent filtration<br />

through 5-mm syringe filters (Sartorius No. 17549, Göttingen, Germany).<br />

In the case of Medicago sativa grown in sandy loam, this approach yielded<br />

total counts of 3.3x10 9 to 6.5x10 8 /g root dry weight from the first to the third<br />

treatment, while hybridizing bacteria remained constant at 1.5x10 8 /g root dry<br />

weight (Mogge et al. 2000). It was calculated that about 88 % of the bacteria<br />

had been desorbed from the rhizoplane by this technique. This result was confirmed<br />

by in situ studies of roots applying confocal laser scanning<br />

microscopy. The roots usually harbor large numbers of phylogenetically different<br />

bacteria, belonging, e.g., to the a-, b- and g-subclasses of proteobacteria.<br />

However, after the third extraction step, no bacteria could be detected any<br />

more on the root <strong>surface</strong> (20 root pieces of 2–3 cm length were scanned).<br />

The suspensions obtained from bulk soil (I), ectorhizosphere (II), and rhizoplane/endorhizosphere<br />

(IIIa-c: merged suspension) can be used for cultivation<br />

and dot blot-hybridizations (see Sect. 3.2). DAPI-staining and FISH can<br />

be applied for counting total and hybridizing bacteria in the three compartments<br />

collected on polycarbonate filters (see Sect. 3.3). PCR-amplification of<br />

16S rDNA and subsequent electrophoretic fingerprinting of the amplification<br />

products as well as clone bank studies can be performed with these fractions<br />

too (see Sect. 3.4). In addition, these compartments can be investigated for<br />

structural and functional microbial diversity by community fatty acid analysis<br />

and community level physiological profiling (see Sect. 3.5).<br />

3.2 Community Analysis by Cultivation and Dot Blot Studies<br />

Serial dilutions (0.85 % NaCl) from bulk soil (compartment I), ectorhizosphere<br />

(compartment II), and rhizoplane/endorhizosphere (compartment III)<br />

suspensions (Fig. 1) were plated onto agar media containing different nutritional<br />

levels (Table 2). The selection of media used for the isolation of soil and<br />

ectorhizosphere-associated bacteria was made to allow the growth of oligotrophic,<br />

slow growing strains as well as fast growers. Minimal media were<br />

suggested because of the sensitivity of soil bacteria to salts (NaCl) or organic


24 Microbial Community Analysis in the Rhizosphere 459<br />

compounds (yeast extracts, casamino acids) as described by Hattori and Hattori<br />

(1980). On the other hand, depending on the lower growth rate and a<br />

longer incubation period, exuberant growth of the fast growers was reduced,<br />

giving the slow growing strains a chance to develop (Gorlach et al. 1994; Mitsui<br />

et al. 1997). In addition, minimal media like M9, were supplemented with<br />

compounds described as root exudates, and with soil or root extracts<br />

(Table 2). Plates were incubated at 20 °C for up to 4 weeks. Cell and colony<br />

morphology was recorded and Gram-test, oxidase and catalase tests performed<br />

according to Gerhardt et al. (1994). Genomic DNA of these isolates<br />

was extracted and purified as described previously (Pukall et al. 1998). The<br />

primer pair 27f and 1500r can be used for the amplification of the almost<br />

complete 16S rRNA gene of the bacterial isolates (Lane 1991). PCR-amplification<br />

of a part of the 23S rDNA was performed using the primer pair 2053r and<br />

990 f.<br />

Using this approach, about 70 % of the bacterial isolates from bulk soil and<br />

ectorhizosphere were identified as Gram-positive bacteria using the oligonucleotide<br />

GP (Rheims et al. 1996), whereas their numbers were reduced to 17 %<br />

in the rhizoplane/endorhizosphere compartment of Medicago sativa (Mogge<br />

et al. 2000). A similar result was obtained by Lilley et al. (1996) and Mahaffee<br />

and Kloepper (1997). On the other hand, the numbers of isolates belonging to<br />

the a-, b-, and g-subclasses of proteobacteria were increased in the rhizoplane<br />

Table 2. Composition of media used to retrieve bacteria from bulk soil, ectorhizosphere<br />

and rhizoplane/endorhizosphere samples<br />

Medium Company or reference<br />

King’s B agar; R2A agar; Actinomycete isolation Difco<br />

agar; nutrient agar<br />

CASO agar Merck<br />

Yeast extract mannitol agar Dunger and Fiedler (1997)<br />

Starch agar with and without root extract Dunger and Fiedler (1997)<br />

Cellulose agar supplemented with soil extract Stotzky et al. (1993)<br />

Planctomyces isolation agar(+N-acetylglucosamin) Schlesner (1994)<br />

Hyphomicrobium isolation agar Moore and Marshall (1981)<br />

Caulobacter isolation agar Poindexter (1964)<br />

Glucose-yeast extract malt agar (GYM) Shirling and Gottlieb (1966)<br />

M9 minimal medium a (+ carbon source b / Sambrook et al. (1989, modified)<br />

+ trace elements c )<br />

a Composed of Na2 HPO 4 10.2, KH 2 PO 4 3.0, NaCl 0.6, and NH 4 Cl 1.2 g/l<br />

b 5 g/l carbohydrates (glucose, glucose and vitamin solution No.6 (Staley 1968), fructose,<br />

sucrose, arabinose) or 2 g/l organic acids (fumaric acid, oxal acetic acid)<br />

c 1 ml of sterile filtered trace element stock solution composed of CaCl2 x6 H 2 O 2.7 g,<br />

MgSO 4 x7 H 2 O 15 g, FeCl 3 0.02 g/l


460<br />

Anton Hartmann et al.<br />

to 13, 26 and 35 % as compared to 4.2, 8.5 and 0.8 % respectively in the ectorhizosphere<br />

as was shown by using the probes ALF1b, BET42a and GAM42<br />

respectively to group the isolates obtained. No differences were found for isolates<br />

of the Cytophaga-Flavobacteria group, which were only a minor portion<br />

in both compartments (3.5 %).<br />

Quantitative population analyses in soil and rhizosphere environments<br />

were also conducted by using strains carrying unique selectable markers. This<br />

was aimed to enumerate one particular introduced strain in the presence of a<br />

large excess of other microbes. Since the usually suitable selectable markers<br />

are missing in wild-type strains, spontaneous or transposon-induced<br />

mutants, which are, e.g., resistant to an antibiotic, are frequently used for<br />

selective plating assays. However, these mutants may be less fit than the wild<br />

type and, therefore, the results of the surveys are biased. De Leij et al. (1998)<br />

demonstrated such effects on environmental fitness in several mutants of<br />

Pseudomonas fluorescens SBW25, constructed by site-directed genomic insertions<br />

of marker genes. Recently, Hirano et al. (2001) selected a site in the gacScysM<br />

intergenic region in Pseudomonas syringae pv. syringae B728, in which<br />

the insertion of an antibiotic resistance marker cassette did not affect the fitness<br />

of the bacterium in the field. They concluded that carefully selected<br />

intergenic regions, which are suitable for the integration of specific marker<br />

cassettes, exist in any bacterium.<br />

3.3 Community Analysis by Fluorescence in Situ Hybridization on<br />

Polycarbonate Filters<br />

Bacterial suspensions (extract of the rhizosphere compartments, Fig. 1) are<br />

fixed overnight at 4 °C with 3 % formaldehyde and concentrated in three parallels<br />

onto 0.2-mm polycarbonate filters (100-ml aliquots). Dehydration of cells<br />

is performed with 50, 80 and 96 % ethanol for 3 min each. For details on the<br />

FISH protocol see Sect. 2.1. The slides are finally mounted with Citifluor AF1<br />

to reduce photobleaching. A Zeiss Axiophot 2 epifluorescence microscope<br />

(Zeiss, Jena, Germany) equipped with filter sets F31–000, F41–001 and<br />

F41–007 (Chroma Tech. Corp., Battleboro, VT, USA) can be used for the enumeration<br />

of bacteria on filters. Total cell counts (DAPI) and hybridizing bacteria<br />

using a set of domain-specific probes (Table 1) are determined by evaluating<br />

at least 10 microscopic fields with 20–100 cells per field.<br />

In the case of the M. sativa roots, the extraction method was also applied to<br />

the rhizoplane/endorhizosphere of roots inoculated with Sinorhizobium<br />

meliloti as well as to inoculated roots after the nodules had been removed<br />

with a sterile scalpel. During the three repeated stomacher/stirring-treatments,<br />

nodules cracked and S. meliloti-bacteroids were released (Mogge et al.<br />

2000). Figure 2E shows a representative photomicrograph of bacteria concentrated<br />

on polycarbonate filters after extraction of roots with nodules. Large


24 Microbial Community Analysis in the Rhizosphere 461<br />

(up to 10-mm long) pleomorphic cells hybridized with a set of FLUOS-labeled<br />

oligonucleotide probes directed against the domain Bacteria and the TRITClabeled<br />

oligonucleotide probe Rhi1247 directed against Rhizobium (Table 1).<br />

Obviously, these large cells were bacteroids originating from crushed nodules<br />

and were missing when the nodules had been removed before the application<br />

of the extraction procedure.<br />

3.4 Community Analysis by (RT) PCR-Amplification of Phylogenetic<br />

Marker Genes, D/TGGE-Fingerprinting and Clone Bank Studies<br />

The differentiated rhizosphere compartments can also be used to isolate<br />

rRNA and genomic DNA following previously described protocols (Felske et<br />

al. 1996; Miethling et al. 2000).A further purification of the DNA extracts, e.g.,<br />

with the Wizard DNA clean-up (Promega, Madison, WI), may be necessary,<br />

before PCR can be applied. For amplification, the highly conserved bacterial<br />

16S rRNA primers U968-GC and L1346 are used.Amplification of 16S rDNA is<br />

performed as described by Felske et al. (1996) using the following PCR-program:<br />

1 cycle at 94 °C for 5 min, 35 cycles at 94 °C for 90 s (denaturation), 61 °C<br />

for 40 s (annealing), 70 °C for 40 s (extension), and a single final extension at<br />

70 °C for 5 min. Amplification of 16S rRNA as well as denaturing temperature<br />

gradient gel electrophoretic (D/TGGE) separation of the PCR-products of<br />

DNA and RNA is performed as described by Miethling et al. (2000).<br />

D/TGGE profiles represent the frequency distribution of PCR-amplified<br />

segments of rDNA or rRNA separated due to their melting behavior in the<br />

electric field of a temperature gradient gel. The resulting profiles represent the<br />

frequency distribution of the most prominent community members in a first<br />

approximation (Muyzer and Smalla 1998). Since the ratio of 16S rDNA and<br />

16S rRNA is dependent on cellular activity (Wagner 1994), comparisons of<br />

TGGE patterns derived from 16S rRNA and 16S rDNA amplicons can provide<br />

interesting information about the active members of the community. Variations<br />

in the relation of band intensities (rRNA/rDNA) indicated shifts in the<br />

relative activity of the respective dominant DNA sequences. In particular, the<br />

composition of the communities are changing along the gradient from bulk<br />

soil to the rhizoplane/endorhizosphere (Mogge et al. 2000, Wieland et al.<br />

2001). Additional sequences show higher evenness visible by larger band formation<br />

in the rhizoplane/endorhizosphere compartment, which is clearly different<br />

from all the other examined habitats. In this compartment, a larger<br />

fraction of the community seems to be active, as deduced from the fraction of<br />

bands common to the rDNA and rRNA patterns of the communities. Using<br />

the same methodological approach, Wieland et al. (2001) have demonstrated<br />

recently that the TGGE-patterns of 16S rRNA did not change during the <strong>plant</strong><br />

development in the bulk soil, whereas some pattern variation could be correlated<br />

to <strong>plant</strong> development in the rhizosphere and rhizoplane habitats. On the


462<br />

Anton Hartmann et al.<br />

root <strong>surface</strong> of different <strong>plant</strong>s and <strong>plant</strong>s growing in different soils, more<br />

apparent differences in the complete TGGE-pattern was obvious. The frequency<br />

distribution of target sequences from the total and active community<br />

members appeared to be mostly identical at the rhizoplane/endorhizosphere<br />

where the most prominent bands of the rRNA-derived pattern are also dominant<br />

in the DNA pattern. However, it has to be taken into account that a high<br />

ribosome content does not always indicate a high physiological activity of<br />

bacterial cells, because different bacteria inherently contain different ribosome<br />

numbers (Fegatella et al. 1998). It is likely that both phenomena play a<br />

role, and this may be different for different bacterial groups (Duarte et al.<br />

1998). Duineveld et al. (2001) applied a similar 16S rDNA/rRNA PCR-amplification<br />

approach followed by DGGE analysis in the Chrysantemum rhizosphere,<br />

but found very little difference between the bacterial community of<br />

root-adhering soil and bulk soil. Heuer et al. (2002) used not only general<br />

PCR-primers for the amplification of bacterial 16S rDNA (between positions<br />

968 and 1401, E. coli numbering according to Brosius et al. 1981), but also the<br />

taxon-specific primers F203alpha for alpha-proteobacteria and F964b for bproteobacteria.<br />

Using this approach, these authors revealed a more differentiated<br />

fingerprint for rhizosphere bacterial communities in DGGE-electrophoresis.<br />

A PCR approach targeting the ribosomal 16S–23S rDNA<br />

intergenic spacer region, called ribosomal intergenic spacer analysis (RISA),<br />

can also reveal insight into the bacterial diversity, because this spacer region<br />

varies considerably in different species. Baudoin et al. (2001) applied this<br />

approach for the assessment of the bacterial community structure along<br />

maize roots and in different growth stages. Weidner et al. (1996) applied<br />

restriction fragment length polymorphism (RFLP) analysis of cloned 16S<br />

rDNA from the roots of the seagrass Halophila stipulacea to investigate unculturable<br />

bacterial rhizosphere communities. Finally, a strain-specific detection<br />

of certain bacterial strains in the rhizosphere based on a highly specific PCRamplification<br />

of the 16S–23S intergenic spacer (IGS) region was recently<br />

developed by Tan et al. (2001). The sequence variability in this region was<br />

used to differentially identify Bradyrhizobium and Rhizobium strains colonizing<br />

rice roots by a nested PCR approach and analysis of the amplification<br />

products on simple agarose gels.<br />

The genomic DNA extracted from the rhizosphere compartments I–III<br />

(Fig. 2) can also be used to create 16S rDNA clone banks or dot blot experiments<br />

with 16S rDNA fragments or probing with specific oligonucleotides.<br />

When the oligonucleotide GP (Rheims et al. 1996) was used, a reduced number<br />

of 16S rDNA clones related to Gram-positive bacteria was detected in the<br />

library generated from the rhizoplane/endorhizosphere of Medicago sativa<br />

(12 %) as compared to the library generated from the bulk soil fraction (26 %;<br />

Mogge et al. 2002). Thus, the results of community analysis using cultivation<br />

techniques and FISH analysis (see Sects. 3.2 and 3.3) were, in general, confirmed<br />

by this PCR-based cultivation independent technique.


3.5 Community Analysis by Fatty Acid Pattern and Community Level<br />

Physiological Profile Studies<br />

The overall microbial diversity in environmental habits can be assessed by<br />

cultivation independent biomarker analysis, different from the phylogenetic<br />

ribosomal genes or other genetic markers. As is the case in chemotaxonomic<br />

studies, the fatty acid patterns are used for this purpose. In one type of analysis,<br />

the fatty acid methyl esters (FAME) are obtained from the fatty acids after<br />

saponification of 5 g of soil or root with adhering soil in methanoic NaOH (at<br />

100 °C, 30 min; Dunfield and Germida 2001). Alternatively, the lipids are<br />

extracted from 5 g of soil with methanol:chloroform (2:1), the phospholipids<br />

are separated by chromatography, and finally hydrolyzed to liberate the phospholipid<br />

fatty acids (PLFA; White and Ringelberg 1998). The PLFA analysis<br />

has the advantage of giving insight into the living community, because PFLA<br />

are efficiently hydrolyzed in dead biomass, while the direct FAME analysis<br />

may contain fatty acids from dead organisms too. The GC-MSanalysis finally<br />

provides much information on the diversity of this biomarker (Zelles 1997;<br />

White and Ringelberg 1998). Using the FAME analysis, Germida et al. (1998)<br />

investigated the diversity of root-associated bacterial communities in canola<br />

and wheat, and Dunfiled and Germida (2001) compared the bacterial communities<br />

in the rhizosphere and endorhizosphere of field-grown genetically<br />

modified varieties of canola (Brassica napus).An example of a recent application<br />

of the PFLA approach in rhizosphere studies is the investigation of the<br />

microbial community response in the rhizosphere of Spartina alterniflora to<br />

changing environmental conditions by Lovell et al. (2001).<br />

An investigation targeting the analysis of the functional abilities of a complex<br />

community is the substrate utilization profile assays using the Biolog R -<br />

plates. Baudoin et al. (2001) applied this approach recently to characterize the<br />

functional microbial diversity in different rhizosphere compartments of<br />

maize <strong>plant</strong>s. The differences between the rhizosphere and nonrhizosphere<br />

soil samples were more pronounced in 4-week-old compared to 2-week-old<br />

<strong>plant</strong>s. In addition, adhering soil from different root zones (ramification, root<br />

hair-elongation, root tip) revealed dissimilar community level physiological<br />

profiles (CLPP). However, this approach needs to be regarded as reflecting the<br />

potential rather than the in situ-activity of most culturable microbes, because<br />

these are known to respond and contribute most to the activity at the incubation<br />

conditions of the CLPP-assay (Garland et al. 1997).<br />

4 Conclusions<br />

24 Microbial Community Analysis in the Rhizosphere 463<br />

Using a polyphasic approach including cultivation-dependent and different<br />

cultivation-independent methods, it could be shown that a high proportion of<br />

culturable bacteria is present in the rhizoplane when a variety of appropriate


464<br />

Anton Hartmann et al.<br />

media are applied. This corroborates the findings of Hengstmann et al. (1999),<br />

who reported similar results in their studies on the microbial community of<br />

the rice rhizosphere. The separation into the three compartments, bulk soil,<br />

ectorhizosphere and rhizoplane/endorhizosphere has to be performed with<br />

great care and actually needs an optimization for each <strong>plant</strong> and soil type<br />

under study. The degree to which adhering soil particles (ectorhizosphere)<br />

are included in the rhizosphere studies considerably influences the outcome<br />

of the study, since these soil particles are carrying a microbial community<br />

resembling, to a varying extent, the soil situation compared to the root <strong>surface</strong><br />

or rhizoplane situation. The microbial population colonizing the root <strong>surface</strong><br />

should be approached only after washing the roots free of adhering soil particles.<br />

In conclusion, the way “rhizosphere” is defined by the experimental protocol<br />

is of crucial importance for the results of root colonization studies.<br />

Certainly, in situ and ex situ studies (with the separated rhizosphere compartments)<br />

both complement each other to give a more comprehensive picture.<br />

Although the microscopic in situ approach has the great advantage of<br />

providing detailed spatial information about root <strong>surface</strong> colonization, quantitative<br />

and qualitative data about the structural and functional diversity of<br />

root colonization can be obtained by a variety of complementary ex situ<br />

approaches.<br />

References and Selected Reading<br />

Amann RI, Binder BJ, Olson RJ, Chisholm SW, Devereux R, Stahl DA (1990) Combination<br />

of 16S rRNA-targeted oligonucleotide probes with flow cytometry for analyzing<br />

mixed microbial populations. Appl Environ Microbiol 56:1919–1925<br />

Amann RI, Ludwig W, Schleifer KH (1995) Phylogenetic identification and in situ detection<br />

of individual microbial cells without cultivation. Microbiol Rev 59:143–169<br />

Andersen JB, Sternberg C, Poulsen LK, Bjorn SP, Givskov M, Molin S (1998) New unstable<br />

variants of green fluorescent protein for studies of transient gene expression in<br />

bacteria. Appl Environ Microbiol 64:2240–2246<br />

Aßmus B, Hutzler P, Kirchhof G, Amann RI, Lawrence JR, Hartmann A (1995) In situ<br />

localization of Azospirillum brasilense in the rhizosphere of wheat with fluorescently<br />

labeled, rRNA-targeted oligonucleotide probes and scanning confocal laser microscopy.<br />

Appl Environ Microbiol 61:1013–1019<br />

Aßmus B, Schloter M, Kirchhof G, Hutzler P, Hartmann A (1997) Improved in situ tracking<br />

of rhizosphere bacteria using dual staining with fluorescence-labeled antibodies<br />

and rRNA-targeted oligonucleotides. Microbial Ecol 33:32–40<br />

Baudoin E, Benizri E, Guckert A (2001) Impact of growth stage on the bacterial community<br />

structure along maize roots, as determined by metabolic and genetic fingerprinting.<br />

Appl Soil Ecol 52:1–11<br />

Braun-Howland EB, Vescio PA, Nierzwicki-Bauer SA (1993) Use of a simplified cell blot<br />

technique and 16S rRNA-directed probes for identification of common environmental<br />

isolates. Appl Environ Microbiol 59:3219–3224<br />

Beringer JE (1974) R factor transfer in Rhizobium leguminosarum. J Gen Microbiol<br />

84:188–198


24 Microbial Community Analysis in the Rhizosphere 465<br />

Bothe H, Jost G, Schloter M, Ward BB, Witzel KP (2000) Molecular analysis of ammonia<br />

oxidation and denitrification in natural environments. FEMS Microbiol Rev 24:<br />

673–690<br />

Brimecombe MJ, De Leij FA, Lynch JM (2001) The effect of root exudates on rhizosphere<br />

microbial populations. In: Pinton R, Varanini Z, Nannipieri P (eds) The rhizosphere.<br />

Marcel Dekker, New York, pp 95–140<br />

Brosius J, Dull TJ, Sleeter DD, Noller HF (1981) Gene organization and primary structure<br />

of a ribosomal RNA operon from Escherichia coli. J Mol Biol 148:107–127<br />

Campbell R, Greaves MP (1990a) Anatomy and community structure of the rhizosphere.<br />

In: Lynch JM (ed) The rhizosphere. Wiley, Chichester, pp 11–34<br />

Campbell R, Greaves MP (1990b) Methods for studying the microbial ecology of the rhizosphere.<br />

Meth Microbiol 22:447–477<br />

Chatzinotas A, Sandaa RA, Schönhuber W,Amann R, Daae FL, Torsvik V, Zeyer J, Hahn D<br />

(1998) Analysis of broad-scale differences in microbial community composition of<br />

two pristine forest soils. Syst Appl Microbiol 21:579–587<br />

Christensen BB, Sternberg C, Andersen JB, Palmer Jr RJ, Nielsen JJ, Givskov M, Molin S<br />

(1999) Molecular tools for study of biofilm physiology. Meth Enzymol 310:20–42<br />

De Leij FAAM, Thomas CE, Bailey MJ, Whipps JM, Lynch JM (1998) Effect of insertion<br />

site and metabolic load on the environmental fitness of a genetically modified<br />

Pseudomonas fluorescens isolate. Appl Environ Microbiol 64:2634–2638<br />

Duarte GF, Rosado AS, Seldin L, Keijzer-Wolter AC, Van Elsas JD (1998) Extraction of<br />

ribosomal RNA and genomic DNA from soil for studying the diversity of the indigenous<br />

bacterial community. J Microbiol Meth 32:21–29<br />

Duineveld BM, Kowalchuk GA, Keijzer A, van Elsas JD, van Veen J (2001) Analysis of bacterial<br />

communities in the rhizosphere of Chrysanthemum via denaturing gradient gel<br />

electrophoresis of PCR-amplified 16S rRNA as well as DNA fragments coding for 16S<br />

rRNA. Appl Environ Microbiol 67:172–178<br />

Dunfield KE, Germida JJ (2001) Diversity of bacterial communities in the rhizosphere<br />

and root interior of field-grown genetically modified Brassica napus. FEMS Microbiol<br />

Rev 38:1–9<br />

Dunger W, Fiedler HJ (1997) Methoden der Bodenbiologie. Gustav Fischer-Verlag, Jena,<br />

pp 89–107<br />

Egener T, Hurek T, Reinhold-Hurek B (1999) Endophytic expression of nif genes of<br />

Azoarcus sp. strain BH72 in rice roots. Mol Plant-Microbe Interact 12:813–819<br />

Fegatella F, Lim J, Kjelleberg S, Cavicchiolli R (1998) Implications of rRNA operon copy<br />

number and ribosome content in the marine oligotrophic ultramicrobacterium<br />

Sphingomonas sp. strain RB2256. Appl Environ Microbiol 64:4433–4438<br />

Felske A, Engelen B, Nübel U, Backhaus H (1996) Direct ribosome isolation from soil to<br />

extract bacterial rRNA for community analysis. Appl Environ Microbiol 62:4162–<br />

4167<br />

Garland JL, Cook KL, Loader CA, Hungate BA (1997) The influence of microbial community<br />

structure and function on community-level physiological profiles. In: Insam<br />

H, Rangger A (eds) Microbial communities: functional versus structural approaches.<br />

Springer, Berlin Heidelberg New York, pp 171–183<br />

Gerhardt P, Murray RGE,Wood WA, Krieg NR (1994) Methods for general molecular bacteriology.<br />

American Society for Microbiology, Washington, DC<br />

Germida JJ, Siciliano SD, de Freitas JR, Seib AM (1998) Diversity of root-associated bacteria<br />

associated with field-grown canola (Brassica napus L.) and wheat (Triticum aestivum<br />

L.) FEMS Microbiol Ecol 26:43–50<br />

Giovannoni SJ, DeLong EF, Olsen GJ, Pace NR (1988) Phylogenetic group-specific<br />

oligodeoxynucleotide probes for identification of single microbial cells. J Bacteriol<br />

170:720–726


466<br />

Anton Hartmann et al.<br />

Gorlach K, Shingaki R, Morisaki H, Hattori T (1994) Construction of eco-collection of<br />

paddy field soil bacteria for population analysis. J Gen Microbiol 40:509–517<br />

Hartmann A, Aßmus B, Kirchhof G, Schloter M (1997) Direct approaches to study soil<br />

microflora. In: van Elsas JD, Trevors JT, Wellington EMH (eds) Modern soil <strong>microbiology</strong>.<br />

Marcel Dekker, New York, pp 279–309<br />

Hartmann A, Lawrence JR, Aßmus B, Schloter M (1998) Detection of microbes by laser<br />

confocal microscopy. In: Akkermans ADL, van Elsas JD, de Bruijn FJ (eds) Molecular<br />

microbial ecology manual, Supplement 3. Kluwer, Dordrecht, Chap. 4.1.10<br />

Hartmann A, Stoffels M, Eckert B, Kirchhof G, Schloter M (2000) Analysis of the presence<br />

and diversity of diazotrophic endophytes. In: Triplett EW (ed) Prokaryotic nitrogen<br />

fixation: A model system for analysis of a biological process. Horizon Scientific Press,<br />

Wymondham, USA, pp 727–736<br />

Hattori R, Hattori T (1980) Sensitivity to salts and organic compounds of soil bacteria<br />

isolated on diluted media. J Gen Appl Microbiol 26:1–14<br />

Hengstmann U, Chin KJ, Janssen PH, Liesack W (1999) Comparative phylogenetic<br />

assignment of environmental sequences of genes encoding 16S rRNA and numerically<br />

abundant culturable bacteria from an anoxic rice paddy soil. Appl Environ<br />

Microbiol 65:5050–5058<br />

Herron PR, Wellington EMH (1990) New method for extraction of streptomycete spores<br />

from soil and application to the study of lysogene in sterile amended and nonsterile<br />

soil. Appl Environ Microbiol 56:1406–1412<br />

Heuer H, Kroppenstedt RM, Lottmann J, Berg G, Smalla K (2002) Effects of T4 lysozyme<br />

release from transgenic potato roots on bacterial rhizosphere communities are negligible<br />

relative to natural factors. Appl Environ Microbiol 68:1325–1335<br />

Hirano SS, Willis DK, Clayton MK, Upper CD (2001) Use of an intergenic region in<br />

Pseudomonas syringae pv. syringae B728a for site-directed genomic marking of bacterial<br />

strains for field experiments. Appl Environ Microbiol 67:3735–3738<br />

Hojberg O, Schnider U, Winteler HV, Sorensen J, Haas D (1999) Oxygen-sensing reporter<br />

strain of Pseudomonas fluorescens for monitoring the distribution of low-oxygen<br />

habitats in soil. Appl Environ Microbiol 65:4085–4093<br />

Hopkins DW, MacNaughton SJ, O’Donnell AG (1991) A dispersion and differential centrifugation<br />

technique for representatively sampling microorganisms from soil. Soil<br />

Biol Biochem 23:217–225<br />

Koch B, Worm J, Jensen LE, Hojberg O, Nybroe O (2001) Carbon limitation induces sigma<br />

s -dependent gene expression in Pseudomonas fluorescens in soil. Appl Environ<br />

Microbiol 67:3363–3370<br />

Kragelund L, Hosbond C, Nybroe O (1997) Distribution of metabolic activity and phosphate<br />

starvation response of lux-tagged Pseudomonas fluorescens reporter bacteria in<br />

the barley rhizosphere. Appl Environ Microbiol 63:4920–4928<br />

Lane DJ (1991) 16S/23S rRNA sequencing. In: Stackebrandt E, Goodfellow M (eds)<br />

Nucleic acid techniques in bacterial systematics. Wiley, Chichester, pp 125–175<br />

Lee S, Malone C, Kemp PF (1993) Use of multiple 16S rRNA-targeted fluorescent probes<br />

to increase signal strength and measure cellular RNA from natural planktonic bacteria.<br />

Mar Ecol Prog Ser 101:193–201<br />

Liesack W, Stackebrandt E (1992) Occurrence of novel groups of the domain bacteria as<br />

revealed by analysis of genetic material isolated from an Australian terrestrial environment.<br />

J Bacteriol 174:5072–5078<br />

Liesack W, Janssen PH, Rainey FA, Ward-Rainey N, Stackebrandt E (1997) Microbial<br />

diversity in soil: the need for a combined approach using molecular and cultivation<br />

techniques. In: van Elsas JD, Trevors JT, Wellington EMH (eds) Modern soil <strong>microbiology</strong>.<br />

Marcel Dekker, New York, pp 375–439


24 Microbial Community Analysis in the Rhizosphere 467<br />

Lilley AK, Fry JC, Bailey MJ, Day MJ (1996) Comparison of aerobic heterotrophic taxa<br />

isolated from four root domains of mature sugar beet (Beta vulgaris). FEMS Microbiol<br />

Ecol 21:231–242<br />

Loper JE, Henkels MD (1997) Availability of iron to Pseudomonas fluorescens in rhizosphere<br />

and bulk soil evaluated with an ice nucleation reporter gene. Appl Environ<br />

Microbiol 60:2944–2948<br />

Lorang JM, Tuori RP, Martinez JP, Sawyer TL, Redman RS, Rollins JA, Wolpert TJ, Johnson<br />

KB, Rodriguez RJ, Dickman MB, Ciuffetti LM (2001) Green fluorescent protein is<br />

lighting up fungal biology. Appl Environ Microbiol 67:1987–1994<br />

Lovell CR, Bagwell CE, Czákó M, Márton L, Piceno YM, Ringelberg DB (2001) Stability of<br />

a rhizosphere microbial community exposed to natural and manipulated environmental<br />

variability. FEMS Microbiol Ecol 38:69–76<br />

Ludwig W, Amann R, Martinez-Romero E, Schönhuber W, Bauer S, Neef A, Schleifer KH<br />

(1998) rRNA based identification and detection systems for rhizobia and other bacteria.<br />

Plant Soil 204:1–19<br />

Macdonald RM (1986) Sampling soil microfloras: dispersion of soil by ion exchange and<br />

extraction of specific microorganisms from suspension by elutriation. Soil Biol<br />

Biochem 18:399–406<br />

Mahaffee WF, Kloepper JW (1997) Temporal changes in the bacterial communities of<br />

soil, rhizosphere, and endorhiza associated with field-grown cucumber (Cucumis<br />

sativus L.). Microb Ecol 34:210–223<br />

Manz W, Amann R, Ludwig W, Wagner M, Schleifer KH (1992) Phylogenetic oligodeoxynucleotide<br />

probes for the major subclasses of proteobacteria: problems and<br />

solutions. System Appl Microbiol 15:593–600<br />

Manz W, Amann R, Ludwig W, Vancanneyt M, Schleifer KH (1996) Application of a suite<br />

of 16S rRNA-specific oligonucleotide probes designed to investigate bacteria of the<br />

phylum cytophaga-flavobacter-bacteroides in the natural environment. Microbiology<br />

142:1097–1106<br />

Metz S (2001) Herstellung von monoklonalen Antikörpern gegen die Cu-abhängige dissimilatorische<br />

Nitritreduktase und deren Anwendung zum in situ-Nachweis der Denitrifikationsaktivität<br />

von Bakterien. Doctoral Thesis, Ludwig-Maximilians-Universität<br />

München, Fakultät für Biologie<br />

Miethling R, Wieland G, Backhaus H, Tebbe CC (2000) Variation of microbial rhizosphere<br />

communities in response to crop species, soil origin and inoculation with the<br />

marker gene-tagged Sinorhizobium meliloti L33. Microb Ecol 40:43–56<br />

Mitsui H, Gorlach K, Lee HJ, Hattori R, Hattori T (1997) Incubation time and media<br />

requirements of culturable bacteria from different phylogenetic groups. J Microbiol<br />

Methods 30:103–110<br />

Mogge B, Lebhuhn M, Schloter M, Stoffels M, Pukall R, Stackebrandt E, Wieland G, Backhaus<br />

H, Hartmann A (2000) Erfassung des mikrobiellen Populationsgradienten vom<br />

Boden zur Rhizoplane von Luzerne (Medicago sativa). In: Hartmann A (ed) Biologische<br />

Sicherheit: Biomonitor und Molekulare Mikrobenökologie. Projektträger BEO,<br />

Jülich, pp 217–224<br />

Moore RL, Marshall KC (1981) Attachment and rosette formation by hyphomicrobia.<br />

Appl Environ Microbiol 42:751–757<br />

Morgan JAW, Whipps JM (2001) Methodological approaches to the study of rhizosphere<br />

carbon flow and microbial population dynamics. In: Pinton R,Varanini Z, Nannipieri<br />

P (eds) The rhizosphere. Marcel Dekker, New York, pp 373–409<br />

Muyzer G, Smalla K (1998) Application of denaturing gradient gel electrophoresis<br />

(DGGE) and temperature gradient gel electrophoresis (TGGE) in microbial ecology.<br />

Antonie van Leuwenhook 73:127–141


468<br />

Anton Hartmann et al.<br />

Poindexter JS (1964) Biological properties and classification of the Caulobacter group.<br />

Bacteriol Rev 28:231–295<br />

Pukall R, Brambilla E, Stackebrandt E (1998) Automated fragment length analysis of fluorescently-labeled<br />

16S rDNA after digestion with 4-base cutting restriction enzymes.<br />

J Micobiol Meth 32:55–63<br />

Ramos C, Molbak L, Molin S (2000) Bacterial activity in the rhizosphere analyzed at the<br />

single-cell level by monitoring ribosome contents and synthesis rates. Appl Environ<br />

Microbiol 66:801–809<br />

Ramos C, Licht TR, Sternberg C, Krogfelt KA, Molin S (2001) Monitoring bacterial<br />

growth activity in biofilms from laboratory flow-chambers, <strong>plant</strong> rhizosphere and<br />

animal intestine. Methods Enzymol 337:21–42<br />

Rheims H, Sproer C, Rainey FA, Stackebrandt E (1996) Molecular biological evidence for<br />

the occurrence of uncultured members of the actinomycete line of descent in different<br />

environments and geographical locations. Microbiology 142:2863–2870<br />

Sambrook J, Fritsch EF, Maniatis T (1989) Molecular cloning. A laboratory manual, 2nd<br />

edn. Cold Spring Harbor Laboratory Press, New York<br />

Schlesner H (1994) Development of media suitable for the microorganisms morphologically<br />

resembling Planctomycetes spp., Pirellula spp., other Planctomycetales from<br />

various aquatic habitats using dilute media. System Appl Microbiol 17:135–145<br />

Schloter M, Borlinghaus R, Bode W, Hartmann A (1993) Direct identification, and localization<br />

of Azospirillum in the rhizosphere of wheat using fluorescence-labelled monoclonal<br />

antibodies and confocal scanning laser microscopy. J Microsc 171:173–176<br />

Schloter M, Assmus B, Hartmann A (1995) The use of immunological methods to detect<br />

and identify bacteria in the environment. Biotechnol Adv 13:75–90<br />

Shirling EB, Gottlieb D (1966) Methods for characterization of Streptomyces species. Int<br />

J Syst Bacteriol 16:313–340<br />

Staley JT (1968) Prosthecomicrobium and Ancalomicrobium: new prosthecate freshwater<br />

bacteria. J Bacteriol 95:1921–1942<br />

Steidle A, Sigl K, Schuhegger R, Ihring A, Schmid M, Gantner S, Stoffels M, Riedel K,<br />

Givskov M, Hartmann A, Langebartels C, Eberl L (2001) Visualization of N-acylhomoserine<br />

lactone-mediated cell-cell communication between bacteria colonizing the<br />

tomato rhizosphere. Appl Environ Microbiol 67:5761–5770<br />

Stoffels M, Castellanos T, Hartmann A (2001) Design and application of new 16S rRNAtargeted<br />

oligonucleotide probes for the Azospirillum-Skermanella-Rhodocista-cluster.<br />

Syst Appl Microbiol 24:83–97<br />

Stotzky G, Broder MW, Doyle JD, Jones RA (1993) Selected methods for the detection and<br />

assessment of ecological effects resulting from the release of genetically engineered<br />

microorganisms to the terrestrial environment. Adv Appl Microbiol 38:1–98<br />

Stubner S, Schloter M, Moeck GS, Coulton JW, Ahne F, Hartmann A (1994) Construction<br />

of umu-fhuA operon fusions to detect genotoxic potential by an antibody-cell <strong>surface</strong><br />

reaction. Environ Tox Water Qual 9:285–291<br />

Tan Z, Hurek T, Vinuesa P, Müller P, Ladha JK, Reinhold-Hurek B (2001) Specific detection<br />

of Bradyrhizobium and Rhizobium strains colonizing rice (Oryza sativa) roots by<br />

16S-23S ribosomal DNA intergenic spacer-targeted PCR. Appl Environ Microbiol<br />

67:3655–3664<br />

Tas É, Lindström K (2001) Identification of bacteria by their intrinsic sequences: Probe<br />

design and testing of their specificity. In: Akkermans ADL,Van Elsas JD, De Bruijn FJ<br />

(eds) Molecular microbial ecology manual, Suppl. 5, Kluwer Academic Press, Dordrecht<br />

Tolker-Nielsen T, Brinch UC, Ragas PC, Andersen JB, Jacobsen CS, Molin S (2000) Development<br />

and dynamics of Pseudomonas sp. biofilms. J Bacteriol 182:6482–6489


24 Microbial Community Analysis in the Rhizosphere 469<br />

Torsvik V, Sorheim R, Goksoyr J (1996) Total bacterial diversity in soil and sediment<br />

communities: a review. J Industr Microbiol 17:170–178<br />

Tsien HC, Bratina BJ, Tsuji K, Hanson RS (1990) Use of oligodeoxynucleotide signature<br />

probes for identification of physiological groups of methylotrophic bacteria. Appl<br />

Environ Microbiol 56:2858–2865<br />

Unge A, Tombolini R, Molbak L, Jansson JK (1999) Simultaneous monitoring of cell<br />

number and metabolic activity of specific bacterial populations with a dual gfpluxAB<br />

marker system. Appl Environ Microbiol 65:813–821<br />

Wagner M, Amann R, Lemmer H, Schleifer KH (1993) Probing activated sludge with<br />

oligonucleotides specific for proteobacteria: inadequacy of culture-dependent methods<br />

for describing microbial community structure. Appl Environ Microbiol<br />

59:1520–1525<br />

Wagner R (1994) The regulation of ribosomal rRNA synthesis and bacterial cell growth.<br />

Arch Microbiol 161:100–106<br />

Weidner S, Arnold W, Pühler A (1996) Diversity of uncultured microorganisms associated<br />

with the seagrass Halophila stipulacea estimated from restriction fragment<br />

length polymorphism analysis of PCR-amplified 16S rRNA genes. Appl Environ<br />

Microbiol 62:766–771<br />

Werner D (2001) Organic signals between <strong>plant</strong>s and microorganisms. In: Pinton R,<br />

Varanini Z, Nannipieri P (eds) The rhizosphere. Marcel Dekker, New York, pp 197–222<br />

White DC, Ringelberg DB (1998) Signature lipid biomarker analysis. In: Burlage RS,Atlas<br />

R, Stahl D, Geesey G, Sayler G (eds) Techniques in microbial ecology. Oxford University<br />

Press, New York, pp 255–272<br />

Wieland G, Neumann R, Backhaus H (2001) Variation of microbial communities in soil,<br />

rhizosphere, and rhizoplane in response to crop species, soil type, and crop development.<br />

Appl Environ Microbiol 67:5849–5854<br />

Zelles L (1997) Phospholipid fatty acid profiles in selected members of soil microbial<br />

communities. Chemosphere 35:275–294


25 Methods for Analysing the Interactions Between<br />

Epiphyllic Microorganisms and Leaf Cuticles<br />

Daniel Knoll and Lukas Schreiber<br />

1 Introduction<br />

The <strong>plant</strong> cuticle forms the solid <strong>surface</strong> environment for epiphyllic microorganisms.<br />

This chapter presents newly developed techniques for analysing the<br />

interactions between epiphyllic microorganisms and leaf cuticles. The methods<br />

take into account the unique physical, chemical and functional characteristics<br />

of the cuticular interface of leaves. Furthermore, a new experimental<br />

approach simulating leaf <strong>surface</strong> microbe interactions on the basis of isolated<br />

cuticular membranes (CM) will be presented. Changes in cuticular properties<br />

in relation to microbial growth can be assessed in vitro under controlled conditions.<br />

2 Physical Characterisation of Cuticle Surfaces by Contact<br />

Angle Measurements<br />

Surface wetting can be determined quantitatively by measuring the contact<br />

angle s of an aqueous droplet applied to a <strong>surface</strong>. The contact angle s is<br />

defined by the angle (°) between the flat leaf <strong>surface</strong> and the line tangent to a<br />

water droplet through the point of contact as demonstrated in Fig. 1. The size<br />

of the contact angle s is directly related to the hydrophobic properties of a<br />

<strong>surface</strong>. Low contact angles indicate well wettable <strong>surface</strong>s (left-hand side of<br />

Fig. 1), whereas high contact angles indicate little wettable <strong>surface</strong>s (righthand<br />

side of Fig. 1). Generally, advancing contact angles are measured with<br />

the aid of a goniometer within the first minute after application of a droplet<br />

onto the <strong>surface</strong>. The droplet volume may vary from 1 to 10 ml, since it has<br />

been previously shown that contact angles were independent of the droplet<br />

size (Schreiber 1996). However, contact angles can be significantly dependent<br />

on the pH values of the buffered aqueous solutions. So-called contact angle<br />

titration measuring contact angles at different pH values ranging between pH<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


472<br />

Contactangle<br />

Daniel Knoll and Lukas Schreiber<br />

<br />

Waterdroplet<br />

Contactangle<br />

Waterdroplet<br />

Leaf <strong>surface</strong> Leaf <strong>surface</strong><br />

Fig. 1. A scheme of contact angles from aqueous droplets on <strong>surface</strong>s of different<br />

hydrophobicity. The contact angle s is related to wetting properties of <strong>surface</strong>s. Low contact<br />

angles indicate well wettable <strong>surface</strong>s (left), whereas high contact angles indicate<br />

rarely wettable <strong>surface</strong>s (right)<br />

3.0 and 11.0 can reveal important additional information about the chemical<br />

nature of interfacial molecules.<br />

Contact angles can be measured on leaf <strong>surface</strong>s and a variety of different<br />

model <strong>surface</strong>s (Knoll and Schreiber 1998, 2000). Prior to the contact angle<br />

measurement on leaf <strong>surface</strong>s, leaves have to be immersed in deionised water<br />

for 10 s and carefully blotted with filter paper. This washing step removes any<br />

deposits and dust particles weakly adsorbed to the leaf <strong>surface</strong>, which might<br />

dissolve in the aqueous drops used for the contact angle measurements. Leaf<br />

strips are cut out from the leaf avoiding central veins and necrotic lesions.<br />

Then leaf strips are attached to microscope slides that are placed in the<br />

goniometer to measure the contact angle of the applied droplet. Contact<br />

angles can be measured on leaf <strong>surface</strong>s that are naturally or artificially<br />

colonised in different degrees with microorganisms.<br />

In order to analyse the impact of cuticular waxes and of epiphytic microorganisms<br />

on wetting properties of leaf <strong>surface</strong>s, both components can be isolated<br />

and applied separately to microscope slides as artificial supports. Isolated<br />

wax is recrystallised from the melt on chloroform-washed microscope<br />

slides. For details about wax extraction, refer to the second part of this chapter.<br />

Wetting properties of different species of epiphytic microorganisms can<br />

be determined after cell adherence to artificial glass supports (Fig. 2).Washed<br />

cell suspensions are incubated with hydrophilic chloroform-washed glass<br />

slides and with highly hydrophobic slides that were obtained by chemical<br />

silanisation of the slides (Leibnitz and Struppe 1984).Washed cell suspensions<br />

(25 ml) are transferred onto sterilised microscope slides in sterile tissue culture<br />

dishes. After incubation for 24 h at 25 °C, microscope slides are carefully<br />

washed using a gentle stream of sterile deionised water and remaining<br />

amounts of water are allowed to evaporate. Contact angles are measured<br />

immediately after drying of the <strong>surface</strong>s. In order to measure contact angles<br />

as a function of cell density glass slides are incubated at 25 °C with different<br />

cell concentrations for 6 and 48 h, respectively.


25 Analysing Interactions between Microorganisms and Cuticles 473<br />

Fig. 2. Contact angles of aqueous solutions of different pH values measured on<br />

colonised glass <strong>surface</strong>s of different hydrophobicity. Untreated, polar and silanised,<br />

unpolar glass <strong>surface</strong>s were inoculated with various microbial cell suspensions for 24 h<br />

at 25 °C. As a control, glass and silanised glass <strong>surface</strong>s were incubated with PBS buffer.<br />

Values are means with 95 % confidence intervals (ci) from at least 20 contact angle measurements<br />

with 10 mM citric buffer (pH 3.0) and 10 mM borate buffer (pH 9.0)<br />

3 Chemical Characterisation of Cuticle Surfaces<br />

The chemical composition of cutin and cuticular waxes is determined via gas<br />

chromatography coupled with flame ionisation, infrared or mass spectrometric<br />

detectors. Further information on chemical wax and cutin chemistry can<br />

be obtained from a series of reviews (Kolattukudy 1996; Holloway 1982; Walton<br />

1990; Riederer and Markstädter 1996). In the following, a brief outline of<br />

the principal steps necessary for wax analysis is given. Sample preparation for


474<br />

Daniel Knoll and Lukas Schreiber<br />

chemical analysis generally includes extraction with organic solvents, concentration<br />

of the samples by solvent evaporation, derivatisation of alcoholic<br />

and carboxylic groups and analysis by gas chromatography.<br />

Cuticular waxes can be easily extracted from <strong>plant</strong> <strong>surface</strong>s using organic<br />

solvents like chloroform. Brief extractions of fresh foliage of around 10 s have<br />

been shown to be sufficient to remove all of the <strong>surface</strong> wax and most of the<br />

embedded wax (Schreiber and Schönherr 1993). After evaporation of the<br />

chloroform, the wax concentration is adjusted to 1 mg/ml and 100 ml of the<br />

extract is transferred into 1-ml reactivials for chemical analysis. In order to<br />

quantify wax components, known amounts of highly pure alkane standards<br />

(e.g., 5 mg Dotriacontane) are added to the sample. Derivatisation is necessary<br />

in order to convert free hydroxyl and carboxyl groups into their corresponding<br />

trimethylsilyl ethers and esters. This is done by treating the dried extracts<br />

with 10–30 ml of pyridine and of N,N-trimethylsilyl-trifluoroacetamide<br />

(BSTFA) at 70 °C for 30 min. Of the silylated samples, 1 µl is then injected into<br />

a gas chromatograph equipped with a flame ionisation detector. Optimised<br />

temperature and pressure programs as well as special fused silica capillary<br />

columns gain the best separation of the larger-molecular-weight aliphatic<br />

components based on their different C-carbon chain lengths.An example of a<br />

wax amount [µg cm -2 ] intensity<br />

200000 ISTD<br />

175000<br />

150000<br />

125000<br />

100000<br />

75000<br />

50000<br />

25000<br />

0<br />

C 24 AN<br />

C 31 AN<br />

10 15 20 25 30 35 40 45 50 55<br />

3<br />

2,5<br />

2<br />

1,5<br />

1<br />

0,5<br />

0<br />

alkanes<br />

alcohols<br />

aldehydes<br />

acids<br />

esters<br />

triterpenoids<br />

C22 C24 C25 C26 C27 C28 C29 C30 C31 C32 C33 C34 C38 C40 C42 C44 C46 C48 C50 Tri2 Tri3 Tri4 Tri5 Tri6 Tri7 Tri8<br />

substances<br />

time [min]<br />

Fig. 3. Gas chromatographic analysis of the leaf <strong>surface</strong> wax of strawberry (Fragaria x<br />

ananassa cv. Elsanta). A Example of an original gas chromatogram of the strawberry<br />

wax analysed on a gas chromatograph equipped with a flame ionisation detector: ISTD<br />

internal standard, C 24 AN tetracosane, C 31 AN untriacontane). B Chain lengths distribution<br />

and quantitative wax coverage of the leaf <strong>surface</strong> of strawberry<br />

A<br />

B


25 Analysing Interactions between Microorganisms and Cuticles 475<br />

gas chromatogram is shown in Fig. 3. The qualitative wax analysis is performed<br />

by gas chromatography combined with a mass spectrometric detector.<br />

Identification of wax components relies on the specific mass spectra of the<br />

molecules. The wax coverage and the wax composition is usually given per<br />

unit area of <strong>plant</strong> <strong>surface</strong>. Therefore, the total area of extracted leaves or cuticles<br />

needs to be determined after wax extraction.<br />

4 A New in Vitro System for the Study of Interactions<br />

Between Microbes and Cuticles<br />

4.1 Isolated Cuticles as Model Surfaces for Phyllosphere Studies<br />

This new experimental system for in vitro studies of leaf <strong>surface</strong>–microbe<br />

interactions is based on isolated cuticles as colonisation <strong>surface</strong>s. Isolated<br />

cuticles are ideal model <strong>surface</strong> for simulation of the phylloplane habitat as<br />

the special interfacial character of the phyllosphere is retained. Surfaces of<br />

cuticular membranes reflect the topography of epidermal cells with anticlinal<br />

cell wall depressions and the course of leaf veins like an reverse imprint of the<br />

Fig. 4. Scanning electron microscope picture of an isolated cuticular membrane of ivy<br />

(Hedera helix L.). View of the physiological inner side of the cuticle. The pattern of epidermal<br />

cell walls and leaf veins is clearly visible


476<br />

Daniel Knoll and Lukas Schreiber<br />

leaf <strong>surface</strong> (Fig. 4). Furthermore, isolated cuticles have a functionally intact<br />

wax layer that leads to an extreme high <strong>surface</strong> hydrophobicity and a reduction<br />

of solute transport across the cuticle. However, cuticular membranes are<br />

still permeable to a lesser extent for water and for anorganic as well as polar<br />

organic molecules. Thus, a boundary layer with higher humidity is formed<br />

above the cuticle <strong>surface</strong> and the naturally occurring leaching process of minerals<br />

or sugars through the cuticle is simulated. Important properties of the<br />

<strong>plant</strong> cuticle, like the cuticular permeability or the barrier function against<br />

microbial penetration, can be measured directly in relation to colonisation of<br />

the cuticle by microorganisms under strictly controlled conditions during<br />

incubation. This allows deeper insight into the mechanisms of possible interactions.<br />

In parallel, microbial population densities can be monitored by determining<br />

the colony forming units (cfu) and by microscope visualisation of the<br />

colonised cuticle <strong>surface</strong>. All methods were established with a strain of the<br />

commonly found epiphyllic leaf bacteria Pseudomonas fluorescens and can be<br />

adapted easily for the studies of other species.<br />

4.2 Enzymatic Isolation of Plant Cuticles<br />

Cuticular membranes are enzymatically isolated from astomatous leaf sides<br />

according to the method of Schönherr and Riederer (1986). Punched leaf<br />

disks with a diameter of 20 mm are vacuum-infiltrated with an enzyme solution<br />

containing 2 % (v/v) cellulase (Celluclast, Novo Nordisk, Bagsvaerd, Denmark)<br />

and 2 % (v/v) pectinase (Trenolin Super DF, Erbslöh, Geisenheim) dissolved<br />

in 10 –2 M citric buffer. 10 –3 M NaN 3 (Sigma, Deisenhofen, Germany) is<br />

added in order to inhibit microbial aerobic growth. After an incubation<br />

period of several days at room temperature, cuticles can be completely separated<br />

from adhering leaf tissue by washing carefully with deionised water.<br />

Subsequently, isolated cuticles are air-dried and stored at room temperature.<br />

For still unknown reasons the enzymatic isolation of cuticular membranes is<br />

limited to a certain number of <strong>plant</strong> species to which Prunus laurocerasus L.,<br />

Hedera helix L. and Juglans regia L. belong.<br />

4.3 The Experimental Set-Up of the System<br />

The experimental set-up of the system consists of stainless steel chambers<br />

(Fig. 5A) that were originally designed to measure cuticular permeability of<br />

volatile chemicals (Bauer 1991). Isolated cuticles are placed on the top of the<br />

chamber and fixed with a metal ring sealing the cuticle/steel interfaces with<br />

high-vacuum silicone grease (Wacker Chemie, Burghausen, Germany). Prior<br />

to assembly, chambers and rings coated with silicone grease at the chamber/ring<br />

interfaces are sterilised by dry hot air at 180 °C for 3 h. Cuticles are


25 Analysing Interactions between Microorganisms and Cuticles 477<br />

sterilised by UV radiation for 30 min on each side. Sterilised cuticles are then<br />

mounted in the chambers under sterile conditions. Care is taken that the<br />

physiological outer side of the cuticles is orientated to the outside. The physiological<br />

inner side of the cuticle faces 800 ml of a highly concentrated nutrition<br />

solution consisting of 20 % (w/v) glucose and 5 % (w/v) yeast extract or<br />

simply water. The inner volume of the chamber is accessible by sampling<br />

ports that can be closed by metal stoppers. Using a sterile plastic syringe, the<br />

solution inside the chamber can be replaced several times during the course<br />

of the experiment. Chambers were incubated upside down on a metal grid in<br />

a climate-controlled incubation box for some hours at 25 °C before the inoculation<br />

with microbial cells. Incubation boxes are 10x20 cm in size and can be<br />

closed with an air-tight lid. Boxes are sterilised with 70 % (v/v) ethanol and<br />

with UV radiation. Sterile pressurised air is conducted through the incubation<br />

box. Air humidity is set by simply changing the temperature of the water<br />

reservoir. At a temperature of 25 °C, the air has a humidity of 100 %. Lower<br />

moisture levels can be set in the incubation box by reducing the temperature<br />

of the water reservoir under 25 °C as the saturation vapour pressure of water<br />

in air is dependent on temperature (Nobel 1991). One incubation box is<br />

equipped with a hygrometer and a temperature sensor in order to verify the<br />

actual climate conditions inside the box.<br />

4.4 Inoculation of Cuticular Membranes with Epiphytic Microorganisms<br />

A cell culture of P. fluorescens is cultivated in glucose-yeast-medium overnight<br />

at 25 °C. Cells are harvested by centrifugation (2120xg, 20 min), resuspended<br />

and washed twice in 10 –2 M phosphate buffered saline (PBS, pH 7.4; Sigma<br />

Chemicals). Prior to inoculation the cell suspension is adjusted to an optical<br />

density of 1.0 that corresponds to 2.5◊10 8 cfu/ml. The outer cuticle <strong>surface</strong> is<br />

inoculated with bacteria by spreading 200 ml of a washed cell suspension of P.<br />

fluorescens evenly over the entire exposed cuticle <strong>surface</strong> (Fig. 5B). Chambers<br />

are incubated for 6 h at 25 °C in a sterile glass Petri dish containing PBSbuffer-moistened<br />

filter papers at the bottom in order to avoid evaporation of<br />

water from the inoculation solution. During the inoculation period, bacterial<br />

cells adhere to the cuticle. After 6 h the suspension is withdrawn and the <strong>surface</strong><br />

is carefully washed five times with 200 ml sterile deionised water to<br />

remove unbound bacteria. Chambers are left in a laminar flow hood until dry.<br />

Immediately after the drying of the washed cuticle <strong>surface</strong>, the chambers are<br />

transferred upside down in the incubation box (Fig. 5C). Furthermore, two<br />

control experiments are performed. One control is necessary for checking<br />

sterile conditions during the course of experiment. Therefore, cuticles are<br />

incubated with 200 ml of sterile PBS and treated in the same way as described<br />

above.Another control is to verify that during the inoculation period bacteria<br />

are not able to pass through the silicone grease from the outer cuticle <strong>surface</strong>


478<br />

A<br />

cuticle<br />

B<br />

Daniel Knoll and Lukas Schreiber<br />

sterilization experimental setup<br />

UV-light<br />

stainless steel<br />

chamber<br />

inoculation washing<br />

C<br />

bacteria<br />

pressurised air<br />

water<br />

reservoir<br />

stainless steel<br />

chamber<br />

metal ring<br />

sampling port<br />

stopper<br />

measurement<br />

lid<br />

filter incubation box metal grid<br />

nutrient<br />

solution<br />

or water<br />

50 µl<br />

petri dish<br />

Fig. 5. Scheme of the experimental set-up for the in vitro study of microorganisms–leaf<br />

cuticle interactions. A Enzymatic isolated cuticular membranes are sterilised by UV<br />

radiation and mounted in a stainless steel chamber. The chamber is filled with nutrient<br />

solution or water. B The physiological outer side of the cuticle is inoculated with a microbial<br />

cell suspension for 6 h at 25 °C. Microbial cells not bound to the cuticle <strong>surface</strong> are<br />

removed by washing the cuticle with deionised water. Samples of the solution inside the<br />

chamber can be taken with a sterile syringe via closable sampling ports. C Inoculated<br />

cuticles are incubated up-side down on a metal grid in sterile incubation boxes at 25 °C.<br />

Pressurised air of the desired moisture level is conducted through the incubation box


into the nutrition solution inside the chamber volume. Therefore, round glass<br />

cover slips that definitely cannot be breached by bacteria are mounted in<br />

place of cuticles in the chambers and inoculated with 200 ml of the cell solution.<br />

4.5 Measurement of Changes in Cuticular Transport Properties<br />

4.5.1 Determination of Cuticular Water Permeability<br />

Cuticular water permeability is measured according to a gravimetric method<br />

of Schönherr und Lendzian (1981). The permeability coefficients P (m/s) for<br />

water are calculated using the equation:<br />

P= F<br />

A¥DC 25 Analysing Interactions between Microorganisms and Cuticles 479<br />

where F is the water flow across the cuticular membrane (g/s), A is the area of<br />

the exposed cuticle <strong>surface</strong> (m 2 ) and DC represents the difference in the water<br />

concentration between the aqueous phase inside the chamber and the outer<br />

atmosphere of the incubation box. The water flow across the cuticular membrane<br />

can be measured by weighing the chambers at periodic intervals on an<br />

electronic balance with an accuracy of ±0.1 mg. The weight loss from the<br />

chambers is plotted against the incubation time and the water flow is calculated<br />

by linear regression analysis (Fig. 6). The sampling ports of the cham-<br />

Fig. 6. Effect of<br />

Corynebacterium fascians<br />

on the cuticular water permeability<br />

of Prunus laurocerasus.<br />

The flow of water<br />

through the cuticular<br />

membrane was increased<br />

by a factor of 2 after treatment<br />

with bacteria,<br />

whereas treatment with<br />

PBS did not significantly<br />

change the cuticular water<br />

flow


480<br />

Daniel Knoll and Lukas Schreiber<br />

bers are additionally sealed with adhesive tape to avoid diffusion of water<br />

through the sampling ports. Chambers are incubated upside down on dried<br />

silica gel in an air-tight polyethylene box at 25 °C. The silica gel adsorbs all<br />

free water of the air resulting in a water concentration inside the polyethylene<br />

box constantly held at 0 %. Thus, the driving force DC for the water flow across<br />

the cuticle corresponds to the density of water (10 3 kg m –3 ). The salt and sugar<br />

concentration of the nutrition solution can be neglected as it does not affect<br />

significantly the water activity a w. Control experiments showed that there was<br />

no significant change in cuticular water permeability when using deionised<br />

water or nutrition solution as the aqueous solution inside the chamber volume.<br />

Sterilisation of cuticles by UV radiation also did not significantly change<br />

water permeability.<br />

4.5.2 Effect of Bacteria on Cuticular Water Permeability<br />

Isolated cuticles are mounted in stainless steel chambers and permeability<br />

coefficients P1 for water are determined for each sample as described above.<br />

Cuticular permeability coefficients P1 determined after UV radiation ranged<br />

between 1.44◊10 –10 m/s for Vinca major leaf cuticles and 10.8x10 –9 m/s for<br />

Lycopersicon esculentum fruit cuticles (Table 1). Then cuticles are inoculated<br />

with bacteria and incubated for 12 days in the incubation box at 25 °C at air<br />

humidity close to 100 %. Control experiments are conducted by inoculating<br />

the cuticles with 200 ml PBS in place of the bacterial cell solution. After incubation<br />

with bacteria chambers are again transferred onto dried silica gel and<br />

cuticular water permeability coefficients P2 are determined after an equilibrium<br />

period of 1 day. The effects of bacteria on water permeability of the<br />

respective cuticular membrane are calculated from the permeance of the cuticle<br />

after treatment with bacteria (P2) divided by the initial permeance (P1).<br />

Effect = P2<br />

P1<br />

Table 1. Cuticular permeability coefficients for water P water (m/s) from different <strong>plant</strong><br />

species. Values are arithmetic means with 95 % confidence intervals (ci) of 14 measured<br />

permeability coefficients for each <strong>plant</strong> species<br />

Species P water ¥10 –10 (m/s) ci 95 %x10 –10 (m/s)<br />

Vinca major 1.44 1.26–1.64<br />

Hedera helix can. 2.16 1.76–2.65<br />

Prunus laurocerasus 2.93 2.34–3.68<br />

Citrus aurantium 4.53 2.97–6.92<br />

Lycopersicon esculentum 10.80 8.85–13.17


An example for the change in water permeability of one cuticular membrane<br />

before and after treatment with bacteria is shown in Fig. 6. The effects<br />

on water permeability for an entire sample unit consisting of at least 12 cuticles<br />

are given as mean values of the effects measured for individual membranes.<br />

Some results are presented in the chapter “Interactions between Epiphyllic<br />

Microorganisms and Leaf Cuticles” by Schreiber et al. (Chap. 9, this<br />

Vol.).<br />

The effects on water permeability need not necessarily be measured before<br />

and after treatment with bacteria, but can also be measured during the incubation<br />

with bacteria by lowering the air humidity inside the incubation box<br />

to, e.g. 90 %. As the driving force for the water flow across the cuticle is<br />

reduced to 1/10, periodical intervals in between weighing the chamber are<br />

increased to 4 days in order to measure a significant weight loss.<br />

4.6 Measuring Penetration of Microorganisms Through Cuticular<br />

Membranes<br />

Penetration of microorganisms through cuticular membranes can be measured<br />

as well using the described in vitro system. The outer side of the cuticle<br />

is inoculated with bacterial cells, whereas the inner side faces a sterile nutrition<br />

solution. If the cuticle, located between microbial cells and nutrition<br />

solution inside the chamber, is penetrated by bacterial cells, microbial growth<br />

will be detectable in the nutrition solution. Thus, penetration of isolated cuticles<br />

by bacteria can be easily monitored by transferring 50 ml of the nutrition<br />

solution inside the chamber onto glucose-supplemented yeast extract agar<br />

plates using a sterile syringe. Subsequent microbial growth on the agar plates<br />

indicates that a penetration event through the cuticular membrane has<br />

occurred. In that way, the amount of cuticles penetrated is determined in daily<br />

intervals. The amount of cuticles penetrated after different periods of incubation<br />

is given in percent of the total amount of inoculated membranes.<br />

%CM =<br />

penetrated<br />

25 Analysing Interactions between Microorganisms and Cuticles 481<br />

Number of CMpenetrated<br />

Number of CMtotal<br />

¥100<br />

An example for a penetration kinetic is shown in Fig. 7. The amount of penetrated<br />

cuticular membranes increased over the incubation period of 12 days.<br />

Some typical characteristics of a penetration kinetic can be used to describe<br />

the barrier function of cuticles quantitatively: (1) at the end of the incubation<br />

period there was a steady increase of penetrated cuticular membranes versus<br />

incubation time. Rates of penetration (% CM penetrated/day) can be calculated<br />

from the slopes of the linear regression. (2) Another meaningful parameter is<br />

the time needed by the microorganisms to penetrate 50 % of inoculated membranes<br />

(T 50 %). High rates of penetration and small T 50 % values indicate low


482<br />

Daniel Knoll and Lukas Schreiber<br />

Fig. 7. Penetration of<br />

Pseudomonas fluorescens<br />

through cuticular membranes<br />

of Vinca major.The<br />

amount of penetrated cuticles<br />

increases with incubation<br />

time. After 9.5 days<br />

50 % of the inoculated cuticles<br />

are penetrated by P.<br />

fluorescens. At the end of<br />

the kinetic there is a linear<br />

increase of penetrated cuticles<br />

with a rate of 6.1 %<br />

penetrated CM per day in<br />

relation to the total amount<br />

of inoculated cuticles<br />

barrier functions of the cuticle for microbial penetration. Once these values<br />

are known, barrier properties of cuticles of different <strong>plant</strong> species that differ<br />

in their morphology like cuticle thickness or chemistry like wax composition<br />

can be compared. Another attractive application is to measure penetration of<br />

different microbial strains or mutants that differ in their array of extracellular<br />

enzymes like cutinase activity.<br />

Several control experiments need to be conducted to ensure bacterial penetration<br />

through isolated cuticles. (1) When glass slides are mounted into the<br />

chambers in place of cuticles there was never any bacterial growth detectable<br />

in the nutrition solution. This gives evidence that bacteria are not able to<br />

bypass the glass <strong>surface</strong> via the silicone grease seal. (2) In addition, no bacterial<br />

growth was detected in the nutrition solution when cuticles were inoculated<br />

with sterile PBS indicating that the system itself is sterile and no other<br />

origins for bacterial growth are possible except from the inoculus on the outer<br />

cuticle <strong>surface</strong>. (3) Finally, a third control consists of applying 200 ml of dead<br />

bacteria. Cells are cultivated as described above and subsequently killed with<br />

paraformaldehyde and stained with the fluorescent dye DAPI. It was checked<br />

that all bacterial cells were killed.After the inoculation period of 6 h the nutrition<br />

solution is checked for the presence of DAPI-stained cells with fluorescence<br />

microscopy. A fraction of about 10 % of the inoculated cuticles was<br />

apparently leaky for dead cells. This might be due to mechanical injuries to<br />

the cuticular membranes during the process of isolation or during the mounting<br />

of cuticular membranes in the chambers. Those membranes were sorted<br />

out and not considered any further. Furthermore, cuticular water permeability<br />

measured prior to inoculation with bacterial cells was very low (Table 1),<br />

indicating that the membranes form high effective barriers for the transport<br />

of water on the molecular level. This also suggests that they build intact barriers<br />

for microbial cells as well. Basically, all control experiments confirmed


25 Analysing Interactions between Microorganisms and Cuticles 483<br />

that after the inoculation period of 6 h bacterial cells are solely present on the<br />

inoculated outer cuticle <strong>surface</strong>.<br />

4.7 Determination of the Viable Cell Number on the Cuticle Surface<br />

In order to document the microbial development on isolated cuticular membranes,<br />

the cfu is determined. The initial cfu on isolated cuticles is determined<br />

directly after inoculation of membranes with microorganisms. As an example,<br />

the initial cfu of P. fluorescens attached to cuticles of V. major was<br />

2.85x10 5 ±0.98x10 5 cfu/CM. Then cfu measurements are done in daily intervals<br />

during the incubation period. First, the nutrition solution inside the<br />

chamber is totally removed with a sterile syringe and kept in sterile glass<br />

tubes to check for microbial growth (see below). After having removed the<br />

nutrition solution, the membrane is cut out of the chamber with a sterile<br />

scalpel blade and transferred in a 1.5-ml tube containing 0.05 g of sterile sand.<br />

The cuticle is ground in 100 ml PBS with a micropestle for 2 min. After<br />

homogenisation of the cuticle, 900 ml PBS is added and the tube contents<br />

mixed. Serial dilutions of 100 µl are incubated on glucose yeast extract agar<br />

plates at 25 °C for 2 days before colonies have been counted.<br />

In order to determine the microbial cfu exclusively on the outer cuticle <strong>surface</strong>,<br />

it is very important to check the nutrition solution inside the chamber<br />

for microbial growth. Therefore, the nutrition solution removed from the<br />

inner chamber volume is simply incubated at 25 °C for 2 days. Only if there is<br />

no microbial growth detectable, is the cfu determined for that cuticle considered<br />

to describe the microbial development on the outer cuticle <strong>surface</strong>.<br />

4.8 Microscope Visualisation of Microorganisms on the Cuticle<br />

Microscopic detection of microbial cells on isolated cuticles gives information<br />

about the colonisation pattern and development. The fluorescent dyes acridine<br />

orange and DAPI are used to stain bacteria. Both dyes are polar substances<br />

with a very high affinity to bind nucleic acids. Thus, microbial cells<br />

adhering to the cuticle <strong>surface</strong> are specifically stained, whereas the hydrophobic<br />

cuticle <strong>surface</strong> itself is not stained. 0.02 % (w/v) acridine orange and<br />

0.001 % (w/v) DAPI are dissolved in deionised water and filtered through<br />

0.2 mm membrane filters to remove dye crystals and dust particles. Care is<br />

taken that staining solutions are protected from daylight. For better handling<br />

cuticles are left mounted in the chambers for staining of bacterial cells. Staining<br />

solution (200 µl) is evenly distributed over the outer cuticle <strong>surface</strong>.<br />

Chambers are incubated in the dark at room temperature on a horizontal<br />

shaker (30 rpm). After different staining times of 5, 20, 40 and 60 min, respectively,<br />

the cuticle <strong>surface</strong>s are washed twice with 200 ml of sterile-filtered


484<br />

Daniel Knoll and Lukas Schreiber<br />

deionised water to remove unbound dye molecules. Cuticles are left over silica<br />

gel until dry. The dried cuticle <strong>surface</strong>s are excised from the chambers<br />

with a scalpel blade and cut into four parts. Cuticle pieces are transferred onto<br />

a thin hydrophobic layer of silicon grease on a microscopic slide. A cover slip<br />

together with one drop of immersion oil is put on the top of the cuticle prior<br />

to microscopic examination. Due to the hydrophobic layer of silicon grease<br />

and the immersion oil, the entire <strong>surface</strong>s of the cuticle pieces are spread<br />

totally flat minimising problems with depth focus. Furthermore, the refraction<br />

of light is markedly reduced allowing fluorescence microscopy with isolated<br />

cuticles. Samples can be viewed with a Zeiss Axioplan microscope<br />

(Zeiss, Oberkochen, Germany) equipped with a 50 W mercury high pressure<br />

bulb, a 40x objective (Zeiss, Plan-Neofluar) and a Zeiss filter set No. 09 (excitation:<br />

450–490 nm; dichroic beamsplitter ≥510 nm; emission ≥520 nm). One<br />

examples of a fluorescence microscopy micrograph of a colonised cuticle <strong>surface</strong><br />

is shown in Fig. 8. The <strong>surface</strong> coverage of the cuticle colonised by bacte-<br />

Fig. 8. Epifluorescent microscope image of an isolated cuticular membrane of Prunus<br />

laurocerasus artificially colonised with Pseudomonas fluorescens (magnification ¥400).<br />

Bacterial cells are stained with acridine orange and viewed at an excitation of<br />

450–490 nm.Approximately 27.4 % of the cuticle <strong>surface</strong> is covered by bacteria. Bacterial<br />

cells are accumulated in small clusters over the entire cuticle <strong>surface</strong>


25 Analysing Interactions between Microorganisms and Cuticles 485<br />

rial cells can be quantified by digital image analysis. Digitised video images<br />

are analysed for the pixel size of stained bacterial cells using Adobe Photoshop<br />

software. Percentage coverage of bacterial cells is calculated as follows:<br />

No. of pixel of bacterial cells of digitized image<br />

% coverage = ¥100<br />

No. of total pixel size of digitized image<br />

Percentage coverage by bacteria is given as the mean value of 12 analysed<br />

digitised images at 400x magnification from randomly chosen sites of at least<br />

three cuticles per sampling point. The influence of staining time with acridine<br />

orange on the area coverage can be seen in Fig. 9A. The optimal staining time<br />

is 20 m. An adhesion kinetic of cells of P. fluorescensto cuticle <strong>surface</strong>s of P.<br />

Fig. 9. Surface coverage of cuticles from Prunus laurocerasus with Pseudomonas fluorescens.<br />

A Dependence of the <strong>surface</strong> coverage by bacterial cells on the staining time<br />

with acridine orange. The optimal staining time was 20 min. B Adhesion of bacterial cells<br />

to the cuticle <strong>surface</strong> over time. Maximal adhesion of 46.9 % occurred after 6 h of inoculation.<br />

Percentage coverage by bacterial cells is given as the mean value with 95 % confidence<br />

intervals of 12 analysed digitised images at x400 magnification from randomly<br />

chosen sites of each of three examined cuticles. Only two membranes could be analysed<br />

for the 60-min time sample


486<br />

Daniel Knoll and Lukas Schreiber<br />

laurocerasus is shown in Fig. 9B. Maximal <strong>surface</strong> coverage of 46.9 % was<br />

reached after 6 h of inoculation with bacterial cell solution.<br />

5 Conclusions<br />

The presented methods allow a detailed analysis of a variety of microbe–cuticle<br />

interactions combining physicochemical, ecophysiological and microbial<br />

ecological aspects. Isolated cuticles are excellent model <strong>surface</strong>s to study the<br />

mechanisms of such interactions. Using the presented in vitro system, even<br />

minor changes in cuticular wax composition or permeability can be examined<br />

in relation to microbial growth. When working with entire leaves, such<br />

changes would probably be masked by the physiological influence of the leaf.<br />

Therefore, this new approach might be very helpful to reveal possible mechanisms<br />

of interactions that occur in reality only in the scale of microhabitats.<br />

The impact of cuticular features will help us to understand the observed heterogeneous<br />

colonisation of the leaf habitat and the formation of microcolonies.<br />

Vice versa, the capacity of microbial cells to change cuticular properties<br />

might be of crucial importance for a successful colonisation of the leaf<br />

<strong>surface</strong>s and could contribute substantially to the microbial fitness of individual<br />

epiphyllic species.<br />

Acknowledgements. The authors gratefully acknowledge financial support of this work<br />

by the Deutsche Forschungsgemeinschaft and the FCI.<br />

References and Selected Reading<br />

Bauer H (1991) Mobilität organischer Moleküle in der pflanzlichen Kutikula. PhD Thesis,<br />

Technical University of Munich, Germany<br />

Holloway PJ (1982) The chemical constitution of <strong>plant</strong> cutins. In: Cutler DF, Alvin KL,<br />

Price CE (eds) The <strong>plant</strong> cuticle. Academic Press, London<br />

Knoll D (1998) Die Bedeutung der Kutikula bei der Interaktion zwischen epiphyllen<br />

Mikroorganismen und Blattoberflächen. PhD Thesis, University of Würzburg, Germany<br />

Knoll D, Schreiber L (1998) Influence of epiphytic micro-organisms on leaf wettability:<br />

wetting of the upper leaf <strong>surface</strong> of Juglans regia and of model <strong>surface</strong>s in relation to<br />

colonization by microorganisms. New Phytol 140:271–282<br />

Knoll D, Schreiber L (2000) Plant-microbe interactions: wetting of ivy (Hedera helix L.)<br />

leaf <strong>surface</strong>s in relation to colonization by epiphytic microorganisms. Microb Ecol<br />

41:33–42<br />

Kolattukudy PE (1996) Biosynthetic pathways of cutin and waxes. In: Kerstiens G (ed)<br />

Plant cuticles: an integrated functional approach. BIOS Scientific Publishers, Oxford,<br />

pp 83–108<br />

Leibnitz E, Struppe HG (1984) Handbuch der Gaschromatographie. Akademische Verlagsgesellschaft,<br />

Leipzig


25 Analysing Interactions between Microorganisms and Cuticles 487<br />

Nobel PS (1991) Physicochemical and environmental <strong>plant</strong> physiology. Academic Press,<br />

San Diego<br />

Riederer M, Markstädter C (1996) Cuticular waxes: a critical assessment of current<br />

knowledge. In: Kerstiens G (ed) Plant cuticles: an integrated functional approach.<br />

BIOS Scientific Publishers, Oxford, pp 189–200<br />

Schönherr J, Lendzian K (1981) A simple and inexpensive method of measuring water<br />

permeability of isolated <strong>plant</strong> cuticular membranes. Z Pflanzenphysiol 102:321–327<br />

Schönherr J, Riederer M (1986) Plant cuticles sorb lipophilic compounds during enzymatic<br />

isolation. Plant Cell Environ 4:459–466<br />

Schreiber L (1996) Wetting of the upper needle <strong>surface</strong> of Abies grandis: influence of pH,<br />

wax chemistry and epiphyllic microflora on contact angles. Plant Cell Environ<br />

19:455–463<br />

Schreiber L, Schönherr J (1993) Mobilities of organic compounds in reconstituted cuticular<br />

wax of barley leaves: Determination of diffusion coefficients. Pestic Sci 38:353–<br />

361<br />

Walton TJ (1990) Waxes, cutin and suberin. Meth Plant Biochem 4:105–158


26 Quantifying the Impact of ACC Deaminase-<br />

Containing Bacteria on Plants<br />

Donna M. Penrose and Bernard R. Glick<br />

1 Introduction<br />

In 1994, we reported that the bacterium, Pseudomonas putida GR12–2 (Lifshitz<br />

et al. 1986), a well-known <strong>plant</strong> growth promoting strain, contained the<br />

enzyme, 1-aminocyclopropane-1-carboxylic acid (ACC) deaminase (Jacobson<br />

et al. 1994). This enzyme hydrolyzes ACC, the immediate precursor of ethylene,<br />

in <strong>plant</strong> tissues (Yang and Hoffman 1984). Ethylene is required for seed<br />

germination by many <strong>plant</strong> species and the rate of ethylene production<br />

increases during germination and seedling growth (Abeles et al. 1992).<br />

Although low levels of ethylene appear to enhance root initiation and growth,<br />

and promote root extension, high levels of ethylene produced by fast growing<br />

roots can lead to inhibition of root elongation (Mattoo and Suttle 1991; Ma et<br />

al. 1998). We have proposed a model that suggests that ACC deaminase-containing<br />

<strong>plant</strong> growth promoting bacteria can lower ethylene levels and thus<br />

stimulate <strong>plant</strong> growth (Glick et al. 1998). It is quite likely that much of the<br />

ACC produced during ethylene biosynthesis is taken up by the bacterium and<br />

subsequently hydrolyzed to a–ketobutyrate and ammonia by ACC deaminase.<br />

The uptake and cleavage of ACC by ACC deaminase would decrease the<br />

amount of ACC, as well as ethylene.<br />

2 Selection of Bacterial Strains that Contain ACC Deaminase<br />

We developed a rapid and novel procedure for the isolation of ACC deaminase-containing<br />

bacteria and used this technique to identify and isolate seven<br />

<strong>plant</strong> growth promoting strains based on their ability to utilize ACC as the<br />

sole source of nitrogen (Glick et al. 1995). These bacterial strains were isolated<br />

from soil samples collected during late summer in Waterloo, Ontario, Canada<br />

and various locations in California, USA from the rhizosphere of seven different<br />

<strong>plant</strong>s (Table 1). Originally, these strains were designated as Pseudomonas<br />

sp., but were re-classified following fatty acid analysis (Shah et al. 1997).<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


490<br />

Donna M. Penrose and Bernard R. Glick<br />

Table 1. ACC-utilizing bacterial strains isolated from Waterloo, Ontario, Canada and<br />

California, USA<br />

Genus and species Strain Soil location Plant source<br />

Pseudomonas putida UW1 Waterloo, Ontario, Canada Bean<br />

Enterobacter cloacae UW2 Waterloo, Ontario, Canada Clover<br />

Pseudomonas putida UW3 Waterloo, Ontario, Canada Maize<br />

Enterobacter cloacae UW4 Waterloo, Ontario, Canada Reeds<br />

Pseudomonas fluorescens CAL1 San Benito, California, USA Oats<br />

Enterobacter cloacae CAL2 King City, California, USA Tomato<br />

Enterobacter cloacae CAL3 Fresco, California, USA Cotton<br />

Our method of isolating bacteria entails screening soil bacteria for the ability<br />

to use ACC as a sole nitrogen source, a trait that is a consequence of the<br />

presence of the activity of the enzyme, ACC deaminase. One gram of soil is<br />

added to 50 ml of sterile medium containing 10 g proteose peptone, 10 g<br />

casein hydrolysate, 1.5 g anhydrous MgSO 4 , 1.5 g K 2 HPO 4 and 10 ml glycerol<br />

(PAF medium) in a 250-ml flask. The flask and its contents are incubated in a<br />

shaking water bath (200 rpm) at either 25 or 30 °C depending on the geographic<br />

location of the soil samples, i.e., the samples collected in the cooler<br />

Canadian climate of Waterloo, Ontario were grown at 25 °C and those from<br />

the warmer weather of California, USA were grown at 30 °C.After 24-h, a 1-ml<br />

aliquot is removed from the growing culture, transferred to 50 ml of sterile<br />

PAF medium in a 250-ml flask and incubated at 200 rpm in a shaking water<br />

bath for 24 h, at either 25 or 30 °C, the same temperature as the first incubation.<br />

Following these two incubations, the population of pseudomonads is<br />

enriched and the number of fungi in the culture is reduced.<br />

A 1-ml aliquot is removed from the second culture and transferred to a<br />

250-ml flask containing 50 ml of sterile minimal medium, DF salts (Dworkin<br />

and Foster 1958; per litre): 4.0 g KH 2 PO 4 , 6.0 g Na 2 HPO 4 , 0.2 g MgSO 4 ·7H 2 O,<br />

2.0 g glucose, 2.0 g gluconic acid and 2.0 g citric acid with trace elements: 1 mg<br />

FeSO 4·7H 2O, 10 mg H 3BO 3, 11.19 mg MnSO 4·H 2O, 124.6 mg ZnSO 4·7H 2O,<br />

78.22 mg CuSO 4·5H 2O, 10 mg MoO 3, pH 7.2 and 2.0 g (NH 4) 2SO 4 as a nitrogen<br />

source. In our lab the DF minimal medium is prepared as follows: (1) the trace<br />

elements (10 mg H 3BO 3, 11.19 mg MnSO 4◊H 2O, 124.6 mg ZnSO 4◊7H 2O,<br />

78.22 mg CuSO 4◊5H 2O, and 10 mg MoO 3) are dissolved in 100 ml of sterile distilled<br />

water and then stored in the refrigerator for up to several months; (2)<br />

FeSO 4 ◊7H 2 O (1 mg) is dissolved in 10 ml of sterile distilled water and is stored<br />

in the refrigerator for up to several months; (3) all of the other ingredients<br />

including 4.0 g KH 2PO 4, 6.0 g Na 2HPO 4, 0.2 g MgSO 4·7H 2O, 2.0 g glucose, 2.0 g<br />

gluconic acid, 2.0 g citric acid, 2.0 g (NH 4) 2SO 4 and 0.1 ml of each of the solutions<br />

of trace elements and FeSO 4◊7H 2O are dissolved in 1 l of distilled water


26 Quantifying the Impact of ACC Deaminase-Containing Bacteria on Plants 491<br />

and autoclaved for no more than 20 min. If this medium is prepared by dissolving<br />

one ingredient at a time, i.e., by not adding another ingredient until<br />

the previous one is completely dissolved, this medium should not contain a<br />

precipitate. Following an incubation of 24 h in a shaking water bath at<br />

200 rpm at either 25 or 30 °C, the same temperature as the first incubation, a<br />

1-ml aliquot is removed from this culture and transferred to 50 ml of sterile<br />

DF salts minimal medium in a 250-ml flask containing 3.0 mM ACC (instead<br />

of (NH 4) 2SO 4) as the source of nitrogen. A 0.5 M-solution of ACC (Calbiochem-Novobiochem<br />

Corp., La Jolla, CA, USA), which is very labile in solution,<br />

is filter-sterilized through a 0.2 mm membrane and the filtrate collected,<br />

aliquoted and frozen at –20 °C. Just prior to inoculation, the ACC solution is<br />

thawed and a 300-ml aliquot added to 50 ml of sterile DF salts minimal<br />

medium; following inoculation, the culture is placed in a shaking water bath<br />

at 200 rpm and grown for 24 h at either 25 or 30 °C, the same temperature as<br />

the previous incubation.<br />

Dilutions of this final culture are plated onto solid DF salts minimal<br />

medium and incubated for 48 h at either 25 or 30 °C, the same temperature as<br />

the previous incubations. These plates are prepared with 1.8 % Bacto-Agar<br />

(Difco Laboratories, Detroit, MI, USA), which has a very low nitrogen content,<br />

and are spread with ACC (30 mmol/plate) just prior to use. Before streaking<br />

with either a loopful of bacterium or an individual colony, the ACC is allowed<br />

to dry fully. The inoculated plates are incubated at the appropriate temperature<br />

– no higher than 35 °C because all of the known ACC deaminases are<br />

inhibited above this temperature – for 3 days and the growth on the plates is<br />

checked daily. Even when apparently nitrogen-free agar is used, and no additional<br />

source of nitrogen is included in the medium, it is almost impossible to<br />

obtain plates with absolutely no growth, but it is possible to obtain plates with<br />

very, very light growth.<br />

The colonies isolated from each of the seven soil samples displayed a similar<br />

colony morphology and rate of growth. In order to avoid isolating multiple<br />

copies of the same bacterium, only a single colony from each soil sample<br />

is selected for further testing. Each selected colony is tested for the synthesis<br />

of siderophores, antibiotics and indole acetic acid, as well as for <strong>plant</strong> growth<br />

stimulation and ACC deaminase activity. It is interesting to note that Belimov<br />

et al. (submitted for publication) used a variant of the procedure described<br />

above to isolate ACC deaminase-containing strains of Bacillus.<br />

3 Culture Conditions for the Induction of Bacterial ACC<br />

Deaminase Activity<br />

The assessment of bacterial ACC deaminase activity and root growth<br />

enhancement both require growth conditions that favor the induction of ACC<br />

deaminase. The bacteria are cultured first in rich medium and then trans-


492<br />

Donna M. Penrose and Bernard R. Glick<br />

ferred to minimal medium with ACC as the sole source of nitrogen. Bacterial<br />

cells are grown to mid- up to late-log phase in 15 ml of rich medium, e.g.,<br />

tryptic soybean broth (TSB; Difco Laboratories, Detroit, MI, USA) divided<br />

between two culture tubes: each tube is inoculated with 5 ml of the appropriate<br />

strain. Cultures are incubated overnight in a shaking water bath at 200 rpm<br />

at either 25 or 30 °C – the temperature most suitable for the bacterial strain.<br />

The accumulated biomass is harvested by centrifugation of the contents of the<br />

combined tubes at 8000xg for 10 min at 4 °C in a Sorvall RC5B/C centrifuge<br />

using an SS34 rotor. The supernatant is removed and the cells are washed with<br />

5 ml of DF salts minimal medium. Following an additional centrifugation for<br />

10 min at 8000xg in the same rotor at 4 °C, the cells are suspended in 7.5 ml of<br />

DF salts minimal medium, in a fresh culture tube. Just prior to incubation, the<br />

frozen 0.5 M ACC solution (prepared as described in Sect. 2) is thawed, and an<br />

aliquot of 45 ml is added to the cell suspension; the final ACC concentration is<br />

3.0 mM. The bacterial cells are returned to the shaking water bath to induce<br />

the activity of ACC deaminase – at 200 rpm for 24 h at the same temperature<br />

as the overnight incubation, either 25 or 30 °C. The bacteria are harvested by<br />

centrifugation at 8000xg for 10 min at 4 °C in an SS34 rotor in a Sorvall<br />

RC5B/C centrifuge. The supernatant is removed, and the cells are washed by<br />

suspending the cell pellet in 5 ml of either 0.1 M Tris-HCl, pH 7.6 if the cells<br />

are to be assayed for ACC deaminase activity, or 0.03 M MgSO 4 if they are to<br />

be used as a bacterial treatment in the gnotobiotic root elongation assay or<br />

the high performance liquid chromatography (HPLC) protocol for measuring<br />

ACC. Following centrifugation at 8000xg at 4 °C for 10 min in the same rotor<br />

and centrifuge, the supernatant is discarded. The washing procedure is<br />

repeated twice to ensure that the pellet is free of medium. The pelleted cells<br />

are stored at either –20 °C for measurement of ACC deaminase activity, or at<br />

4 °C for seed treatment in the gnotobiotic root elongation assay or HPLC measurement<br />

of ACC.<br />

4 Gnotobiotic Root Elongation Assay<br />

The gnotobiotic root elongation assay is used as a method of assessing the<br />

effect of various bacterial strains on the growth of canola seedlings. Each of<br />

the seven strains of ACC deaminase-containing soil bacteria isolated in our<br />

lab was assayed by the root elongation assay and was shown to promote<br />

canola seedling growth under gnotobiotic conditions. The protocol described<br />

below is a modification of the procedure developed by Lifshitz et al. (1987)<br />

and is used to measure the elongation of canola roots from seeds treated with<br />

different strains of bacteria or chemical ethylene inhibitors. The bacterial cell<br />

pellet, prepared as described in Section 3, is suspended in 0.5 ml of sterile<br />

0.03 M MgSO 4 and then placed on ice. A 0.5-ml sample is removed from the<br />

cell suspension and diluted eight to ten times in 0.03 M MgSO 4; the


26 Quantifying the Impact of ACC Deaminase-Containing Bacteria on Plants 493<br />

absorbance of the sample is measured at 600 nm. This measurement is used to<br />

adjust the absorbance at 600 nm, of the bacterial suspension, to 0.15 with sterile<br />

0.03 M MgSO 4.<br />

Seed-pack growth pouches (Northrup King Co., Minneapolis, MN, USA)<br />

are prepared for the gnotobiotic assay of canola root elongation. Following<br />

the addition of 12 ml of distilled water to each one, the growth pouches are<br />

wrapped in aluminum foil in groups of ten, placed in an upright position to<br />

prevent water loss, and autoclaved at 121 °C for 15 min.<br />

Canola seeds (Brassica campestris) are disinfected immediately before use.<br />

(Tomato seeds may also be used in this assay.) The seeds (approximately<br />

0.2 g/treatment) are soaked in 70 % ethanol for 1 min in glass Petri dishes<br />

(60¥15 mm); the ethanol is removed and replaced with 1 % sodium hypochlorite<br />

(household bleach). After 10 min the bleach solution is suctioned off and<br />

the seeds are thoroughly rinsed with sterile distilled water at least five times,<br />

sterile distilled water is added to the dish of seeds, swirled and removed by<br />

suction. Each dish is incubated at room temperature for 1 h with the appropriate<br />

treatment: sterile 0.03 M MgSO 4 (used as a negative control) or bacterial<br />

suspensions in sterile 0.03 M MgSO 4. Following incubation with each treatment,<br />

the seeds are placed in growth pouches with sterilized forceps: six seeds<br />

are set in each growth pouch and ten pouches are used for each treatment.<br />

The pouches are grouped together according to treatment and placed upright<br />

in a rack (Northrup King Co., Minneapolis, MN, USA) ensuring that the<br />

pouches are not touching. Two empty pouches are placed at the ends of each<br />

rack. Racks are placed in a clean plastic bin containing sterile distilled water,<br />

to a depth of approximately 3 cm, and covered loosely with clear plastic wrap<br />

to prevent dehydration. Pouches are incubated in a growth chamber (Conviron<br />

CMP 3244, Controlled Environments Ltd., Winnipeg, MB, Canada) which<br />

is maintained at 20±1 °C with a cycle beginning with 12 h of dark followed by<br />

12 h of light (18 mmol m –1 s –1 ). Each rack is positioned such that the center of<br />

the row of pouches is 8in. below and 5 in. lateral to the light source. The primary<br />

root lengths are measured on the fifth day of growth and the data are<br />

analyzed. Seeds that fail to germinate 2 days after they were sown are marked<br />

and the roots that subsequently develop from these seeds are not measured.<br />

5 Measurement of ACC Deaminase Activity<br />

ACC deaminase activity is assayed according to the method of Honma and<br />

Shimomura (1978) which measures the amount of a-ketobutyrate when the<br />

enzyme, ACC deaminase, cleaves ACC. The number of mmoles of a-ketobutyrate<br />

produced by this reaction is determined by comparing the absorbance<br />

at 540 nm of a sample to a standard curve of a-ketobutyrate ranging between<br />

0.1 and 1.0 mmol (Fig. 1). A stock solution of 100 mM a-ketobutyrate (Sigma-<br />

Aldrich Co.) is prepared in 0.1 M Tris-HCl pH 8.5 and stored at 4 °C. Just prior


494<br />

Absorbance at 540 nm<br />

1.6<br />

1.4<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

Donna M. Penrose and Bernard R. Glick<br />

0<br />

0 0.2 0.4 0.6 0.8 1<br />

-ketobutyrate, moles<br />

to use, the stock solution is diluted with the same buffer to make a 10-mM<br />

solution from which a standard concentration curve is generated. Each in a<br />

series of known a-ketobutyrate concentrations is prepared in a volume of<br />

200 ml and transferred to a glass test tube (100x13 mm); each point in the<br />

series is assayed in duplicate. Three hundred ml of the 2,4-dinitrophenylhydrazine<br />

reagent (0.2 % 2,4-dinitrophenyl-hydrazine in 2 N HCl; Sigma-<br />

Aldrich Co.) is added to each glass tube and the contents are vortexed and<br />

incubated at 30 °C for 30 min during which time the a-ketobutyrate is derivatized<br />

as a phenylhydrazone. The color of the phenylhydrazone is developed by<br />

the addition of 2.0 ml of 2 N NaOH; after mixing, the absorbance of the mixture<br />

is measured at 540 nm.<br />

5.1 Assay of ACC Deaminase Activity in Bacterial Extracts<br />

Fig. 1. Standard curve of<br />

a-ketobutyrate versus<br />

absorbance at 540 nm<br />

5.1.1 Preparation of Bacterial Extracts<br />

ACC deaminase activity is measured in bacterial extracts prepared in the following<br />

manner. Bacterial cell pellets, prepared as described in Section 3, are<br />

each suspended in 1 ml of 0.1 M Tris-HCl, pH 7.6 and transferred to a 1.5-ml<br />

microcentrifuge tube. The contents of the 1.5-ml microcentrifuge tube are<br />

centrifuged at 16,000xg for 5 min in a Brinkmann microcentrifuge and the<br />

supernatant is removed with a fine-tip transfer pipette. The pellet is suspended<br />

in 600 ml of 0.1 M Tris-HCl, pH 8.5. Thirty ml of toluene is added to the<br />

cell suspension and vortexed at the highest setting for 30 s.At this point, a 100ml<br />

aliquot of the “toluenized cells” is set aside and stored at 4 °C for protein<br />

assay at a later time. The remaining toluenized cell suspension is immediately<br />

assayed for ACC deaminase activity.


26 Quantifying the Impact of ACC Deaminase-Containing Bacteria on Plants 495<br />

5.1.2 Measurement of ACC Deaminase Activity<br />

All sample measurements are carried out in duplicate. Two hundred ml of the<br />

toluenized cells is placed in a fresh 1.5-ml microcentrifuge tube; 20 ml of 0.5 M<br />

ACC is added to the suspension, briefly vortexed, and then incubated at 30 °C<br />

for 15 min. Following the addition of 1 ml of 0.56 N HCl, the mixture is vortexed<br />

and centrifuged for 5 min at 16,000xg in a Brinkmann microcentrifuge<br />

at room temperature. One ml of the supernatant is vortexed together with<br />

800 ml of 0.56 N HCl in a clean glass tube (100x13 mm). Thereupon, 300 ml of<br />

the 2,4-dinitrophenylhydrazine reagent (0.2 % 2,4-dinitrophenylhydrazine in<br />

2 N HCl) is added to the glass tube, the contents vortexed and then incubated<br />

at 30 °C for 30 min. Following the addition and mixing of 2 ml of 2 N NaOH,<br />

the absorbance of the mixture is measured at 540 nm.<br />

The absorbance of the assay reagents including the substrate, ACC, and the<br />

bacterial extract are taken into account. After the indicated incubations, the<br />

absorbance at 540 nm of the assay reagents in the presence of ACC is used as<br />

a reference for the spectrophotometric readings; it is subtracted from the<br />

absorbance of the bacterial extract plus the assay reagents in the presence of<br />

ACC. The contribution of the extract, i.e., the absorbance at 540 nm of extract<br />

and the assay reagents without ACC, is determined and subtracted from the<br />

absorbance value calculated above. This value is used to calculate the amount<br />

of a-ketobutyrate generated by the activity of ACC deaminase.<br />

6 Measurement of ACC in Plant Roots, Seed Tissues and Seed<br />

Exudates<br />

In order to be able to test the model described earlier, we required a method<br />

of measuring ACC in <strong>plant</strong> tissues. Since all of the available methods for ACC<br />

quantification had problems and limitations associated with their use, we<br />

adapted the Waters AccQ•Tag Method, designed to measure amino acids, for<br />

ACC analysis. This procedure is simple and relatively sensitive. ACC, which is<br />

an amino acid, is derivatized with the Waters AccQ•Fluor reagent; the ACC<br />

derivatives are separated by reversed phase HPLC and quantified by fluorescence.We<br />

have used this procedure to quantify the amount of ACC in extracts<br />

of germinating canola seeds, seedling roots, and seed exudate (Penrose et al.<br />

2001; Penrose and Glick 2001).<br />

6.1 Collection of Canola Seed Tissue and Exudate During Germination<br />

Canola seed tissue and exudate is collected from 200-seed samples exposed to<br />

various treatments and then incubated in the dark for up to 50 h. The seeds<br />

are disinfected immediately before use. Two hundred seeds (0.400±0.008 g)


496<br />

Donna M. Penrose and Bernard R. Glick<br />

are measured into an aluminum weigh boat and soaked in 5 ml of 10 % hydrogen<br />

peroxide at room temperature (Bayliss et al. 1997).After 2 min, the hydrogen<br />

peroxide solution is removed by suction and the seeds are rinsed with<br />

sterile distilled water at least four times. Each dish is then incubated at room<br />

temperature for 1 h with 5 ml of the appropriate treatment: 0.03 M MgSO 4,<br />

(used as a negative control) or bacterial cells (grown as described in Sect. 3)<br />

suspended in 0.03 M MgSO 4 and diluted to an absorbance of 0.15 at 600 nm.<br />

Following incubation, the solution used for seed treatment (0.03 M MgSO 4 or<br />

bacterial suspension) is removed from the seeds and they are rinsed twice<br />

with sterile distilled water.After the water is removed by suction, the seeds are<br />

transferred to a 100-mm nylon sterile cell strainer (Becton Dickinson Labware,<br />

Franklin Lakes, NJ, USA) set into a sterile disposable polypropylene<br />

Petri plate (60x15 mm). One ml of autoclaved distilled water is added to each<br />

Petri dish and the Petri plates are placed in loosely covered plastic containers.<br />

The containers are incubated in the dark at 20±1 °C. After 20 h of incubation,<br />

1 ml of sterile water is added to the remaining Petri dishes and following 44 h<br />

of incubation, another1 ml of water is added to the samples.<br />

At specified times after seed treatment, duplicate Petri plates are removed<br />

from the growth chamber. The cell sieve is removed from each Petri plate and<br />

the seeds transferred by sterile forceps to autoclaved screw-capped 1.5-ml<br />

microcentrifuge tubes (VWR Canlab, Canada). The tubes are immediately<br />

placed in liquid nitrogen and the frozen seeds stored at –80 °C.<br />

After the germinating seedlings have been gathered from the strainers at<br />

each time point, the seedling exudate is removed from the Petri plate (and any<br />

clinging to the cell strainer) with a 1-ml sterile disposable syringe fitted with<br />

a #20 gauge needle (Becton Dickinson Labware, Franklin Lakes, NJ, USA). The<br />

exudate is filtered through a 0.2-mm sterile syringe filter (Gelman Sciences,<br />

Ann Arbour, MI, USA), pre-wetted with sterile distilled water. The filtrate is<br />

collected into 1.5-ml glass vials (12x32 mm) capped with silicon septa (75/10)<br />

and polypropylene open top lids (Chromatographic Specialities Inc., Brockville,<br />

ON, Canada) and immediately frozen at –80 °C.<br />

6.2 Preparation of Plant Extracts<br />

We used a modification of the protocol described by Siefert et al. (1994) to<br />

make extracts of the canola seed-samples and the roots of the 4.5-day-old<br />

seedlings grown for the root elongation assay. Roots excised from the approximately<br />

60 seedlings grown for the root elongation assay, are set in aluminum<br />

weigh boats, immediately frozen in liquid nitrogen and stored at –80 °C.All of<br />

the glassware used in the preparation of crude <strong>plant</strong> extracts, i.e., mortars and<br />

pestles, solution bottles, centrifuge tubes, pipettes, Pasteur pipets, and glass<br />

vials and silicon septa, is heated overnight at 275 °C and cooled to room temperature<br />

just prior to use. Each of the frozen tissue samples is ground in a pre-


26 Quantifying the Impact of ACC Deaminase-Containing Bacteria on Plants 497<br />

chilled mortar and pestle, suspended in 2.5 ml of 0.1 M sodium acetate pH 5.5<br />

and kept on ice for 15 min. The contents of the mortar are scraped into a 15ml<br />

glass centrifuge tube and the mortar and pestle are rinsed with 0.5 ml of<br />

the same buffer. The ground tissue suspension, together with the rinses, is<br />

centrifuged in an SS34 rotor at 17,500xg in a Sorvall R5C/B centrifuge for<br />

15 min at 4 °C to remove cell debris. The supernatant is collected and clarified<br />

by centrifugation in a Beckman L8–70 ultracentrifuge at 100,000¥g in a 70.1<br />

Ti rotor for 1 h at 4 °C and then, if necessary, by an additional centrifugation<br />

at 100,000xg for 15 min. The clarified supernatant is collected and distributed<br />

into 1-ml aliquots, some of which are stored at –80 °C in glass vials for ACC<br />

determination by HPLC, and the remainder stored in 1.5-ml microcentrifuge<br />

tubes at 4 °C for protein determination.<br />

6.3 Protein Concentration Assay<br />

The protein concentrations are measured according to a protocol based on<br />

the method of Bradford (1976) and BSA (bovine serum albumin) is used as<br />

the standard protein. Each point on the standard curve and all of the samples<br />

are assayed in triplicate.<br />

6.3.1 Protein Concentration Assay of Bacterial Extracts<br />

The 100-ml aliquots of toluenized cell suspensions, which have been set aside<br />

and stored at 4 °C during the preparation of crude bacterial cell extracts, are<br />

each mixed with 100 ml of 0.1 N NaOH and incubated for 10 min at 100 °C.<br />

After the mixtures have cooled, between 20 and 50 ml of each sample is transferred<br />

to a clean glass test tube (100x13 mm); the volume is adjusted to 100 ml<br />

with 0.1 M Tris-HCl pH 8.5, and 5 ml of the diluted dye reagent is added to the<br />

tube. The contents of the tube are vortexed and incubated for 5–20 min at<br />

room temperature. The absorbance of the samples is measured at 595 nm.<br />

6.3.2 Protein Concentration Assay of Plant Extracts<br />

Aliquots of the <strong>plant</strong> extracts, set aside and stored at 4 °C, are each transferred<br />

to clean glass test tubes (100x13 mm) and the volume is adjusted to 100 ml<br />

with 0.1 M sodium acetate pH 5.5. Varying amounts of the different extracts<br />

are transferred to the tubes, depending on the concentration of the extract:<br />

routinely, 30 ml of seed extract and 100 ml of root extract are used. Sufficient<br />

buffer is added to each tube to bring the volume up to 100 mL.After 5 ml of the<br />

diluted dye reagent are added to each test tube, it is vortexed and incubated at<br />

room temperature between 5 and 20 min. The absorbance of each sample is<br />

measured at 595 nm.


498<br />

Donna M. Penrose and Bernard R. Glick<br />

6.4 Measurement of ACC by HPLC<br />

6.4.1 Chemicals<br />

The Waters AccQ •Fluor Reagent Kit, AccQ•Tag eluent A concentrate (a premixed<br />

concentrated acetate-phosphate buffer) and the amino acid standard, a<br />

mixture of 17 amino acids (tryptophan, glutamine, and asparagine not<br />

included) each at a concentration of 2.5 mM with the exception of cysteine<br />

which is 1.25 mM, are supplied by Waters Limited. The Waters AccQ•Fluor<br />

Reagent Kit contains the chemicals for derivatization: AccQ•Fluor reagent<br />

powder (6-aminoquinolyl-N-hydroxysuccinimidyl carbamate; AQC), AccQ•<br />

Fluor reagent borate buffer and AccQ•Fluor reagent diluent (acetonitrile).<br />

ACC, b- and g-aminobutyric acid are purchased from Calbiochem-<br />

Novabiochem Corp. (La Jolla, CA, USA), HPLC grade acetonitrile from Caledon<br />

Laboratories (Georgetown, ON, Canada), a-aminobutyric acid from<br />

Fisher Scientific, and L-a-(2-amino-ethoxyvinyl) glycine hydrochloride<br />

(AVG) from Sigma-Aldrich Co. All water used is purified by a Milli-Q Water<br />

System (Millipore Co. Bedford, MA, USA), autoclaved and then filtered<br />

through a 0.45-mm HA filter (Millipore Co. Bedford, MA, USA).<br />

6.4.2 Treatment of Glassware<br />

All glassware used in this procedure is washed and then flushed at least six<br />

times with tap water, twice with deionized water and twice more with distilled<br />

water. Just prior to use, the cleansed glassware is wrapped in aluminum foil,<br />

heated overnight at 275 °C and cooled to room temperature. Solutions and<br />

samples are stored in heat-treated bottles and vials (including septa and lids).<br />

6.4.3 Preparation of Standard Solutions<br />

Stock 2.5-mM solutions of ACC,a-aminobutyric acid,b-aminobutyric acid,gaminobutyric<br />

acid, and a mixture of 17 amino acids are prepared in 25 ml of<br />

0.1 N HCl in a 25-ml volumetric flask. These solutions are diluted with sterile<br />

distilled water to yield a concentration of 0.1 mM. The 2.5-mM and 0.1-mM<br />

stock solutions are divided into 0.5-ml aliquots, frozen at –20 °C, thawed once<br />

when needed and then discarded.With the exception of ACC,the 0.1-mM solutions<br />

are further diluted with sterile distilled water to generate concentrations<br />

between 5 and 25 pmol/20 ml injection. Dilutions of the 0.1-mM solutions of<br />

ACC yield between 1 and 25 pmol ACC/20 ml injection. Standard mixtures of<br />

ACC,a-,b-,and g - aminobutyric acids are prepared in sterile distilled water to<br />

yield 12.5 pmol/20 ml injection. The amino acid standard is diluted such that<br />

each 20-ml injection included 25 pmol of each of the 17 amino acids with the<br />

exception of the amount of cysteine which was 12.5 pmol.Aliquots of the standard<br />

solutions are frozen at –20 °C, and when required, thawed once and used.


26 Quantifying the Impact of ACC Deaminase-Containing Bacteria on Plants 499<br />

6.4.4 Derivatization Procedure<br />

Standard solutions of ACC; ACC, a-, b- and g -aminobutyric acids; the amino<br />

acids and <strong>plant</strong> extracts are coupled with ACQ according to the directions in the<br />

Waters AccQ•Fluor Reagent Kit Instruction Manual.The AccQ•Fluor derivatization<br />

reagent, once reconstituted, is stable for 1 week. The derivatization reagent<br />

is reconstituted by adding 1 ml of acetonitrile (vial 2B) to the AccQ•Fluor<br />

reagent powder, vortexing for 10 s, and heating on top of a 55 °C heating block<br />

for no more than 10 min to dissolve the powder.The concentration of the reconstituted<br />

AccQ•Fluor reagent is approximately 10 mM in acetonitrile; amino acid<br />

derivatization is optimal when the reconstituted AccQ•Fluor reagent is in excess<br />

and the pH is between pH 8.2 and 10. The derivatization reactions are carried<br />

out in duplicate in 6x55 mm glass sample tubes (Waters Limited).Ten ml of standard<br />

or sample solution is placed in each tube; 70 ml of AccQ•Fluor borate buffer<br />

is added to it and the mixture is immediately vortexed for several seconds. Following<br />

the addition of 20 ml of reconstituted AccQ•Fluor, the mixture is briefly<br />

vortexed again,allowed to stand at room temperature for 1 min and then heated<br />

at 55 °C for 2 min in a heating block. Once cooled to room temperature<br />

(5–10 min) the solution may be injected immediately or sometime during the<br />

next week.Amino acids derivatized by this procedure are quite stable and can be<br />

stored at room temperature for at least 1 week.<br />

6.4.5 HPLC Determination of ACC Content<br />

The AccQ•Tag Column, a high-efficiency 4 mm Nova-Pak C 18 column specifically<br />

certified for use with the AccQ•Tag Method (Waters Limited) is used to<br />

separate the amino acid derivatives produced by the AccQ•Fluor derivatization<br />

reaction, and a Hewlett Packard column heater is used to maintain the<br />

column temperature at 37 °C. Amino acid derivatives are detected and measured<br />

by using a Hewlett Packard HPLC system which consists of a 1050<br />

Series Quaternary Pump and a 104a Programmable Fluorescence Detector. A<br />

PC computer system (DTK 3300 386/33) is used to run the supporting computer<br />

software, i.e., Hewlett Packard’s ChemStation (DOS Series).<br />

The solvent system includes eluent A, a diluted solution of Waters AccQ•Tag<br />

acetate-phosphate buffer concentrate prepared daily, (50 ml concentrate<br />

diluted with 500 ml 18 Megohm Milli–Q water), eluent B, HPLC-grade acetonitrile,<br />

and eluent C, 18 Megohm Milli-Q water. The solvents are continuously<br />

sparged with helium and the solvent lines are purged for at least 60 s<br />

prior to use to remove any air bubbles present. The AccQ•Tag column is conditioned<br />

with 60 % eluent B/40 % eluent C at a flow rate of 1 ml/min for 30 min<br />

and then equilibrated with 100 % eluent A for 10 min at a flow rate of 1 ml/min<br />

before injection of the first sample. The gradient recommended by Waters<br />

Limited for separation of the AccQ•Tag-labelled amino acids was modified to<br />

enhance resolution of the ACC peak (Table 2).


500<br />

Donna M. Penrose and Bernard R. Glick<br />

Table 2. Gradient table for Waters AccQ•Tag system modified for ACC elution<br />

Time (min) Flow rate (ml/min) A (%) B (%) C (%)<br />

0 1.0 100.0 0 0<br />

0.5 1.0 99.0 1.0 0<br />

3.0 1.0 91.0 9.0 0<br />

13.0 1.0 88.0 12.0 0<br />

14.0 1.0 83.0 17.0 0<br />

16.0 a 1.0 0 60 40<br />

18.0 1.0 100.0 0 0<br />

23.0 1.0 100.0 0 0<br />

Abbreviations: A, Waters AccQ•Tag acetate-phosphate buffer concentrate (50 ml diluted<br />

with 500 ml 18 Megohm Milli-Q water); B, HPLC-grade acetonitrile; C, 18 Megohm Milli-<br />

Q water<br />

a From this point in the gradient, the column is washed and conditioned for the next<br />

sample<br />

The Hewlett Packard 104a Programmable Fluorescence Detector is set up<br />

according to the Waters AccQ•Tag Amino Acid Analysis Method and is turned<br />

on at least 40 min prior to sample injection. The settings are as follows: excitation<br />

wavelength, 250 nm; emission wavelength, 395 nm; response time, 4;<br />

pmt gain, 15, and lamp setting, 3–5 W/220 Hz.<br />

Once the column is conditioned and equilibrated, and the detector is<br />

warmed up, a standard solution, containing 12.5 pmol of a-, b-, and g -<br />

aminobutyric acid, is injected. Following the injection of standard solutions,<br />

samples are injected and analyzed; the run time for each sample is 23 min and<br />

includes washing and re-equilibrating the column following the separation of<br />

the derivatized amino acids. Duplicates of each standard and sample are<br />

derivatized and injected. The needle port is rinsed with eluent A prior to each<br />

injection in order to reduce contamination from previously injected samples.<br />

The injection volume of all samples including blanks, standards and <strong>plant</strong><br />

extracts is 20 ml. Plant tissue extracts are diluted just prior to derivatization.<br />

The quantity of sample hydrolyzed and derivatized in 20 ml is estimated to be<br />

0.1–1.0 mg (4–40 pmol) of protein, based on a protein average molecular<br />

weight of 25,000 Daltons.<br />

6.4.6 Quantification of ACC<br />

The amount of ACC in samples is quantified by using an ACC standard curve<br />

that is linear between 1 and 25 pmol of ACC per sample (Fig. 2). The ACC standard<br />

curve is prepared from a fresh stock solution of ACC (0.1 mM) diluted<br />

with sterile distilled water to yield between 1 and 25 pmol of ACC/20-ml injection.<br />

The ACC dilutions are derivatized, and following injection, are eluted


26 Quantifying the Impact of ACC Deaminase-Containing Bacteria on Plants 501<br />

Fig. 2. Standard curve of<br />

ACC measured in fluorescence<br />

units<br />

Fluorescence units<br />

20000<br />

15000<br />

10000<br />

5000<br />

from the AccQ•Tag column at approximately 7.6 min. Similar standard curves<br />

may be prepared for a-, b- and g- aminobutyric acid, metabolites of ACC,<br />

which are eluted from the AccQ•Tag column at 8.2, 8.7 and 9.2 min, respectively.<br />

References and Selected Reading<br />

0 5 10 15 20 25<br />

ACC, pmoles<br />

Abeles FB, Morgan PW, Saltveit ME Jr (1992) Ethylene in <strong>plant</strong> biology, 2nd edn. Academic<br />

Press, New York<br />

Bayliss C, Bent E, Culham DE, MacLellan S, Clarke AJ, Brown GL, Wood JM (1997) Bacterial<br />

genetic loci implicated in the Pseudomonas putida GR12–2R3 – canola mutualism:<br />

identification of an exudate-inducible sugar transporter. Can J Microbiol<br />

43:809–818<br />

Bradford M (1976) A rapid and sensitive method for the quantitation of microgram<br />

quantities of protein utilizing the principle of protein-dye binding. Anal Biochem<br />

73:248–258<br />

Dworkin M, Foster J (1958) Experiments with some microorganisms which utilize<br />

ethane and hydrogen. J Bacteriol 75:592–601<br />

Glick BR, Karaturovíc DM, Newell PC (1995) A novel procedure for rapid isolation of<br />

<strong>plant</strong> growth promoting pseudomonads. Can J Microbiol 41:533–536<br />

Glick BR, Penrose DM, Li J (1998) A model for the lowering of <strong>plant</strong> ethylene concentrations<br />

by <strong>plant</strong> growth-promoting bacteria. J Theor Biol 190:63–68<br />

Honma M, Shimomura T (1978) Metabolism of 1-aminocyclopropane-1-carboxylic acid.<br />

Agric Biol Chem 42:1825–1831<br />

Jacobson CB, Pasternak JJ, Glick BR (1994) Partial purification and characterization of 1aminocyclopropane-1-carboxylate<br />

deaminase from the <strong>plant</strong> growth promoting rhizobacterium<br />

Pseudomonas putida GR12–2. Can J Microbiol 40:1019–1025<br />

Lifshitz R, Kloepper JW, Scher FM, Tipping EM, Laliberté M (1986) Nitrogen-fixing<br />

Pseudomonads isolated from roots of <strong>plant</strong>s grown in the Canadian High Arctic.Appl<br />

Environ Microbiol 51:251–255


502<br />

Donna M. Penrose and Bernard R. Glick<br />

Lifshitz R, Kloepper JW, Kozlowski M, Simonson C, Carlson J, Tipping EM, Zaleska I<br />

(1987) Growth promotion of canola (rapeseed) seedlings by a strain of Pseudomonas<br />

putida under gnotobiotic conditions. Can J Microbiol 33:390–395<br />

Ma J-H, Yao J-L, Cohen D, Morris B (1998) Ethylene inhibitors enhance in vitro formation<br />

from apple shoot cultures. Plant Cell Rep. 17:211–214<br />

Mattoo AK, Suttle CS (1991) The <strong>plant</strong> hormone ethylene. CRC Press, Boca Raton, FL, p<br />

337<br />

Penrose DM, Glick BR (2001) Levels of 1-aminocyclopropane-1-carboxylic acid (ACC) in<br />

exudates and extracts of canola seeds treated with <strong>plant</strong> growth-promoting bacteria.<br />

Can J Microbiol 47:368–372<br />

Penrose DM, Moffatt BA, Glick BR (2001) Determination of 1-aminocyclopropane-1-carboxylic<br />

acid (ACC) to assess the effects of ACC deaminase-containing bacteria on<br />

roots of canola seedlings. Can J Microbiol 47:77–80<br />

Shah S, Li J, Moffatt BA, Glick BR (1997) ACC deaminase genes from <strong>plant</strong> growth promoting<br />

bacteria. In: Ogoshi A, Kobayashi K, Homma Y, Kodama F, Kondo N, Akino S<br />

(eds) Plant growth-promoting rhizobacteria: present status and future prospects.<br />

OECD, Paris, pp 320–324<br />

Siefert F, Langebartels C, Boller T, Grossmann K (1994) Are ethylene and 1-aminocyclopropane-1-carboxylic<br />

acid involved in the induction of chitinase and b-1,-3-glucanase<br />

activity in sunflower cell-suspension cultures? Planta 192:431–440<br />

Yang SF, Hoffman NE (1984) Ethylene biosynthesis and its regulation in higher <strong>plant</strong>s.<br />

Annu Rev Plant Physiol 35:155–189


27 Applications of Quantitative Microscopy in<br />

Studies of Plant Surface Microbiology<br />

Frank B. Dazzo<br />

1 Introduction<br />

“Sometimes what counts can’t be counted,<br />

and what can be counted doesn’t count.”<br />

(Albert Einstein)<br />

Whereas the animal carries its major community of indigenous microflora<br />

(generally of a beneficial kind) on the moist warm walls of its peristaltic gut,<br />

the <strong>plant</strong> does likewise, but on its entire exposed <strong>surface</strong>s, from apical tip to<br />

root cap. These <strong>plant</strong> <strong>surface</strong>s represent an oozing, flaking layer of integument<br />

which discharges a wide range of substances that support a vast number of<br />

spatially discrete and specialized microbial communities, including parasites<br />

and symbionts that can have a major impact on <strong>plant</strong> growth and development.<br />

A modern view of the <strong>plant</strong> <strong>surface</strong> is now seen as a dynamic adaptable<br />

envelope, flexible in both its import and export of materials, forming a<br />

<strong>plant</strong>–microbe ecosystem in its own right and the first barrier between the<br />

moist, concentrated, balanced <strong>plant</strong> cell and a hostile ever-changing external<br />

environment.<br />

Manipulation of the <strong>plant</strong> <strong>surface</strong> microflora to improve its health is a longstanding<br />

goal in <strong>plant</strong> <strong>microbiology</strong>. However, efforts to exploit this type of<br />

biological control have frequently been impeded because of major technical<br />

difficulties that must be overcome to fully understand the microbial ecology<br />

of this ecosystem, especially the lack of ability to extract in situ data that are<br />

both informative and quantifiable at spatial scales relevant to the ecological<br />

niches of the microorganisms involved.<br />

Most of this chapter describes the author’s development and utilization of<br />

quantitative microscopy in studies of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>. The majority<br />

of this work has been done to gain a better understanding of the Rhizobium-legume<br />

root-nodule symbiosis. Various types of microscopy have been<br />

employed, including brightfield, phase-contrast, Nomarski-interference contrast,<br />

polarized light, real-time and time-lapse video, darkfield, conventional<br />

and laser scanning confocal epifluorescence, scanning electron, transmission<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


504<br />

Frank B. Dazzo<br />

electron, and field-emission scanning/transmission electron microscopies<br />

combined with visual counting techniques and manual interactive applications<br />

of image analysis. More recently, the author has led a team of scientists<br />

to develop a new generation of innovative, customized image analysis software<br />

designed specifically to analyze digital images of microbial populations<br />

and communities and extract all the informative, quantitative data of in situ<br />

microbial ecology from them at spatial scales relevant to the microbes themselves.<br />

We have begun to apply this new computer-assisted imaging technology<br />

to the fascinating field of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>. The chapter<br />

includes many figures that exemplify how the awesome resolving power of the<br />

microscope has significantly enhanced our understanding of <strong>plant</strong> <strong>surface</strong><br />

<strong>microbiology</strong>, and richly illustrates how this topic area is even more enhanced<br />

with the added dimension of quantitation using computer-assisted digital<br />

image analysis.<br />

2 Quantitation of Symbiotic Interactions Between<br />

Rhizobium and Legumes by Visual Counting Techniques<br />

2.1 The Modified Fåhraeus Slide Culture Technique for Studies of the<br />

Root–Nodule Symbiosis<br />

The slide culture technique of Fåhraeus (1957) was the single, most important<br />

method developed to facilitate the microscopical examination of the infection<br />

process in the Rhizobium-legume symbiosis, especially with small-seeded<br />

legumes like white clover in symbiosis with its root-nodule endosymbiont, R.<br />

leguminosarum bv. trifolii. This simple method of culturing the symbionts<br />

under microbiologically controlled conditions made it possible to examine<br />

the interactions between the <strong>plant</strong> and microbial symbionts by various types<br />

of microscopy, including a classic time-lapse cinema depicting the developmental<br />

morphology of clover root hair infection (Nutman et al. 1973). Phase<br />

contrast microscopy using this slide culture technique also revealed the paramount<br />

importance of host specificity in the infection process at the stage of<br />

infection thread formation within host root hairs (Li and Hubbell 1969).<br />

The original Fåhraeus slide method involved vertical cultivation of a<br />

seedling on a microscope slide within a large enclosed tube containing an isotonic<br />

nitrogen-free <strong>plant</strong> culture medium, and with its root inoculated with<br />

rhizobia embedded in an agar medium beneath a large cover slip (Fåhraeus<br />

1957).Various modifications of this slide culture technique have been made to<br />

further facilitate detailed microscopical examinations of the infection<br />

process. For instance, the embedding agar was found unnecessary even for<br />

cultivation of two seedlings per slide. Elimination of the embedding agar permitted<br />

the symbionts to interact unimpeded by this fibrous matrix, the roots<br />

to be processed more consistently and efficiently after an appropriate period


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 505<br />

of incubation, and more detailed microscopy to be performed with a cleaner,<br />

phase-transparent background. These added features have significantly<br />

improved the signal-to-noise ratio of image quality, making it possible to<br />

accurately quantify many of the pre-infection and post-infection events<br />

occurring on root hairs in vivo at single bacterial cell resolution, including<br />

rhizobial attachment to root hairs (phenotype Roa) of constant length by<br />

phase-contrast microscopy (Dazzo et al. 1976; Dazzo 1982). The results of<br />

studies using this quantitative microscopical counting technique revealed<br />

important spatio-temporal aspects of the Roa phenotype, including its distinct<br />

cellular orientations/patterns/phases of adhesion, the positive relationship<br />

of certain patterns of attachment to host specificity, the importance of<br />

cell-<strong>surface</strong> glycoconjugates and saccharide-binding host lectins to symbiont<br />

recognition, the inhibition of symbiont recognition and infection by combined<br />

nitrogen, and the manipulation of rhizobial genes affecting cell <strong>surface</strong><br />

components and rhizobial attachment to host root hairs (Dazzo and Hubbell<br />

1975; Dazzo et al. 1976, 1978, 1984; Dazzo and Brill 1978, Sherwood et al. 1984;<br />

Rolfe et al. 1996). This same modification of the slide culture technique also<br />

made it possible to quantitate clover root hair infection. Thus, quantitative<br />

microscopy of the infection process resulted in the discovery of potent, stimulating<br />

infection-related biological activities of various purified rhizobial<br />

components required for primary host infection by R. leguminosarum bv.<br />

trifolii, including its clover lectin-binding acidic heteropolysaccharides and<br />

corresponding oligosaccharide repeat unit fragments which retained their<br />

affinity for the clover lectin, its clover lectin-binding lipopolysaccharide glycoform,<br />

and its diverse family of membrane chitolipooligosaccharides that<br />

modulate cell wall architecture and growth physiology of these target differentiated<br />

host cells (Abe et al. 1984; Dazzo et al. 1991, 1996). Further applications<br />

of this modified Fåhraeus slide technique to study the R. leguminosarum<br />

bv. trifolii-white clover symbiosis have utilized real-time video<br />

microscopy and digital image analysis of track-reconstructions to define the<br />

quantitative influence of root secretions on rhizobial motility in situ in the<br />

aqueous, external clover root environment (Dazzo and Petersen 1989), and of<br />

cells and purified lectin-binding lipopolysaccharide of Rhizobium on cytoplasmic<br />

streaming in root hairs indicating activation of their cytoskeleton<br />

activity (Dazzo and Petersen 1989, Dazzo et al. 1991).Another modification of<br />

the Fåhraeus slide technique was to culture seedlings vertically and flat on<br />

small agarose-solidified plates with a portion of their roots covered with the<br />

same nitrogen-free medium and small coverslips. This modification plus the<br />

customized construction of a “horizontal growth station” created the opportunity<br />

to perform real-time and time-lapse video microscopy of seedling<br />

roots grown axenically and geotropically with as little as 10 ml volumes of bacterial<br />

test solutions. Applications of this technique resulted in the detection<br />

and quantitation of symbiosis-related growth responses of clover root hairs to<br />

minute quantities of several different types of bioactive metabolites made by


506<br />

Frank B. Dazzo<br />

the microsymbiont, R. leguminosarum bv. trifolii under strict microbiologically<br />

controlled conditions (Dazzo et al. 1987, 1996; Dazzo and Petersen 1989,<br />

Hollingsworth et al. 1989, Philip-Hollingsworth et al. 1991; Orgambide et al.<br />

1994, 1996). This technique was also used in conjunction with engineered rhizobia<br />

containing reporter gene fusions to locate attached rhizobial cells<br />

expressing pSym nod genes in situ on root hair tips (Dazzo et al. 1988).<br />

2.2 Attachment of Rhizobia to Legume Root Hairs<br />

Although attachment of rhizobia to legume root hairs (Roa [Root attachment]<br />

phenotype) has often been described as a simple, one-step event lacking any<br />

form of specificity, this is a gross oversimplification of the real case. Instead,<br />

quantitative time-resolved microscopy at single bacterial-cell resolution<br />

reveals that Roa is a dynamic, multiphase process including distinct nonspecific<br />

and host-specific events. Figure 1 summarizes a unified view of this<br />

dynamic sequence of events involved in attachment of encapsulated rhizobia<br />

to host legume root hairs (Dazzo et al. 1984). This model culminates in the<br />

development of the specific Roa-3 pattern of R. leguminosarum bv. trifolii<br />

attachment to white clover root hairs in modified Fåhraeus slide cultures prepared<br />

with a relatively small, defined size inoculum of fully encapsulated cells<br />

(Dazzo et al. 1984). This pattern of rhizobial attachment to root hairs (an<br />

immobilized aggregate of cells at the root hair tip and individual polarly<br />

attached cells along the shaft of the same root hair) requires the intervention<br />

of bacterial proteins and polysaccharides, host lectin, and enzymes that<br />

Fig. 1. Diagram of the<br />

dynamic phases of rhizobial<br />

attachment to<br />

host root hairs (Roa),<br />

based on studies using<br />

phase contrast light<br />

microscopy, scanning<br />

electron microscopy,<br />

and transmission electron<br />

microscopy. Cell<br />

sizes are approximately<br />

proportional. Reprinted<br />

with permission from<br />

the American Society<br />

for Microbiology


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 507<br />

Fig. 2. Phase contrast microscopy (A, C, E) and scanning electron microscopy (B, D, F)<br />

of distinct patterns of attachment of R. leguminosarum bv. trifolii to white clover root<br />

hairs. A, B Phase 1A=Roa-1, C, D phase 1C=Roa-2, E phase 1A+1C=Roa-3, F phase 2 with<br />

associated microfibrils. Scale bar A and C 20 mm, B 2 mm, D, F 1 mm, E 15 mm


508<br />

Frank B. Dazzo<br />

degrade the bacterial polysaccharides; it exhibits host-selectivity and is found<br />

on approximately 95 % of successfully infected root hairs in the Rhizobiumwhite<br />

clover symbiosis (Dazzo and Hubbell 1975; Dazzo et al. 1976, 1982, 1984;<br />

Dazzo and Brill 1979; Sherwood et al. 1984; Rolfe et al. 1996; Smit et al. 1992).<br />

The Phase 1A pattern of randomly oriented attachment occurs within 15 min<br />

of inoculation, and involves an initial nonhost-specific interaction of a rhizobial<br />

<strong>surface</strong> protein “rhicadhesin” on individual bacteria with the root hair tip<br />

(Smit et al. 1992), followed within the first hour by a more host-specific aggregation<br />

of bacterial cells immobilized at the root hair tip and mediated by an<br />

excreted, multivalent host lectin. Cells that have not yet attached to the host<br />

root become polarly encapsulated in the external root environment during<br />

the next 4–8 h (Phase 1B), due to the combined action of “polarase” enzymes<br />

in root exudate and de novo synthesis of a new capsule at one cell pole (Dazzo<br />

et al. 1982; Sherwood et al. 1984). Beginning approximately 4 h after inoculation,<br />

these polarly encapsulated cells attach “end-on”, i.e., perpendicular to<br />

the <strong>surface</strong> along the sides of the same root hair (phase 1C). Phase 1 attachment<br />

is distinguished from phase 2 adhesion by the significantly increased<br />

strength of adhesion of attached cells detected approximately 12 h after inoculation,<br />

concurrent with the elaboration of extracellular microfibrils that<br />

increase the degree of contact of the attached bacteria to the root hair <strong>surface</strong><br />

(Dazzo et al. 1984). Indeed, this strength of Phase 2 rhizobial adhesion to<br />

legume host root hairs is immense, exceeding that which anchors some root<br />

hairs onto the root itself! Figure 2A–F is a series of phase contrast light micrographs<br />

and scanning electron micrographs that illustrate each of these distinct<br />

patterns of rhizobial attachment to white clover root hairs (Dazzo and<br />

Brill 1979; Dazzo et al. 1984).<br />

2.3 Rhizobium-Induced Root Hair Deformations<br />

Root hairs on axenic seedlings are straight, but become deformed (Had [Hair<br />

deformation] phenotype) during growth in response to various bioactive<br />

metabolites made by rhizobia. Four different morphotypes of white clover<br />

Had are induced under axenic conditions by minute quantities of purified<br />

bioactive Nod metabolites made by R. leguminosarum bv. trifolii. These are<br />

root hair distortions, tip swellings, branches, and corkscrews induced by rhizobial<br />

membrane chitolipooligosaccharides, N-acetylglutamic acid, and<br />

diglycosyl diacylglycerol glycolipids (Philip-Hollingsworth et al. 1991;<br />

Orgambide et al. 1994, 1996; Dazzo et al. 1996a, b;). Collectively called moderate<br />

Had, these various types of root hair deformations are less symbiont-specific<br />

than marked curling of the root hair tip (commonly referred to as the<br />

“Shepherd’s crook” Hac [Hair curling] phenotype). This Hac morphotype is<br />

illustrated in Fig. 3 and requires close proximity of viable cells of the homologous<br />

symbiont (Li and Hubbell 1969; Yao and Vincent 1976). This figure is a


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 509<br />

Fig. 3. Portion of a white clover root hair that<br />

has undergone a markedly curled deformation<br />

induced by Rhizobium leguminosarum<br />

bv. trifolii. This optisection obtained using<br />

laser scanning confocal microscopy and<br />

immunofluorescence staining with a strainspecific<br />

monoclonal antibody to the bacterial<br />

LPS provides direct evidence that the center of<br />

the shepherd’s crook overlap contains a clump<br />

of rhizobia. Scale bar 7.5 mm<br />

laser scanning confocal epifluorescence micrograph that elegantly provides<br />

direct evidence that the overlap of the shepherd’s crook entraps a clump of<br />

rhizobial cells, as has long been predicted, but not convincingly shown before.<br />

In this case, the confocal image is an optisection located at the optical median<br />

plane of the curled root hair cell, and it definitively shows the immunofluorescent<br />

rhizobia detected by using a fluorescent monoclonal antibody to their<br />

lipopolysaccaride (LPS) somatic O-antigen. It has been predicted that the<br />

confining morphological structure of the shepherd’s crook serves to concentrate<br />

in a localized region the metabolic events of microsymbiont penetration<br />

while preventing lysis of the root hair during primary host infection (Napoli<br />

et al. 1975a; Napoli and Hubbell 1976; Dazzo and Hubbell 1982).<br />

2.4 Primary Entry of Rhizobia into Legume Roots<br />

Figure 4 illustrates a rhizobial-induced infection thread in white clover root<br />

hairs. Successful infections of this type typically exhibit a bright refractile<br />

spot in the center overlap of markedly curled root hair tips, and infection<br />

threads that have elongated through the root hair to its base.A central event of<br />

this infection process in the Rhizobium-legume symbiosis is the modification<br />

of the host cell wall barrier to form a portal of entry large enough for bacterial<br />

penetration. Transmission electron microscopy indicates that rhizobia<br />

enter the legume root hair through a completely eroded hole that is slightly<br />

larger than the bacterial cell (generally 2–3 mm in diameter) and is presumably<br />

created by localized enzymatic hydrolysis of the host cell wall (Napoli<br />

and Hubbell 1976; Callaham and Torrey 1981). Time-lapse cinema microscopy<br />

(Nutman et al. 1973) has elegantly shown that the root hair ceases to


510<br />

Frank B. Dazzo<br />

Fig. 4. Phase contrast micrograph of primary<br />

host infection in the Rhizobium–legume symbiosis.<br />

Note the prominent infection thread<br />

(arrow) within the deformed root hair cell.<br />

Scale bar 10 mm<br />

elongate during the inward growth of the infection thread, which proceeds at<br />

approximately the same elongation rate. This inward growth of the infection<br />

thread is led by a mobile nucleus and a flurry of cytoplasmic streaming within<br />

the root hair (Nutman et al. 1973). Successful infections are best quantitated<br />

by visual counting while viewed by phase contrast microscopy; light staining<br />

of the infection thread with methylene blue can enhance contrast to detect<br />

them. Distinctions of successful vs. unsuccessful infections can be made by<br />

detailed microscopical examination to assess whether the infection thread<br />

has grown to the root hair base and penetrated into the underlying subepidermal<br />

cortical cell. Infective rhizobia engineered with Gus or green fluorescent<br />

protein reporter genes can facilitate the detection of infected root hairs,<br />

but this is overkill for skilled microscopists.<br />

An alternate primitive route of primary host infection of legumes leading<br />

to effective nodule formation is the crack entry of rhizobia into natural<br />

wounds of the host <strong>plant</strong> epidermis. This commonly occurs in many tropical<br />

legumes (Napoli et al. 1975b) and some temperate legumes, but can also occur<br />

infrequently in anomalous ineffective nodulations by rhizobia outside their<br />

normal cross-inoculation group (Hrabak et al. 1985). In the aquatic legume<br />

Neptunia natans where root hairs do not normally develop, the natural splitting<br />

of the epidermis during development of the spongy aerenchyma and<br />

emergence of adventitious and lateral roots create openings that allow “crack<br />

entry” of the rhizobial symbiont, Allorhizobium undicola, Rhizobium undicola,<br />

or Devosia neptuniae, as the normal mode of primary host infection<br />

(Subba-Rao et al. 1995).<br />

Recently, an interesting novel combination of infection events has been<br />

found to occur in development of the root-nodule symbiosis of rhizobia with<br />

tagasaste (Chamaecytisus proliferus L.), a legume indigenous to the Canary<br />

Islands near the west coast of Africa. In this symbiosis, primary host infection<br />

initially involves rhizobial deformation and penetration of host root hairs, but<br />

all these primary host infections abort and the rhizobia then revert to a crack<br />

entry mode of invasion directly into the emerging root nodules without<br />

development of infection threads (Vega-Hernandez et al. 2001). Quite a<br />

remarkable, unique mode of <strong>plant</strong> infection by <strong>surface</strong> rhizobia!


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 511<br />

2.5 In Situ Molecular Interactions Between Legumes Roots and Surface-<br />

Colonizing Rhizobia<br />

Microscopy has played a central role in elucidating molecular events important<br />

to the development of the Rhizobium-legume root-nodule symbiosis. The<br />

use of various molecular probes combined with the awesome resolving power<br />

of the microscope has made it possible to dissect and locate key molecules<br />

that participate in primary host infection, including the cell <strong>surface</strong> interfaces<br />

during symbiotic recognition, attachment, deformation, and root hair penetration,<br />

and also in root nodule development. Various types of microscopy<br />

that can view intact living cells noninvasively have added new dimensions to<br />

unraveling the symbiotic interactions of potent rhizobial signal molecules<br />

with host cells, including the precise localization of specific binding receptor<br />

sites on the host root <strong>surface</strong>, the rapid internalization of certain rhizobial signal<br />

communication molecules within root hairs and their transfer to underlying<br />

cortical cells, and various other infection-related host cell responses. The<br />

significance of all of these studies is improved when the various microscopical<br />

techniques are accompanied by quantitative methods of data acquisition.<br />

Some examples of in situ “molecular microscopy” in studies of <strong>plant</strong> <strong>surface</strong><br />

<strong>microbiology</strong> are illustrated here.<br />

2.6 Cross-Reactive Surface Antigens and Trifoliin A Host Lectin<br />

Rhizobium leguminosarum bv. trifolii and white clover roots share related<br />

<strong>surface</strong> components that are antigenically cross-reactive (Dazzo and Hubbell<br />

1975; Dazzo and Brill 1979). Quantitative immunofluorescence microscopy<br />

indicates that these cell-<strong>surface</strong> antigens are transient, symbiont-specific,<br />

infection-related, and participate in the host lectin-mediated stage of symbiont<br />

recognition on the clover root hair <strong>surface</strong> (Dazzo and Hubbell 1975;<br />

Dazzo and Brill 1979; Dazzo et al. 1979). Transformation of Azotobacter<br />

vinelandii with DNA from R. leguminosarum bv. trifolii resulted in hybrid<br />

recombinants that expressed these symbiotic cross-reactive antigens (Bishop<br />

et al. 1977), and these recombinants gained the ability to carry out the phase<br />

1A pattern of bacterial cell attachment to white clover root hair tips (Dazzo<br />

and Brill 1979). The cell <strong>surface</strong> location of these epitopes plus their infection-related<br />

symbiont-specificity, interaction with the multivalent white<br />

clover root lectin, and role in cell attachment formed the basis for proposing<br />

their involvement as cell-<strong>surface</strong> receptors in a lectin cross-bridging model<br />

of symbiont recognition during early stages of primary host infection<br />

(Dazzo and Hubbell 1975; Dazzo and Brill 1979). Recent studies using <strong>plant</strong><br />

molecular biology techniques have provided substantial evidence supporting<br />

the validity of this cross-bridging model (van Rhijn et al. 1998; Hirsch<br />

1999).


512<br />

Frank B. Dazzo<br />

Fig. 5. Symbiont-specific interaction of trifoliin A white clover lectin and R. leguminosarum<br />

bv. trifolii. A, B Transmission electron microscopy, C–F conventional immunofluorescence<br />

microscopy using antibody to purified trifoliin A. A The historical micrograph<br />

which suggested the involvement of a particulate cross-bridging clover lectin in<br />

the attachment of encapsulated R. leguminosarum bv. trifolii cells to host root hairs.<br />

B Negatively stained particles of purified trifoliin A white clover lectin. C Distribution of<br />

trifoliin A on root hair tips of white clover seedlings. D Intense binding of root-derived<br />

trifoliin A to R. leguminosarum bv. trifolii. E In situ binding of trifoliin A to the polar<br />

capsule of R. leguminosarum bv. trifolii cultured in the external clover root environment.<br />

F Direct detection of trifoliin A at the contact interface (arrow) of rhizobial cells polarly<br />

attached to a white clover root hair. Scale bar A 1 mm, B 25 nm, C 50 mm, D, E F 2 mm.<br />

Reprinted with permission from the American Society for Microbiology


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 513<br />

The ultrastructure of the docking stage of rhizobial attachment to the<br />

clover root hair <strong>surface</strong> is illustrated in Fig. 5A. This transmission electron<br />

micrograph revealed the electron-dense granules accumulated on the outer<br />

face of the hair wall that interact with the fibrillar capsule of R. leguminosarum<br />

bv. trifolii (Dazzo and Hubbell 1975). Since this granular material<br />

also occurred on the <strong>surface</strong> of axenic root hairs, it was presumably of host<br />

origin and predictably a carbohydrate-binding lectin (Dazzo and Hubbell<br />

1975). In follow-up studies, a lectin was purified from white clover seed,<br />

shown to exist as an aggregated particle of glycoprotein and to accumulate on<br />

white clover root hairs, especially at their tips, as shown by transmission electron<br />

microscopy and immunofluorescence microscopy (Dazzo et al. 1978;<br />

Gerhold et al. 1985; Fig. 5B, C). This white clover lectin displayed symbiontspecificity<br />

in agglutination of R. leguminosarum bv. trifolii and was named<br />

trifoliin A (Dazzo et al. 1978). The intense, saccharide-inhibitable binding of<br />

root trifoliin A to encapsulated cells of R. leguminosarum bv. trifolii cells is<br />

illustrated in the immunofluorescence micrograph of Fig. 5D. Subsequently, it<br />

was shown that most of the trifoliin A glycoprotein synthesized de novo in<br />

roots of white clover seedlings was excreted into the external root environment<br />

where it interacted in situ with encapsulated cells of R. leguminosarum<br />

bv. trifolii (Dazzo and Hrabak 1981, Dazzo et al. 1982; Sherwood et al. 1984;<br />

Truchet et al. 1986; Fig. 5E). Direct evidence indicating that trifoliin A accumulated<br />

at the contact interface between polarly attached R. leguminosarum<br />

bv. trifolii cells and the <strong>surface</strong> of the white clover root hair wall was shown by<br />

conventional immunofluorescence microscopy viewed with the pre-confocal<br />

optics of a high magnification objective having a narrow depth of focus<br />

(Dazzo et al. 1984; Fig. 5F). Quantitative immunofluorescence microscopy<br />

indicated that hybrid recombinants of R. leguminosarum bv. viciae carrying<br />

multicopy plasmids of cloned pSym nod genes of R. leguminosarum bv. trifolii<br />

controlling clover host specificity acquired the ability to bind trifoliin A in<br />

situ in the external white clover root environment (Philip-Hollingsworth et al.<br />

1989b).All of these findings contributed to the proposal that host lectin mediates<br />

symbiont recognition during host-specific events that precede primary<br />

host infection in the Rhizobium-legume symbiosis. Subsequent elegant <strong>plant</strong><br />

molecular biology studies by Kijne and colleagues (Diaz et al. 1989), and more<br />

recently Hirsch and colleagues (van Rhijn et al. 1998; Hirsch 1999), have confirmed<br />

that the host-encoded lectins play a crucial role in microsymbiont<br />

recognition and host specificity in the Rhizobium-legume symbiosis, as originally<br />

predicted.<br />

2.7 Rhizobium Acidic Heteropolysaccharides<br />

Rhizobium leguminosarum bv. trifolii normally produces a profound true<br />

capsule that is revealed by ruthenium red staining and transmission electron


514<br />

Frank B. Dazzo<br />

microscopy. The bulk of this capsule consists of a large acidic heteropolysaccharide<br />

(Dazzo and Hubbell 1975). Bioassays scored by quantitative phase<br />

contrast microscopy indicate that oligosaccharide fragments produced by<br />

enzymatic depolymerization of this polysaccharide are biologically active in<br />

promoting root hair infectibility in white clover seedlings inoculated with R.<br />

leguminosarum bv. trifolii (Abe et al. 1984; Hollingsworth et al. 1984). The<br />

complete structures of the acidic heteropolysaccharides of several strains of<br />

R. leguminosarum bv. trifolii have been elucidated and shown to consist of<br />

repeated octasaccharide units of 5Glc:2GlcA:1Gal containing a tetrasaccharide<br />

backbone of 2Glc:2GlcA substituted with O-acetate and a tetrasaccharide<br />

sidechain of 3Glc:1Gal bearing pyruvyl substitutions on the terminal Gal and<br />

penultimate Glc, and a O-hydroxybutyrate substitution on the terminal Gal<br />

(Hollingsworth et al. 1988; Philip-Hollingsworth et al. 1989a). Trifoliin A<br />

binds selectively to this acidic heteropolysaccharide, and the symbiont-specificity<br />

in this protein–carbohydrate interaction involves recognition of the<br />

sites of linkage and stoichiometry of noncarbohydrate substitutions in the<br />

octasaccharide repeat unit (Abe et al. 1984; Hollingsworth et al. 1984, 1988;<br />

Philip-Hollingsworth et al. 1989b). Subsequent biochemical studies revealed<br />

host-range related structural features of R. leguminosarum bv. trifolii acidic<br />

heteropolysaccharides that distinguish these cell <strong>surface</strong> polymers and those<br />

of the closely related pea symbiont, R. leguminosarum bv. viciae, based on<br />

subtle differences in molar stoichiometry and positions of attachment of<br />

these noncarbohydrate substitutions (Philip-Hollingsworth et al. 1989a, b).<br />

Other studies have shown a link between rhizobial genes involved in determining<br />

the acidic heteropolysaccharide structures and the legume host-range<br />

in R. leguminosarum and Rhizobium sp. (Acacia; Philip-Hollingsworth et al.<br />

1989b; Lopez-Lara et al. 1993, 1995). This relationship is expressed in some,<br />

but not all genetic backgrounds of R. leguminosarum (Orgambide et al. 1992).<br />

Recently, we have presented a micrograph of a portion of an isolated molecule<br />

of the R. leguminosarum bv. trifolii acidic polysaccharide acquired using a<br />

field-emission scanning/transmission electron microscope at extremely high<br />

magnification (Dazzo and Wopereis 2000). Image analysis of the branches<br />

projecting perpendicular to the main polymer backbone in that micrograph<br />

indicate that they are within the same size range as the predicted 20±2<br />

angstrom length of the substituted tetrasaccharide side-chain. Molecular<br />

microscopy!<br />

A role of the capsular polysaccharide from R. leguminosarum bv. trifolii in<br />

symbiotic recognition was clearly shown by labeling this polymer with the<br />

fluorochrome FITC and documenting its direct interaction with white clover<br />

roots using epifluorescence microscopy (Dazzo and Brill 1977). Figure 6A<br />

illustrates the result, providing direct evidence for the existence and distribution<br />

of receptor sites on clover root hairs that specifically recognized the capsular<br />

polysaccharide of this rhizobial microsymbiont. Further studies using<br />

fluorescence microscopy indicated that these receptor sites are saturable,


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 515<br />

Fig. 6. Role of Rhizobium<br />

acidic heteropolysaccharide<br />

in symbiotic development<br />

with legumes. A<br />

Direct detection of symbiont-specific<br />

receptor<br />

sites for R. leguminosarum<br />

bv. trifolii acidic heteropolysaccharide<br />

on root<br />

hairs of white clover<br />

seedlings. B Quantitative<br />

microscopy of symbiotic<br />

phenotypes of an R. leguminosarum<br />

bv. trifolii<br />

ANU437 Exo – mutant relative<br />

to its Exo + wild-type<br />

ANU794 parent scored on<br />

the white clover host. The<br />

significant requirement of<br />

the bacterial acidic heteropolysaccharide<br />

in<br />

expression of its important<br />

Roa-3, Hac, and Inf symbiotic<br />

phenotypes is clearly<br />

indicated. Reprinted with<br />

permission from the<br />

American Society for<br />

Microbiology<br />

match the cellular distribution of trifoliin A on the root <strong>surface</strong>, and are<br />

specifically hapten-inhibitable, thus implicating an involvement of this root<br />

hair lectin in recognition of the rhizobial acidic heteropolysaccharide (Dazzo<br />

and Brill 1977; Dazzo et al. 1978).<br />

Further symbiotic roles of the acidic heteropolysaccharide from R. leguminosarum<br />

bv. trifolii in clover root nodulation were shown by detailed<br />

microscopy of the phenotypes exhibited by mutants blocked in its synthesis.<br />

A common symbiotic phenotype of “exo-minus” mutants of many fast-growing<br />

rhizobia is their defective ability to invade nodules on their respective<br />

host <strong>plant</strong> (Leigh et al. 1987; Lopez-Lara et al. 1993, 1995; Rolfe et al. 1996;<br />

Sanchez et al. 1997). Figure 6B summarizes the results of detailed, quantitative<br />

microscopical analysis of symbiotic phenotypes in exo-minus mutants of R.<br />

leguminosarum bv. trifolii scored on white clover seedling roots prior to nodule<br />

invasion (Rolfe et al. 1996). These quantitative microscopy results clearly<br />

indicate that the acidic heteropolysaccharide of R. leguminosarum bv. trifolii<br />

plays a crucial role in several early events of the infection process, including


516<br />

Frank B. Dazzo<br />

the rhizobial expression of the symbiont-specific (Roa-3) pattern of attachment<br />

to root hairs, the induction of markedly curled shepherd’s crooks at root<br />

hair tips (Hac), and the formation of successful infection threads in root hairs<br />

(Inf), but not the induction of moderate root hair deformations (Had) or root<br />

nodule primordia (Noi). Thus, the acidic heteropolysaccharide of R. leguminosarum<br />

bv. trifolii is a very important cell <strong>surface</strong> component needed to<br />

accomplish symbiont recognition, Roa-3, Hac, and Inf events crucial to primary<br />

host infection in the Rhizobium-clover symbiosis, as predicted (Dazzo<br />

and Hubbell 1975; Dazzo and Brill 1977, 1979; Dazzo et al. 1984; Sherwood et<br />

al. 1984; Philip-Hollingsworth et al. 1989a, b; Orgambide et al. 1992; Rolfe et al.<br />

1996). In concurrence with our earlier findings using R. leguminosarum bv.<br />

trifolii and white clover, detailed microscopy has more recently revealed the<br />

importance and essential requirement of extracellular acidic heteropolysaccharide<br />

from wild-type Rhizobium leguminosarum bv. viciae and Sinorhizobium<br />

meliloti in successful root hair infection of their corresponding hosts,<br />

vetch and alfalfa (van Workum et al. 1998; Cheng and Walker 1998; Pellock et<br />

al. 2000).<br />

2.8 Rhizobium Lipopolysaccharides<br />

The lipopolysaccharide (LPS) is another cell <strong>surface</strong> component of R. leguminosarum<br />

bv. trifolii that was predicted to play a role in symbiotic infection<br />

when it was found to bind trifoliin A and contain the glycosyl component<br />

quinovosamine (2-amino-2,6-dideoxyglucose) in its structure, which turned<br />

out to be a potent saccharide hapten inhibitor of trifoliin A-Rhizobium polysaccharide<br />

interactions (Dazzo and Brill 1979; Hrabak et al. 1981; Sherwood et<br />

al. 1984; Dazzo et al. 1991). Quantitative bioassays of root hair infections on<br />

white clover scored directly by phase contrast microscopy indicated a role of<br />

a transient, trifoliin A-binding glycoform (K90) of R. leguminosarum bv. trifolii<br />

LPS in activating the infection process (Dazzo et al. 1991). This infectionrelated<br />

biological activity significantly increased the frequency of successful<br />

infection threads that grew the entire length of the root hair and penetrated<br />

into the underlying cortical cells (Dazzo et al. 1991). Further studies using<br />

immunofluorescence and immunoelectron microscopy revealed the direct<br />

interaction between this bioactive LPS glycoform and white clover root hairs<br />

(Dazzo et al. 1991), including its localized binding to root hair tips where trifoliin<br />

A accumulates (Fig. 7A, B), and its uptake and internalization within the<br />

root hair cell (Fig. 7C). Real-time video microscopy and quantitative image<br />

analysis revealed that this specific interaction of the trifoliin A-binding glycoform<br />

of R. leguminosarum bv. trifolii LPS and white clover root hairs induced<br />

rapid changes in cytoplasmic streaming indicative of altered cytoskeleton<br />

activity, and 2-D gel electrophoresis revealed changes in levels of several specific<br />

root hair proteins made in response to LPS exposure (Dazzo et al. 1991).


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 517<br />

Fig. 7. Direct interaction of Rhizobium lipopolysaccharide with host root hairs.Adsorption<br />

of the trifoliin A-binding glycoform of LPS from R. leguminosarum bv. trifolii to the<br />

tips of white clover root hairs, and their internalization of this bioactive Rhizobium signal<br />

molecule are shown by immunofluorescence microscopy (A), conventional transmission<br />

electron microscopy (B), and immunoelectron microscopy (C). Scale bar A<br />

10 mm, B, C 3 mm. Reprinted with permission from the American Society for Microbiology


518<br />

Frank B. Dazzo<br />

In contrast, quantitative microscopy revealed that similar treatment of<br />

white clover roots with LPS from heterologous wild-type rhizobia (e.g., R.<br />

leguminosarum bv. viciae or S. meliloti) resulted in very incompatible root<br />

hair responses (Dazzo et al. 1991). These included a reduction in frequency of<br />

successful infections made by wild-type R. leguminosarum bv. trifolii, a corresponding<br />

increase in proportion of aborted infections accompanied by accumulation<br />

of intensely autofluorescent material at the arrested infection thread<br />

within the root hairs, and the suppression in levels of some of the newly synthesized<br />

root hair proteins plus elevation in levels of other specific root hair<br />

proteins (Dazzo et al. 1991). These results indicate that Rhizobium LPS is a<br />

potent signal molecule that rapidly communicates with host root hairs before<br />

bacterial penetration, triggering signal transduction of various molecular<br />

and physiological changes in these host cells that modulate infection thread<br />

development and compatibility/incompatibility events during primary host<br />

infection (Dazzo et al. 1991).<br />

2.9 Chitolipooligosaccharide Nod Factors<br />

Microscopy has played a major role in showing that chitolipooligosaccharides<br />

(CLOS), first described by Lerouge et al. in S. meliloti (Lerouge et al.<br />

1990), are one group of several different types of Nod factor molecules made<br />

by R. leguminosarum bv. trifolii capable of inducing Had and Ccd/Noi on<br />

white clover roots (Hollingsworth et al. 1989; Philip-Hollingsworth et al.<br />

1991, 1997; Orgambide et al. 1994, 1995, 1996; Dazzo et al. 1996a; Dazzo et al.<br />

1996b; ). Consistent with their amphiphilic physicochemistry, CLOSs of true<br />

wild type (i.e., not genetically manipulated) R. leguminosarum bv. trifolii<br />

accumulate three log cycles higher in their cellular membranes rather than<br />

in the extracellular milieu, and comprise a diverse family of at least 23 different<br />

types of CLOS that vary in O-acetyl and N-fattyacyl substitution, and<br />

in degree of oligomerization (Orgambide et al. 1995; Philip-Hollingsworth et<br />

al. 1995).<br />

Because these wild-type Nod factors are primarily associated with membranes<br />

rather than secreted extracellularly (contrary to dogma), it was important<br />

to establish if they represent the symbiotically relevant forms. Quantitative<br />

microscopy bioassays on axenic seedlings showed that this was definitely<br />

the case. The family of wild-type membrane CLOSs from R. leguminosarum<br />

bv. trifolii was fully active in its ability to induce Had, Ccd and Noi in white<br />

clover roots at subnanomolar concentrations (Orgambide et al. 1996). Furthermore,<br />

these symbiotic activities of R. leguminosarum bv. trifolii membrane<br />

CLOSs were host-specific in that they elicited no mitogenic Ccd or Noi<br />

activity in hairy vetch or alfalfa roots (heterologous legumes of different<br />

cross-inoculation groups), no Had in alfalfa at any concentration tested, and<br />

only elicited a weak Had response in hairy vetch requiring a 10 4 -fold higher


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 519<br />

threshold concentration than in the homologous host white clover (Orgambide<br />

et al. 1996).<br />

We combined organic chemical synthesis and quantitative microscopy<br />

approaches to dissect the molecular structural features of wild-type CLOS<br />

molecules required for their Had and Ccd/Noi symbiotic activities. A variety<br />

of small analog molecules bearing various motifs of CLOS glycolipids were<br />

chemically synthesized and bioassayed on axenic legume <strong>plant</strong>s (Philip-<br />

Hollingsworth et al. 1997). The results of this study were straightforward and<br />

very informative. Quantitative brightfield microscopy indicated that<br />

nanomolar concentrations of a single glucosamine residue bearing a longchain<br />

fatty N-acyl substitution were required and sufficient to induce Had<br />

and Ccd/Noi activity on both white clover and alfalfa, without any structural<br />

requirements for sulfation, O-acetylation, oligomerization of the glucosamine<br />

backbone, or unsaturation of the N-acyl fatty acid moiety of<br />

CLOSs (Philip-Hollingsworth et al. 1997). Further molecular dissection of<br />

the polar head group (e.g., removal of the C5 and C6 groups from the pyranose<br />

ring) rendered the amphiphilic CLOS analog inactive in these Had and<br />

Ccd/Noi bioassays (Philip-Hollingsworth et al. 1997). Contrary to “dogma”,<br />

these studies on the molecular determinants of CLOS action showed that the<br />

minimal portion of the native CLOS molecule that is both essential and sufficient<br />

for these symbiotic activities resides simply at the nonreducing glucosamine<br />

terminus substituted with an N-acylated long-chain fatty acid, and<br />

the remaining variations in components of the CLOS molecule leading to<br />

their native diverse family restrict which host (white clover or alfalfa) will<br />

respond to them rather than serve as required, positive effectors of their Had<br />

and Noi bioactivities per se (Philip-Hollingsworth et al. 1997). These key<br />

results which show that N-fatty acyl polyunsaturation and sulfation (for<br />

alfalfa) are not essential components of the minimal active structural component<br />

of CLOS for Ccd/Noi in legumes have been independently confirmed<br />

(Vernoud et al. 1999; Diaz et al. 2000). Consistent with these findings, other<br />

related studies show that perception of NodRm CLOS factors by membrane<br />

fractions of alfalfa have no significant structural requirement for N-fatty<br />

acyl polyunsaturation nor sulfation (Bono et al. 1995). Collectively, these significant<br />

findings have profound impact on the validity of models that assign<br />

the physiological location of CLOS in rhizobia, as well as their structural<br />

requirements for perception and symbiotic bioactivities in legume hosts like<br />

white clover, alfalfa, and vetch.<br />

In this same study (Philip-Hollingsworth et al. 1997), we developed various<br />

fluorescent molecular probes to investigate the in vivo fate and uptake of<br />

bioactive CLOS molecules into living root cells of intact white clover<br />

seedlings. By chemically labeling the reducing N-acetylglucosamine terminus<br />

of wild-type R. leguminosarum bv. trifolii CLOSs with NBD fluorochrome, we<br />

were able to produce a family of fluorescent NBD-CLOS derivatives with minimal<br />

molecular perturbation that retained their Had and Ccd/Noi inducing


520<br />

Frank B. Dazzo<br />

biological activities on white clover roots (Philip-Hollingsworth et al. 1997).<br />

This approach is far superior to conjugation of CLOSs with certain alternative<br />

fluorochromes, e.g., biodipi, whose relatively large and hydrophobic molecular<br />

structure could significantly perturb the physiological bioactivity of the<br />

CLOS molecules. This NBD-CLOS molecular probe was applied to axenic<br />

seedling roots under microbiologically controlled conditions. At various time<br />

points thereafter, the specimens were rinsed free of unbound conjugate and<br />

examined in vivo by laser scanning confocal microscopy, with results<br />

acquired in real time at subcellular resolution (Philip-Hollingsworth et al.<br />

1997). Figure 8A–H illustrates the key in vivo results of these studies, providing<br />

direct microscopical evidence that the NBD-CLOSs made by wild-type R.<br />

leguminosarum bv. trifolii interact rapidly with clover root hairs, traverse<br />

their cell walls, absorb to their cell membrane, and within minutes are then<br />

internalized within these living cells, where they migrate to the base of the<br />

root hairs and translocate to underlying cortical cells in a discrete region of<br />

the root. Quantitative fluorescence microscopy indicated that NBD-CLOSs<br />

from wild-type R. leguminosarum bv. trifolii were internalized by a significantly<br />

higher proportion of root hairs from the host legume white clover than<br />

from the nonhost legume alfalfa (Philip-Hollingsworth et al. 1997). As predicted,<br />

the structural requirements for internalization of NBD-CLOS analogs<br />

in living root hairs matched those required for Had and Noi bioactivities of<br />

CLOSs in white clover and alfalfa as described above. In contrast, the fluorescent<br />

analog NBD-chitotriose (without a linked lipid) was not taken up by living<br />

clover root hairs or cortical cells, indicating that in vivo internalization of<br />

Fig. 8. Laser scanning confocal microscopy of the direct, dynamic interaction of chitolipooligosaccharides<br />

(CLOSs) from wild-type R. leguminosarum bv. trifolii ANU843<br />

with living cells of white clover roots. Purified CLOSs were conjugated with the fluorochrome<br />

NBD to produce a fluorescent molecular probe with minimal molecular perturbation<br />

that retained Had and Noi bioactivities on white clover roots. A When applied<br />

to roots, these labeled Nod factors rapidly adsorbed to the root hairs. Closer examination<br />

of a time-series sequence of images showed that the NBD-tagged CLOSs adsorbed<br />

to the root hair cell membrane, and then within minutes were internalized within these<br />

epidermal cells (B–F), some migrating to the base of the root hair cell (B–D) and others<br />

remaining on the cell membrane or inside the root hair nucleus (E, F). Within 30 min,<br />

some NBD-CLOSs were translocated to a discrete region of the underlying root cortex<br />

and internalized within selected cortical cells (G, H). Arrowheads in the paired micrographs<br />

of (E, F) point to the root hair nucleus that internalized some labeled CLOSs. The<br />

NBD-CLOSs of ANU843 were internalized by a significantly higher proportion of the<br />

root hairs on white clover than alfalfa roots. Further studies using synthetic CLOS<br />

analogs and axenic seedling bioassays evaluated by these microscopy techniques established<br />

the minimal structural features of these Nod factor molecules that are required<br />

and sufficient for uptake and Had/Noi-inducing activities on both white clover and<br />

alfalfa roots. Scale bar A 50 mm, B–D 15 mm, E, F 10 mm, H 100 mm. Reprinted with permission<br />

from Lipid Research, Inc.


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 521


522<br />

Frank B. Dazzo<br />

NBD-labeled CLOSs and CLOS analogs by these host cells require the long<br />

chain N-acyl fatty acid moiety, but it does not have to be polyunsaturated.<br />

These results also indicated that the observed fluorescence was not due to autofluorescence<br />

of root cells, nor to uptake of a cleavage product of NBD-CLOSs<br />

degraded by <strong>plant</strong> chitinases, and that the root epidermis of seedlings used in<br />

these experiments had no open cracks through which NBD-CLOSs could passively<br />

diffuse into the root.Another interesting finding was that the interior of<br />

the clover root hair nucleus was a specific target reached rapidly by some of<br />

the internalized fluorescent NBD-CLOSs applied to white clover roots, as<br />

illustrated in the paired images of a root hair using phase contrast light<br />

microscopy (Fig. 8E) and the corresponding, longitudinal epifluorescence<br />

optisection obtained by laser scanning confocal microscopy that samples<br />

through the fluorescent nucleoplasm of its nucleus (Fig. 8F). These findings<br />

(Philip-Hollingsworth et al. 1997) impact profoundly on our understanding of<br />

the very early fate of rhizobial CLOS molecules before primary infection and<br />

nodule induction, and on the nature, location, and molecular specificity of<br />

putative host receptor sites for these Nod factors in the host legume root.<br />

2.10 Epidermal Pit Erosions<br />

Recently, we used various types of microscopy and enzymology to further<br />

clarify how rhizobia modify root epidermal cell walls in order to shed new<br />

light on the mechanism of primary host infection in the Rhizobium-legume<br />

symbiosis (Mateos et al. 2001). A thorough scanning electron microscope<br />

(SEM) examination of the epidermal <strong>surface</strong> of white clover roots inoculated<br />

with R. leguminosarum bv. trifolii revealed a nonuniform distribution of<br />

eroded pits that follow the contour of the Rhizobium cell (Fig. 9A). Their localized<br />

structure suggested that rhizobia have cell-bound wall-degrading<br />

enzymes, and indeed, follow-up biochemical studies confirmed that rhizobia<br />

produce multiple cell-bound isozymes of cellulase and polygalacturonase<br />

(Mateos et al. 1992, 1996; Jiminez-Zurdo et al. 1996). Quantitative SEM indi-<br />

Fig. 9. Epidermal eroded pits induced by Rhizobium leguminosarum bv. trifolii on white<br />

clover roots. A Scanning electron micrograph of the root epidermis pitted by attached<br />

cells of rhizobia (arrows). B Transmission electron micrograph showing ultrastructural<br />

details of the pitted interface between an attached cell of rhizobia and the clover epidermal<br />

root cell wall. Note that the localized erosion is restricted to amorphous regions and<br />

not the ordered microfibrillar wall layer (arrows). C (control), E, G Phase contrast<br />

microscopy and D, F Nomarski interference contrast microscopy of the Hot (Hole on the<br />

tip) reaction representing the complete erosion of a transmuro hole made by purified<br />

cellulase from R. leguminosarum bv. trifolii through the noncrystalline wall at root hair<br />

tips (arrows). Reprinted with permission from the Canadian National Research Council


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 523


524<br />

Frank B. Dazzo<br />

cated that the spatial density of these rhizobia-associated eroded pits was significantly<br />

higher on the root epidermis of host rather than nonhost legume<br />

combinations, was inhibited by high nitrate supply, and was not induced by<br />

immobilized wild-type R. leguminosarum bv. trifolii chitolipooligosaccharide<br />

Nod factors reversibly adsorbed to latex beads. Transmission electron microscope<br />

(TEM) examination of these highly localized epidermal pits indicated<br />

that they were only partially eroded, i.e., only the outer amorphous region of<br />

the <strong>plant</strong> wall in direct contact with the bacterial cell was disrupted, whereas<br />

the underlying highly ordered portion(s) of the wall remained ultrastructurally<br />

intact (Fig. 9B). Further studies using phase contrast and polarized<br />

light microscopy indicated that (1) the structural integrity of clover root hair<br />

walls is dependent on wall polymers that are valid substrates for the purified<br />

cell-bound polysaccharide-degrading enzymes (e.g., C2 cellulase isozyme)<br />

from rhizobia (Fig. 9C–G); (2) the major site where these rhizobial cell-bound<br />

enzymes can completely erode through the root hair wall is highly localized at<br />

the isotropic, noncrystalline apex of the root hair tip (Fig. 9C–G), and (3) the<br />

degradability of clover root hair walls by these rhizobial polysaccharidedegrading<br />

enzymes is enhanced by modifications induced during growth in<br />

the presence of CLOS Nod factors from wild-type clover rhizobia. These<br />

results suggest that these eroded <strong>plant</strong> structures represent incomplete<br />

attempts of bacterial penetration that had only progressed through isotropic,<br />

noncrystalline layers of the <strong>plant</strong> cell wall, and that the rhizobial cell-bound<br />

glycanases and chitolipooligosaccharides participate in complementary roles<br />

that ultimately create the localized transmuro portal of entry for successful<br />

primary host infection (Munoz et al. 1998; Mateos et al. 2001).<br />

2.11 Elicitation of Root Hair Wall Peroxidase by Rhizobia<br />

Many investigators have proposed that successful infection of legumes by rhizobia<br />

may depend on the microsymbiont’s ability to escape, suppress, or avoid<br />

host defense responses that normally protect <strong>plant</strong>s against invasive microorganisms<br />

(Vance 1983; Djordjevic et al. 1987; Parniske et al. 1990, 1991). To test<br />

this hypothesis, we performed in situ enzyme cytochemistry at subcellular<br />

resolution using brightfield microscopy followed by in vitro enzyme assays to<br />

detect changes in activity of <strong>plant</strong> wall-bound peroxidase as an indication of<br />

a localized host defense response following inoculation of white clover and<br />

pea roots with compatible and incompatible combinations of rhizobial symbionts<br />

(R. leguminosarum biovars trifolii and viciae; Salzwedel and Dazzo<br />

1993). For compatible combinations, elevated peroxidase activity was initially<br />

delayed, but subsequently located precisely at infection-related sites: the center<br />

of markedly deformed shepherd’s crooks and at penetration sites of incipient<br />

infection thread formation, but not elsewhere on the infected root hairs<br />

including the intracellular infection thread itself. In contrast, the incompati-


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 525<br />

ble combinations rapidly elicited elevated <strong>plant</strong> peroxidase activity over<br />

larger areas of the uninfected root hairs corresponding to their entire irregularly<br />

deformed root hair tips. Studies using various pSym nod mutant strains<br />

(provided by B. Rolfe, Australian National University) indicated a role of<br />

extracellular factors and the host-specific nodulation genes nodEL in this<br />

Rhizobium-controlled modulation of root hair peroxidase activity (Salzwedel<br />

and Dazzo 1993). Thus, active suppression of host defense responses by compatible<br />

rhizobia prior to primary host infection are implicated by these studies.<br />

Induction of white clover root peroxidase by compatible and incompatible<br />

rhizobial symbionts has been independently confirmed by differential display<br />

<strong>plant</strong> molecular biology techniques (Crockard et al. 1999).<br />

2.12 In Situ Gene Expression<br />

Reporter strains of Rhizobium with gene fusions encoding b-galactosidase, bglucuronidase,<br />

and green fluorescent protein are expanding the contribution<br />

of microscopy in unraveling many mysteries of the fascinating infection<br />

process in the Rhizobium-legume symbiosis. A common application of this<br />

technology is the use of reporter strains to locate primary host infections<br />

since they occur infrequently. Another informative application is the use of<br />

merodiploid reporter strains to locate at single cell resolution where, and at<br />

what stage of infection do rhizobia express symbiotic genes in situ with minimal<br />

risk of disturbing their symbiotic phenotypes. This application was used<br />

in quantitative microscopy studies that documented the in situ expression of<br />

pSym nodA by R. leguminosarum bv. trifolii cells during their early interaction<br />

with the root <strong>surface</strong> of the white clover host, especially those bacterial<br />

cells that have been clumped together on white clover root hair tips by trifoliin<br />

A during the first few hours of phase 1 attachment (Dazzo et al. 1988).<br />

Several methods have been used to detect expression of host symbiotic<br />

genes during early interactions of rhizobia with their legume host. One<br />

approach has been to use darkfield microscopy with in situ hybridization of<br />

DNA probes to specific mRNAs in <strong>plant</strong> tissue to locate which legume root<br />

cells express early nodulins [“Enods”] in response to inoculation with rhizobia<br />

(McKhann and Hirsch 1993). Such in situ localization studies can be<br />

enhanced even further if accompanied by immunofluorescence microscopy at<br />

single cell resolution (Dazzo and Wright 1996; McDermott and Dazzo 2002),<br />

to determine if the antigenic gene product of interest remains with the same<br />

cell(s) expressing the gene and/or is redistributed to other cells in the tissue.<br />

A second method to examine the cellular location and timing of expression of<br />

symbiotically important host genes induced by rhizobia makes use of<br />

chimeric fusions of the Gus-reporter gene in transgenic <strong>plant</strong>s. For instance,<br />

recent microscopical examination of transgenic alfalfa <strong>plant</strong>s stained for GUS<br />

activity has shown that nod mutants of S. meliloti although blocked in ability


526<br />

Frank B. Dazzo<br />

to introduce polyunsaturation of the N-acyl fatty acid moiety, in O-acetylation<br />

and in sulfation of CLOS Nod factors, are still capable of inducing<br />

ENOD20 (a marker of cortical cell activation) and (most importantly) eliciting<br />

cortical cell divisions in this legume host (Vernoud et al. 1999). This result<br />

is fully consistent with our studies described earlier that defined the minimal<br />

structural requirements for uptake and bioactivity of rhizobial CLOS analogs<br />

in legume roots, including induction of alfalfa and white clover cortical cell<br />

divisions (Philip-Hollingsworth et al. 1997), contrary to the dogma indicating<br />

that those structural features of CLOS dictate host specificity in the S.<br />

meliloti-alfalfa symbiosis. Finally, a third powerful approach to detect target<br />

mRNA is based on staining tissue sections for in situ PCR-amplified antisense<br />

riboprobes. This approach has recently been used to detect a novel<br />

Enod [dd23b] in white clover roots induced within 6 h after inoculation with<br />

wild-type R. leguminosarum bv. trifolii or the corresponding purified wildtype<br />

CLOS (Crockard et al. 2002).<br />

3 Quantitation of Symbiotic Interactions Between<br />

Rhizobium and Legumes by Image Analysis<br />

The value of quantitative microscopy for <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> can be<br />

enhanced even further when coupled with computer-assisted digital image<br />

analysis (Hollingsworth et al. 1989; Orgambide et al. 1996). This fast-growing<br />

technology utilizes the digital computer to derive numerical information<br />

regarding selected image features. Although image analysis technology cannot<br />

add anything that is not already present, its ability to extract the maximum<br />

amount of data from the image, as well as to quickly store, retrieve, and<br />

electronically transmit that data makes it an invaluable research tool for the<br />

microscopist. Computer-assisted microscopy has been used to enhance developmental<br />

morphology studies of the Rhizobium-legume symbiosis since 1989<br />

(Dazzo and Petersen 1989). Here, I highlight a few examples of new information<br />

on the Rhizobium-legume symbiosis derived from microscopical studies<br />

utilizing digital image analysis, and later illustrate how we have opened new<br />

ground in <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> by development and implementation of<br />

innovative image analysis software tailored to studies of in situ microbial<br />

ecology.<br />

3.1 Definitive Elucidation of the Nature of Rhizobium Extracellular<br />

Microfibrils<br />

The extracellular microfibrils made by R. leguminosarum bv. trifolii in pure<br />

culture were isolated and shown by chemical analysis to consist of microcrystalline<br />

cellulose (Napoli et al. 1975a). However, the nature of the microfibrils


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 527<br />

associated with rhizobia that highlight the beginning of their Phase 2 firm<br />

adhesion to the legume root epidermis (Fig. 2F) was more difficult to define.<br />

The combined use of scanning electron microscopy, enzyme cytochemistry,<br />

and computer-assisted digital image analysis provided direct in situ evidence<br />

of the cellulosic nature of the extracellular microfibrils extending from R.<br />

leguminosarum bv. trifolii cells colonized on the white clover root epidermis<br />

(Mateos et al. 1995).<br />

3.2 Rhizobial Modulation of Root Hair Cytoplasmic Streaming<br />

Many studies have shown that rhizobia influence the cytoplasmic streaming<br />

of host root hairs (beginning with the classic microscopical studies of root<br />

hair infection by Fåhraeus (1957) and Nutman et al. (1973), but very few have<br />

gone the extra mile to quantitate the changes in velocity of this early host<br />

cytoskeletal event in vivo. We utilized high resolution, phase-contrast video<br />

microscopy and play-back digital image analysis in real time to establish that<br />

the velocity of cytoplasmic streaming within living root hairs of white clover<br />

is increased by 35 and 63 % soon after exposure to cells or isolated clover<br />

lectin-binding lipopolysaccharide of R. leguminosarum bv. trifolii, respectively<br />

(Dazzo and Petersen 1989; Dazzo et al. 1991).<br />

3.3 Motility of Rhizobia in the External Root Environment<br />

Rhizobium leguminosarum bv. trifolii is peritrichously flagellated. How fast<br />

does it swim in the external root environment of its host, white clover? By<br />

focusing the phase objective lens just below the coverslip in modified<br />

Fåhraeus slide cultures without the agar matrix, it is possible to record<br />

enough examples of long swimming runs of individual cells within the depth<br />

of focus to perform image analysis on track reconstructions of real-time<br />

video recorded images played back in slow motion. Quantitation of this activity<br />

by digital image analysis showed that R. leguminosarum bv. trifolii swims<br />

in this external clover root environment at an average velocity of 52 mm/s<br />

(around 40 times its cell length, compared to around 60 body lengths for E.<br />

coli under ideal testing conditions), and cells tethered by their lateral flagella<br />

to the underside of the coverslip rotate at a frequency of 5–6 Hz/s in this slide<br />

culture environment (Dazzo and Petersen 1989).<br />

When Fåhraeus slide cultures of white clover seedlings and R. leguminosarum<br />

bv. trifolii are prepared using 0.4 % agarose, two zones of bacterial<br />

chemotropic swarming can be visualized by darkfield illumination. Digital<br />

image analysis indicates that one of these bacterial chemotropic responses<br />

forms a hollow sphere whose center is at the root tip and an intercept of<br />

radius approximately 4 mm above the root tip. The second chemotropic


528<br />

Frank B. Dazzo<br />

response is less structured, but accumulates in a cylindrical zone surrounding<br />

the root 2–4 mm from the root tip. These microscopical observations suggest<br />

that Rhizobium responds chemotactically in situ to different, multiple chemical<br />

gradients in the external environment surrounding the clover root.<br />

3.4 Root Hair Alterations Affecting Their Dynamic Growth Extension<br />

and Primary Host Infection<br />

Quantitative microscopy has played a major role in analyzing the developmental<br />

morphology of white clover root hairs to elucidate the mechanisms of<br />

rhizobial CLOS action in modulating the growth dynamics and symbiont<br />

infectibility of these target host cells (Dazzo et al. 1996a).We performed timelapse<br />

video microscopy of axenic seedling roots treated with nanomolar concentrations<br />

of wild-type R. leguminosarum bv. trifolii CLOSs and grown geotropically<br />

under microbiologically controlled conditions, followed by a<br />

quantitative time-series image analysis of individual root hair growth in the<br />

acquired video-recorded images at 4-s resolution (Dazzo et al. 1996a). This<br />

analysis indicated that the earliest discernible root hair deformations occur<br />

within 2.12±0.65 h after application of the wild-type CLOS, and that the morphological<br />

basis of the dominant type of CLOS-induced Had is a short-range<br />

alteration in direction of polar extension growth of the root hair tip rather<br />

than distortion of an already elongated root hair wall, resulting in a redirection<br />

of tip growth that deviates from the medial axis of the root hair cylinder.<br />

Further studies of quantitative microscopy indicated that CLOS action<br />

extends the growing period of active root hair elongation for ~ 5.2 h beyond<br />

its normal duration without affecting the elongation rate per se (~19 mm/h),<br />

resulting in mature root hairs that are on average about 100 mm longer. This<br />

extended growth period predictably increases the duration in which the root<br />

hair’s “window of infectibility” remains open before cessation of growth. Consistent<br />

with this hypothesis, CLOS action was shown by polarized light<br />

microscopy to induce localized isotropic alterations in the otherwise<br />

anisotropic, ordered crystalline architecture of root hair walls and shown by<br />

phase contrast light microscopy to significantly increase the number of<br />

potential infection sites and promote their infectibility by wild-type R. leguminosarum<br />

bv. trifolii (Dazzo et al. 1996a). These studies gave new information<br />

on the mechanisms of CLOS action that participate in activating root hair<br />

infectibility in the Rhizobium-legume symbiosis.


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 529<br />

4 A Working Model for Very Early Stages of Root Hair<br />

Infection by Rhizobia<br />

These various studies that capitalize on the added dimension of quantitative<br />

microscopy at cellular and subcellular resolution (Abe et al. 1984; Dazzo et al.<br />

1982, 1991, 1996; Mateos et al. 1992, 1996, 2001; Salzwedel and Dazzo 1993;<br />

Rolfe et al. 1996; Sanchez et al. 1997) have led to a working model for primary<br />

infection of white clover root hairs by the N 2-fixing symbiont, R. leguminosarum<br />

bv. trifolii. This model includes a transient, rapid nodEL-dependent<br />

suppression of host peroxidase activity during the initial period in which root<br />

hair infectibility is activated by trifoliin A-binding CPS oligosaccharides and<br />

K90 LPS, and CLOS-induced growth extension and disruptions in crystalline<br />

architecture of the growing root hair wall. The infection-related pattern of<br />

rhizobial attachment allows for the short-range combined action of these<br />

bioactive molecules to result in an increased localized susceptibility of this<br />

host wall barrier to a highly controlled degradation by cell-bound rhizobial<br />

enzymes that eventually form a small, but complete transmuro erosion site<br />

that ultimately becomes the primary portal of bacterial entry while still<br />

enclosed within the center overlap of the root hair shepherd’s crook. An<br />

increased flurry of cytoplasmic streaming within the root hair stimulated by<br />

the rhizobial symbiont is proposed to facilitate the delivery of new host cell<br />

components involved in initiation and continued inward growth of the walled<br />

tubular infection thread, while simultaneously directing the traffic of internalized<br />

membrane-associated CLOS signal molecules to the root hair nucleus.<br />

Later, a localized host wound response at the site of incipient bacterial penetration<br />

elevates peroxidase activity that cross-links structural polymers of the<br />

eroded wall in order to avoid lysis of the root hair protoplast after bacterial<br />

entry and infection thread formation. In contrast, rapid elicitation of clover<br />

peroxidase activity in the incompatible combinations (rhizobia with heterologous<br />

nodEL) may represent a localized discriminating host defense response<br />

that rapidly increases cross-linking of wall polymers, thus making the primary<br />

host barrier of the root hair wall more resistant to bacterial penetration.<br />

This unifying model assigns the ability of Rhizobium to modulate the plasticity<br />

(i.e., the summation of softening and hardening processes) of the root hair<br />

wall as a major symbiotic event controlling successful host infection.<br />

5 Improvements in Specimen Preparation and Imaging<br />

Optics for Plant Rhizoplane Microbiology<br />

Residual rhizosphere soil remaining on <strong>plant</strong> roots after gentle washing significantly<br />

obscures the underlying rhizoplane microflora. We have addressed<br />

this major limitation in <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> using very young white<br />

clover seedlings (£2 days old) grown in a sandy loam soil. By empirically opti-


530<br />

Frank B. Dazzo<br />

mizing the gyrorotary angular velocity and duration of gentle washing of<br />

excavated roots of white clover in isotonic Fåhraeus medium, we have largely<br />

solved this technical problem to expose the underlying pioneer microflora<br />

that develops rapidly on root <strong>surface</strong>s of seedlings germinated in soil. The<br />

optimal conditions to solve this problem vary with the density and length of<br />

root hairs, hence the <strong>plant</strong> species used. Quantitative image analysis of the<br />

white clover seedling roots indicate that this optimized washing procedure<br />

uncovers the vast majority (≥80 %) of the rhizoplane <strong>surface</strong> for viewing<br />

microbes without fragmentation of the root hairs.<br />

A major limitation of conventional epifluorescence microscopy used to<br />

examine the natural rhizoplane microflora that develops on soil-grown roots<br />

stained with fluorochromes is the background of blurred fluorescence outside<br />

the plane of focus used to produce the image. In this case, useful morphological<br />

information can only be extracted from images of cells lacking a background<br />

of out-of-focus fluorescence. A significant development in microscopy<br />

is the use of laser scanning confocal microscopy (LSCM) combined<br />

with digital image processing techniques. The unique feature of LSCM is that<br />

it utilizes pinholes at the laser light source and at the detection of the object’s<br />

image. This optical design eliminates the stray and out-of-focus light that<br />

interferes with the formation of the object’s image (a major limitation of the<br />

conventional fluorescence microscope), thereby only allowing signals from<br />

the focused plane to be detected (McDermott and Dazzo 2002). This optical<br />

design also improves the resolution and contrast of microbial cells in natural<br />

environments by greatly diminishing objectionable background fluorescence<br />

arising from <strong>plant</strong> tissue, soil particles, or organic debris. Because light from<br />

outside the plane of focus is not included in image formation, the 2-D (x–y)<br />

image becomes an accurate optidigital thin section with a thickness<br />

approaching the theoretical 0.2-mm resolution of the light microscope. Also,<br />

by digitizing a sequential series of 2-D images while focusing through the<br />

specimen in the third (z) dimension, a 3-D computer-reconstructed digital<br />

image can be produced, rotated,‘resectioned’ in another plane, displayed, and<br />

quantitatively analyzed.<br />

Because LSCM imaging technology solves so many problems inherent in<br />

conventional fluorescence microscopy, it is receiving wide application for in<br />

situ studies of microbial ecology. The first LSCM examination of the general<br />

rhizoplane microflora in situ was done with acridine orange-stained roots of<br />

white clover seedlings grown in soil (Dazzo et al. 1993). This approach eliminated<br />

the major background fluorescence due to dye absorption into the<br />

root interior, which makes conventional epifluorescence microscopy impossible<br />

for this type of specimen. Subsequently, Schloter et al. (1993) demonstrated<br />

the usefulness of LSCM for immunofluorescence examination of<br />

Azospirillum on wheat roots. They used a dual laser system to produce the<br />

green autofluorescence of the root background upon which the distinctive<br />

red immunofluorescence of Azospirillum (probed with tetramethylrho-


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 531<br />

damine isothiocyanate-labeled monoclonal antibodies) could be easily seen.<br />

They also utilized the noninvasive optical sectioning ability of the confocal<br />

microscope to locate the Azospirillum cells within the root mucigel layer.<br />

More recently, we have used optical sectioning by LSCM to document the<br />

entry of neptunia-nodulating rhizobia into crevices at lateral root emergence<br />

of the aquatic legume Neptunia natans (Subba-Rao et al. 1995), and azorhizobia<br />

colonized on the root <strong>surface</strong> and within cortical cells of intact rice<br />

roots (Reddy et al. 1997).<br />

6 CMEIAS: A New Generation of Image Analysis Software for<br />

in Situ Studies of Microbial Ecology<br />

6.1 CMEIAS v. 1.27: Major Advancements in Bacterial Morphotype<br />

Classification<br />

A major challenge in microbial ecology is to develop reliable methods of<br />

computer-assisted microscopy that can analyze digital images of microbial<br />

populations and complex microbial communities at single cell resolution,<br />

and compute useful ecological characteristics of their organization and<br />

structure in situ without cultivation. To address this challenge, we are developing<br />

customized semi-automated image analysis software capable of<br />

extracting the full information content in digital images of actively growing<br />

microbial populations and communities. This analytical tool, called CMEIAS<br />

(Center for Microbial Ecology Image Analysis System) consists of plug-in<br />

files for the free downloadable program UTHSCSA ImageTool (Wilcox et al.<br />

1997) operating in a personal computer running Windows NT 4.0/2000. The<br />

first release version of CMEIAS was developed primarily to perform morphotype<br />

classification of bacteria in segmented digital images of complex<br />

microbial communities (Liu et al. 2001). This CMEIAS version 1.27 uses pattern<br />

recognition algorithms optimized by us to recognize bacterial morphotypes<br />

with an overall classification accuracy of 97 %, and a sensitivity that<br />

can classify morphotypes present in the community at a frequency as low as<br />

~0.1 % (Liu et al. 2001). CMEIAS v. 1.27 can recognize 11 major morphotypes,<br />

including cocci, spirals, curved rods, U-shaped rods, regular straight<br />

rods, clubs, ellipsoids, prosthecates, unbranched filaments, rudimentary<br />

branched rods, and branched filaments, representing a complexity level of<br />

morphological diversity equivalent to 98 % of the genera described in the 9th<br />

edition of Bergey’s Manual of Determinative Bacteriology (Holt et al. 1994).<br />

An interactive edit feature is included in CMEIAS v. 1.27 to revise the output<br />

of automatic classification data if necessary (occurring at a 3 % error rate),<br />

and add up to five additional morphotypes not included in the automatic<br />

classification routine (Liu et al. 2001). Our first major application of CMEIAS<br />

v. 1.27 was to contribute data on dynamic changes in community structure,


532<br />

Frank B. Dazzo<br />

including its resistance, resilience, and ecological succession in a polyphasic<br />

taxonomy study of microbial community responses to nutrient perturbation,<br />

using complex anaerobic bioreactors as the model system (Fernandez et al.<br />

2000; Hashsham et al. 2000). CMEIAS v. 1.27 will soon be released for free<br />

Internet download at a website linked to the Michigan State University Center<br />

for Microbial Ecology (http://cme.msu.edu/cmeias).<br />

6.2 CMEIAS v. 3.0: Comprehensive Image Analysis of Microbial<br />

Communities<br />

A significantly upgraded version of CMEIAS is being developed with several<br />

new analytical modules designed to extract four ecologically relevant, in situ<br />

features of microbial communities in digital images: (1) morphotype classification<br />

and diversity,(2) microbial abundance for both filamentous and nonfilamentous<br />

morphotypes, (3) in situ studies of microbial phylogeny/autecology/metabolism,<br />

and (4) in situ spatial distribution analysis of microbial<br />

colonization on various <strong>surface</strong>s. Significant new features will include an<br />

advanced morphotype classifier that incorporates default size and shape<br />

dimensional borders that are taxonomically relevant and has user-defined<br />

flexibility to discriminate any customized level of morphological diversity;<br />

various computations of cell density, biovolume, biomass carbon, bio<strong>surface</strong><br />

area, and filamentous length; color recognition of foreground objects stained<br />

with fluorescent molecular probes; various measurement features of plot-less,<br />

plot-based,and georeferenced patterns of spatial distribution analysis; spreadsheet<br />

macros for automatic data preparation, sampling statistics and spatial<br />

statistics analyses; and automated image editing routines (Reddy et al.2002a,b;<br />

see http://lter.kbs.msu.edu/Meetings/2003_All_inv_Meeting/Abstracts.dazzo.<br />

htm). Data extracted from images by CMEIAS can be used in other advanced<br />

ecological statistics programs, e.g., EcoStat (Towner 1999), and GS+Geostatistics<br />

(Robertson 2002), to compute numerous other statistical indices that further<br />

characterize microbial community structure. Our vision is for CMEIAS to<br />

become an accurate, robust and user-friendly software tool that can analyze<br />

microbial communities without cultivation, thereby creating many new<br />

approaches to study microbial ecology in situ at spatial scales physiologically<br />

relevant to the individual microbes.<br />

To illustrate some of the awesome computational power of CMEIAS that<br />

can be applied to in situ studies of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>, examples of<br />

analyses have been performed on (1) the abundance and spatial distribution<br />

of Rhizobium leguminosarum bv. trifolii cells colonized on a white clover<br />

seedling root in gnotobiotic culture; (2) a comparison of the morphological<br />

diversity and distribution of abundance in natural microbial communities<br />

that colonize the phylloplane leaf <strong>surface</strong> of two different varieties of fieldgrown<br />

corn, and (3) the in situ spatial patterns of root colonization by the pio-


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 533<br />

neer microflora that develop on the white clover seedling rhizoplane during<br />

their first 2 days of growth in soil, and (4) the spatial scale of quorum sensing<br />

of signal molecules by rhizobacteria colonized on the root <strong>surface</strong>.<br />

6.3 CMEIAS v. 3.0: Plotless and Plot-Based Spatial Distribution Analysis<br />

of Root Colonization<br />

For this first example, CMEIAS image analysis was performed on a scanning<br />

electron micrograph of a region of the white clover seedling root <strong>surface</strong> colonized<br />

by cells of Rhizobium leguminosarum bv. trifolii wild-type strain<br />

ANU843 to extract many different types of quantitative data relevant to <strong>plant</strong><br />

<strong>surface</strong> <strong>microbiology</strong> (Fig. 10A). Figure 10B illustrates the frequency distribution<br />

of their first and second nearest neighbor distances (distance between<br />

object centroids), as two examples of their spatial distribution in a plot-less<br />

analysis. Table 1 lists 15 quantitative features relevant to <strong>plant</strong> <strong>surface</strong> micro-<br />

Fig. 10. Colonization<br />

of the white clover<br />

root <strong>surface</strong> by wildtype<br />

R. leguminosarum<br />

bv. trifolii ANU843 in<br />

gnotobiotic culture.<br />

A Scanning electron<br />

micrograph of a region<br />

of the root <strong>surface</strong> colonized<br />

by the bacteria.<br />

Bar scale 1 mm.<br />

B CMEIAS plot-less<br />

spatial distribution<br />

analysis of bacterial<br />

cells in (A) measured<br />

as the frequency distribution<br />

of each cell’s<br />

first and second nearest<br />

neighbor distance


534<br />

Frank B. Dazzo<br />

Table 1. Quantitative data relevant to <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong> extracted from<br />

Fig. 10A by CMEIAS image analysis<br />

Measurement feature Type of analysis Value<br />

Number of cells Microbial abundance 138<br />

Avg. cell biovolume (mm 3 ) Microbial abundance 0.17<br />

Avg. cell biomass C (fg) Microbial abundance 34.66<br />

Avg. cell bio<strong>surface</strong> area (mm 2 ) Microbial abundance 1.72<br />

Cumulative bacterial biovolume (mm 3 ) Microbial abundance 23.91<br />

Cumulative bacterial biomass C (fg) Microbial abundance 4783.18<br />

Cumulative bacterial bio<strong>surface</strong> area (mm 2 ) Microbial abundance 237.86<br />

Cumulative area covered by bacteria (mm 2 ) Microbial abundance 60.25<br />

Avg. first nearest neighbor distance (mm) Plotless spatial 0.84±0.30<br />

distribution<br />

Avg. second nearest neighbor distance (mm) Plotless spatial 1.10±0.32<br />

Average aggregation (cluster) index (mm –1 ) Plotless spatial 1.31±0.38<br />

distribution<br />

Holgate’s A value of spatial randomness Plotless spatial 0.622<br />

distribution =clumped<br />

Significance value of Holgate’s A (p) Plotless spatial 0.001<br />

distribution<br />

Spatial density of bacteria (cells/mm 2 ) Plot-based spatial 427,245<br />

distribution<br />

Microbial cover (%) Plot-based spatial 18.7<br />

distribution<br />

Uncovered root <strong>surface</strong> area (%) Plot-based spatial 81.3<br />

distribution<br />

biology that were extracted from this same image, eight features that measure<br />

microbial abundance, and seven (four plot-less plus three plot-based) features<br />

that measure their spatial distribution. The Aggregation (Cluster) Index is a<br />

plot-less spatial distribution measurement feature that we have introduced,<br />

equal to the inverse of the first nearest neighbor distance (Dazzo et al. 2003).<br />

The Holgate’s method for plot-less spatial analysis is a statistical test for spatial<br />

randomness requiring that n random points be selected and that the distance<br />

to the two nearest individuals be measured. This method computes Holgate’s<br />

A, a measure of aggregation. Values of A are 0.5 for randomly spaced<br />

populations, >0.5 for clumped populations, and 0.5, their spatial distribution is clumped, and the Z-test for spatial<br />

randomness is rejected at the statistically significant level of 99.9 %. Definitive<br />

quantitative spatial distribution data acquired by computer-assisted microscopy!


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 535<br />

6.4 CMEIAS v. 3.0: In Situ Analysis of Microbial Communities on Plant<br />

Phylloplanes<br />

In the second example, CMEIAS was used to perform an in situ image analysis<br />

of the microbial communities that developed on phylloplane <strong>surface</strong>s of<br />

two corn varieties grown under field conditions: one was a genetically modified<br />

corn genotype engineered to express the insecticide protein made by the<br />

bacterium Bacillus thuriengensis (BT-corn variety Pioneer 3573), and the second<br />

was a control corn (non-BT variety Pioneer 36N05) receiving no insecticide.<br />

Corn leaf disks (4 mm in diameter) were sampled in July, 1999 from<br />

mature field-grown <strong>plant</strong>s cultivated in an Long-Term Ecological Research<br />

[LTER] experimental site at the Michigan State University Kellogg Biological<br />

Station (KBS). Adjacent quadrats (n=26) of digital images were acquired by<br />

scanning electron microscopy at ¥1000 and at ¥100 to resolve the prokaryotic<br />

Fig. 11. Scanning electron<br />

micrographs of the phylloplane<br />

microflora developing<br />

on the leaf <strong>surface</strong> of<br />

field-grown corn. Images<br />

were acquired at 1000x (A)<br />

and 100x (B) to locate and<br />

analyze the prokaryotic<br />

(bacteria) and eukaryotic<br />

(fungi) microorganisms in<br />

the phylloplane community,<br />

respectively. Scale bar<br />

1 mm in A and 100 mm in B


536<br />

Frank B. Dazzo<br />

and eukaryotic components of the microbial communities, respectively, on<br />

the upper corn leaf <strong>surface</strong>s (Fig. 11A, B), and then analyzed by CMEIAS to<br />

characterize their community structures in situ. Figure 12 compares the morphological<br />

diversity of the prokaryotic microorganisms in these two communities,<br />

with data presented in a rank-order pareto plot of their richness and<br />

percent numerical abundance of operational morphological units (OMU utilizing<br />

both shape and size classification schemes), plus a table insert of their<br />

morphological diversity index (based on Shannon’s Diversity Index H’ using<br />

nearly equivalent community sample sizes and OMUs rather than species), J<br />

evenness distribution of OMUs, and a proportional similarity index that is<br />

weighted according to OMU dominance. The latter three indices are derived<br />

from computations of the CMEIAS data in EcoStat. These results indicate that<br />

the prokaryotic component of the phylloplane communities developed on<br />

these two corn varieties had quite similar values of OMU richness, morphological<br />

diversity indices, and J evenness in distribution of OMUs, but deeper<br />

CMEIAS data mining indicate that they have a proportional similarity index<br />

in prokaryotic morphological diversity of only 64.2 % due to major differ-<br />

Fig. 12. Rank-abundance diversity plots of CMEIAS morphotype classification data<br />

among prokaryotic microorganisms that colonize the phylloplane <strong>surface</strong> of control<br />

(non-BT) corn and BT-corn expressing the bacterial insecticide. The insert table reports<br />

the similarities and differences in indices of community structure based on morphological<br />

analyses using CMEIAS


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 537<br />

ences in relative abundance of various sized regular rods, cocci, and ellipsoid<br />

OMUs (Fig. 12). One should be able to readily appreciate from this example<br />

how CMEIAS can augment other methods of polyphasic taxonomy (e.g., 16S<br />

rDNA sequence analysis) to analyze and quantitatively compare complex<br />

microbial communities in situ without cultivation.<br />

CMEIAS offers several different algorithms to compute microbial biovolume,<br />

with the most accurate overall being adaptive to shape, i.e., CMEIAS first<br />

classifies the cell shape and then applies the most appropriate formula to<br />

compute its volume based on that particular shape. Figure 13 summarizes the<br />

CMEIAS analysis of total abundance and relative distribution of biovolume<br />

among the various prokaryotic morphotypes in these two different corn phylloplane<br />

communities. The results clearly indicate a significantly greater abundance<br />

of prokaryotic biovolume per unit of phylloplane <strong>surface</strong> area for the<br />

Pioneer 36NO5 (control) variety than the Pioneer 3573 (BT-corn) variety<br />

(Fig. 13A), and substantial differences in relative distribution of prokaryotic<br />

biovolume for certain dominant morphotypes in these communities<br />

(Fig. 13B).<br />

Fig. 13. CMEIAS analysis<br />

of biovolume abundance<br />

in microbial communities<br />

developed on the phylloplane<br />

of field-grown control<br />

corn and BT-corn.<br />

Above Total standing crop<br />

of prokaryotic biovolume.<br />

Below Distribution of<br />

community biovolume<br />

among different prokaryotic<br />

morphotypes


538<br />

Frank B. Dazzo<br />

Table 2. In situ plot-based spatial distribution analysis of corn phyllosphere prokaryotic<br />

microbial communities<br />

Parameter Control BT corn Interpretation<br />

corn (Pioneer<br />

(Pioneer 3573)<br />

36N05)<br />

Spatial density (cells/mm 2 ) 214,615 172,692 Higher on control corn<br />

Morista dispersion value 1.3086 1.7805 Clumped distribution<br />

Variance/mean ratio 7.7346 13.6624 Clumped distribution<br />

Negative binomial K distribution 1.9238 0.6779 Clumped distribution<br />

Lloyd’s patchiness value 1.3144 1.7931 Clumped distribution<br />

Nonfilamentous microbial cover (%) 2.2 0.4 Higher on control corn<br />

The spatial distribution of prokaryotic microorganisms on the phylloplane<br />

<strong>surface</strong> of these two corn varieties was compared by analyzing several CMEIAS<br />

in situ plot-based measurement features on a sample set of 104 quadrat images<br />

(52 quadrats each). The mean values of their spatial density (cells/unit of <strong>surface</strong><br />

area) and percent nonfilamentous microbial cover indicated a significantly<br />

higher level of bacterial colonization on the phylloplane of the Pioneer<br />

36N05 (control corn) variety (Table 2). An ascending sort plot of the entire<br />

range of spatial density for each image quadrat provided further insight into<br />

the basis for this difference in spatial distribution,with clear indication that the<br />

overall density of bacteria on the BT-corn phylloplane was lower because that<br />

habitat contained more image quadrats with no bacterial cover (Fig. 14).<br />

Table 2 lists several other computations that define the patterns of spatial<br />

distribution for microbes that colonize these <strong>plant</strong> leaf <strong>surface</strong>s, all derived<br />

from in situ plot-based data extracted from image quadrats by CMEIAS and<br />

computed in EcoStat. The Morista Index measures the degree of dispersion,<br />

with values 1 for a clumped pattern. The variance/mean ratio from the<br />

observed pattern of frequencies (proportion of quadrats that contain organisms)<br />

to those predicted by a Poisson distribution is approximately 1 for randomly<br />

spaced populations, significantly >1 for clumped spacing, and


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 539<br />

Fig. 14. CMEIAS spatial<br />

density analysis of<br />

prokaryotic microorganisms<br />

colonized on the<br />

phylloplane of fieldgrown<br />

control corn<br />

(36N05) and BT-corn<br />

(3573)<br />

Table 3. Spatial abundance of filamentous fungi on the phylloplane of field-grown corn.<br />

Values reported are per mm 2 phylloplane <strong>surface</strong> area<br />

Spatial abundance parameter Control (36N05) corn BT (3573) corn<br />

Filamentous hyphae cover (%) 12.7 6.4<br />

Cumulative hyphal length (mm) 1737 803<br />

Hyphal bio<strong>surface</strong> area (mm 2 ) 4244 2035<br />

Hyphal biovolume (mm 3 ) 816 439<br />

Hyphal biomass carbon (fg) 163 88<br />

Table 3 summarizes the CMEIAS-based analyses of spatial abundance for<br />

the filamentous fungi on these corn phylloplanes. CMEIAS recognizes filamentous<br />

microorganisms in digital images based on their shape characteristic<br />

of a length/width ratio >16 without wave form periodicity, and classifies<br />

unbranched and branched filaments separately based on whether they have<br />

more than two cell poles (Liu et al. 2001).All five CMEIAS measurement parameters<br />

indicated an approximate 99 % higher spatial abundance of filamentous<br />

fungi biomass on the Pioneer 36N05 control corn variety.<br />

These results illustrated in this second example indicate that CMEIAS performs<br />

admirably in the in situ analysis of phylloplane microbial communities.<br />

One could definitively conclude from the results that different microbial communities<br />

developed on the phylloplane sampled from field-grown Pioneer<br />

36N05 and Pioneer 3573 varieties of corn, but more thorough studies would<br />

be necessary before reaching any firm conclusion regarding the possible


540<br />

Frank B. Dazzo<br />

involvement of the BT insecticide itself in influencing how these microbial<br />

communities developed to these different states.<br />

6.5 CMEIAS v. 3.0: In Situ Geostatistical Analysis of Root Colonization<br />

by Pioneer Rhizobacteria<br />

In the third example, CMEIAS was used to analyze the pattern of spatial distribution<br />

for the pioneer rhizobacterial community that first colonizes<br />

seedling roots grown in soil. For this study, Dutch white clover seeds were<br />

<strong>plant</strong>ed in a wetted sandy loam soil sampled at the KBS-LTER field site.<br />

Seedling roots were harvested after 2 days of germination, then gently washed<br />

free of rhizosphere soil by optimized gyrorotary rotation in Fåhraeus<br />

medium, stained briefly with a 1:10,000 aqueous solution of acridine orange,<br />

rinsed in 1 % Na pyrophosphate, and mounted in Vectashield photobleaching<br />

retardant reagent. Fluorescent micrographs of the natural pioneer rhizobacterial<br />

communities that developed on the clover rhizoplane were acquired as a<br />

series of optisection, grayscale images georeferenced to the root tip landmark<br />

using laser scanning confocal microscopy with the 63x oil immersion objective<br />

and direct through-the-ocular confocal viewing. These digital images<br />

were segmented and used to produce a large continuous montage in Adobe<br />

Photoshop. The montage images were analyzed by CMEIAS to locate the x, y<br />

Cartesian coordinates of each individual microbial cell on the rhizoplane and<br />

compute its Cluster index (inverse of first nearest neighbor distance) in situ as<br />

the z variate. These CMEIAS data were then analyzed by the spatial geostatistics<br />

modeling techniques of semivariogram autocorrelation and kriging<br />

analyses (Murray 2002) using GS+ software (Robertson 2002).<br />

The variogram of Fig. 15 is the first of its kind in showing that an isotropic<br />

exponential model best fits the semivariance autocorrelation data of spatial<br />

dependence for pioneer root colonization by microorganisms in soil. It further<br />

clearly indicates that there is a spatial dependence in the nearest neighbor<br />

distribution of rhizoplane colonization for microorganisms, with a spatial<br />

scale of influence up to a separation distance of ~52 mm. Thus, microbes separated<br />

from each other by distances up to this spatial limit do influence each<br />

other’s root colonization pattern. Such information is fundamentally new in<br />

that it provides a real world perspective of the in situ spatial scales that are<br />

truly relevant to microbial colonization of <strong>plant</strong> root <strong>surface</strong>s in soil. A first<br />

for in situ microbial ecology!


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 541<br />

Fig. 15. CMEIAS/GS+ analysis of spatial geostatistics (autocorrelation semivariogram)<br />

for rhizobacteria during pioneer colonization of white clover seedlings grown in soil.<br />

This graph indicates the highly significant, fundamentally new finding that pioneer colonization<br />

of seedling roots by bacteria in soil has an in situ spatial dependence over a<br />

spatial scale up to 52 mm<br />

6.6 CMEIAS v. 3.0: Quantitative Autecological Biogeography of the<br />

Rhizobium–Rice Association<br />

In the fourth example, CMEIAS is being used to study the biogeography of R.<br />

leguminosarum bv. trifolii strain E11, a <strong>plant</strong> growth-promoting endocolonizer<br />

of rice roots isolated in the Nile delta where rice and berseem clover<br />

have been rotated since antiquity (Yanni et al. 1997). We are using this strain<br />

in a model study designed to define the autecological biogeography of rhizobial<br />

PGPR endophytes of rice at two spatial scales, one relevant to the organisms<br />

(its colonization of rice roots), and second relevant to the rice farmer<br />

who would be using such strains as rice biofertilizer inoculants to enhance<br />

rice production with less dependence on chemical fertilizer N (Yanni et al.<br />

2001). Figure 16A is an SEM image quadrat of the rice root <strong>surface</strong> after gnotobiotic<br />

cultivation with strain E11. Note that the root hair cells above the<br />

plane of focus have obscured some of the root <strong>surface</strong>, and therefore the full<br />

distribution of bacteria in this sampled area cannot be examined directly.<br />

This problem in microbial biogeography is solved by a geostatistical analysis<br />

of the spatial distribution data acquired by CMEIAS using a kriging analysis<br />

to interpolate spatial dependence information on a continuous scale even in<br />

areas not sampled. Figure 16B shows the 2-D krig map that provides a statistically<br />

defendable interpolation of the spatial density of bacteria in a continuous<br />

mode, even in these areas obscured by the overlying root hairs (Fig. 16B).<br />

The power of CMEIAS geostatistical analysis!


542<br />

Frank B. Dazzo<br />

Fig. 16. Geostatistical analysis of the spatial distribution of a <strong>plant</strong> growth-promotive<br />

strain of Rhizobium leguminosarum bv. trifolii colonized on the rice root <strong>surface</strong>. A Typical<br />

colonization pattern as shown by scanning electron microscopy. Scale bar 10 mm. B<br />

2-D interpolation kriging map of the spatial density of bacterial cells in A


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 543<br />

6.7 CMEIAS v. 3.0: Spatial Scale Analysis of in Situ Quorum Sensing by<br />

Root-Colonizing Bacteria<br />

In the fifth example, CMEIAS is being used to extract information that sheds<br />

new light on the spatial scale at which cell-cell communication of quorum<br />

sensing occurs in situ during bacterial colonization of roots. This work is<br />

being done in a collaboration of the author with Prof. Anton Hartmann,<br />

Stephan Gantner and Christine Duerr in Germany. Confocal fluorescence<br />

images of roots are acquired to locate the positions of the red fluorescent protein<br />

reporter strain of bacteria that produces and secretes the acyl homoserine<br />

lactone quorum signal (source cells) and the green GFP-reporter strain of<br />

bacteria that cannot produce these signal molecules, but is nevertheless activated<br />

by them (sensor cells). The range of distances between each green sensor<br />

cell and its nearest red source cell neighbor then becomes a measure of the<br />

spatial scale at which the cell-to-cell communication of quorum-sensing signal<br />

molecules occurs in situ during root colonization. Early indications are<br />

that this spatial scale is close to the same range found for spatial dependence<br />

in root colonization as described in Fig. 15 above. Figure 17 further illustrates<br />

Fig. 17. CMEIAS/GS+<br />

spatial geostatistical<br />

analysis of in situ<br />

quorum sensing<br />

among neighboring<br />

bacteria colonizing<br />

the root <strong>surface</strong>. A<br />

Dot map indicating<br />

the location of bacteria<br />

that provide the<br />

source of the extracellular<br />

quorum signal<br />

molecule (N-acyl<br />

homoserine lactone).<br />

Scale bar 10 mm. B 2-<br />

D interpolation kriging<br />

map of the predicted<br />

gradients of<br />

the quorum sensing<br />

molecule in situ on<br />

the root <strong>surface</strong> based<br />

on the localized cluster<br />

indices of colonized<br />

bacteria


544<br />

Frank B. Dazzo<br />

the power of geostatistical kriging as a spatial modeling technique that can<br />

provide a statistically defendable graphical display of the predicted gradients<br />

of quorum sensing signals that would diffuse from aggregates of “source cell”<br />

bacteria colonized on the root <strong>surface</strong>. Figure 17A shows the sample point<br />

location of signal source bacteria in an image quadrat and Fig. 17B is a 2-D<br />

kriging map of their cluster index on a continuous scale. This new technique<br />

in computer-assisted quantitative microscopy made possible by CMEIAS<br />

image analysis will undoubtedly impact on our understanding of <strong>plant</strong> <strong>surface</strong><br />

<strong>microbiology</strong> and rhizoplane microbial ecology.<br />

7 Conclusions<br />

This chapter has illustrated with many examples from the author’s work on<br />

the Rhizobium–legume symbiosis how quantitative microscopy can make<br />

important contributions to the field of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>. In addition,<br />

numerous examples illustrate how our CMEIAS software can “count<br />

what really counts” to enhance the quantitative analysis of microbial communities<br />

and populations in situ without cultivation. This opportunity created by<br />

development of CMEIAS will undoubtedly yield fundamentally new information<br />

on <strong>plant</strong>–microbe interactions, and by so doing, expand our understanding<br />

of this fascinating subject of <strong>plant</strong> <strong>surface</strong> <strong>microbiology</strong>.<br />

Acknowledgments. Funds to support portions of the research reported in this chapter<br />

were provided by the Michigan State University Center for Microbial Ecology (National<br />

Science Foundation Grant NO. DEB-91–20006 and the MSU Research Excellence Fund),<br />

the MSU Kellogg Biological Station Long-Term Ecological Research project, the<br />

US–Egypt Science and Technology Joint Fund (projects BI02–001–017–98 and BI05–<br />

001–015), and the Michigan Agricultural Experiment Station. The author thanks Jim<br />

Tiedje, Phil Robertson, Rawle Hollingsworth, Youssef Yanni, Howard Towner, Dominic<br />

Trione, and Edward Marshall for advice and assistance, and the MSU Center for<br />

Advanced Microscopy for use of their facilities.<br />

References and Selected Reading<br />

Abe M, Sherwood JE, Hollingsworth RI, Dazzo FB (1984) Stimulation of clover root hair<br />

infection by lectin-binding oligosaccharides from the capsular and extracellular<br />

polysaccharides of Rhizobium trifolii. J Bacteriol 160:517–520<br />

Bishop P, Dazzo FB, Applebaum E, Maier R, Brill W (1977) Intergeneric transfer of symbiotic<br />

genes from Rhizobium trifolii to Azotobacter vinelandii. Science 198:938–940<br />

Bono JJ, Riond J, Nicolaou KC, Bockovich NJ, Estevez VA, Cullimore JV, Ranjeva R (1995)<br />

Characterization of a binding site for chemically synthesized lipo-oligosaccharidic<br />

NodRm factors in particulate fractions prepared from roots. Plant J 7:253–260<br />

Callaham D, Torrey J (1981) The structural basis for infection of root hairs of Trifolium<br />

repens by Rhizobium. Can J Bot 59:1647–1664


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 545<br />

Cheng HP, Walker GC (1998) Succinoglycan is required for initiation and elongation of<br />

infection threads during nodulation of alfalfa by Rhizobium meliloti. J Bacteriol<br />

180:5183–5191<br />

Crockard MA, Bjourson AJ, Cooper JE (1999) A new peroxidase cDNA from white clover:<br />

Its characterization and expression in root tissue challenged with homologous rhizobia,<br />

heterologous rhizobia or Pseudomonas syringae. Mol Plant-Microbe Interact<br />

12:825–828<br />

Crockard MA, Bjourson AJ, Dazzo FB, Cooper JE (2002) A white clover nodulin gene,<br />

dd23b, encoding a cysteine cluster protein (CCP), is expressed in roots during the<br />

very early stages of interaction with Rhizobium leguminosarum biovar trifolii and<br />

after treatment with chitolipooligosaccharide Nod factors. J Plant Res 115:439–447<br />

Dazzo FB (1982) Leguminous root nodules. In: Burns R, Slater J (eds) Experimental<br />

microbial ecology. Blackwell, Cambridge, pp 431–446<br />

Dazzo FB, Hubbell DH (1975) Cross-reactive antigens and lectin as determinants of<br />

symbiotic specificity in the Rhizobium-clover association. Appl Microbiol 30:1017–<br />

1033<br />

Dazzo FB, Brill WJ (1977) Receptor sites on clover and alfalfa roots for Rhizobium.Appl<br />

Environ Microbiol 33:132–136<br />

Dazzo FB, Brill W (1978) Regulation by fixed nitrogen of host-symbiont recognition in<br />

the Rhizobium-clover symbiosis. Plant Physiol 62:18–21<br />

Dazzo FB, Brill W (1979) Bacterial polysaccharide which binds Rhizobium trifolii to<br />

white clover root hairs. J Bacteriol 137:1362–1373<br />

Dazzo FB, Hrabak EM (1981) Presence of trifoliin A, a Rhizobium-binding lectin, in<br />

clover root exudate. J Supramol Struct Cell Biochem 16:133–138<br />

Dazzo F, Hubbell DH (1982) Control of root hair infection. In: Broughton W (ed) Ecology<br />

of nitrogen fixation: vol II. Rhizobium. Oxford University Press, Oxford, pp 274–310<br />

Dazzo FB, Petersen MA (1989) Applications of computer-assisted image analysis for<br />

microscopic studies of the Rhizobium-legume symbiosis. Symbiosis 7:193–210<br />

Dazzo FB, Wright SF (1996) Production of anti-microbial antibodies and their use in<br />

immunofluorescence microscopy. In: Akkermans A, van Elsas J, de Bruijn F (eds) Molecular<br />

microbial ecology manual. vol 4.12. Kluwer, Dordrecht, pp 1–27<br />

Dazzo FB, Wopereis J (2000) Unraveling the infection process in the Rhizobium-legume<br />

symbiosis by microscopy. In: Triplett E (ed) Prokaryotic nitrogen fixation: a model<br />

system for the analysis of a biological process. Horizon Scientific Press,Wymondham,<br />

UK, pp 295–347<br />

Dazzo FB, Napoli C, Hubbell DH (1976) Adsorption of bacteria to roots as related to host<br />

specificity in the Rhizobium-clover symbiosis. Appl Environ Microbiol 32:166–177<br />

Dazzo FB,Yanke W, Brill W (1978) Trifoliin: a Rhizobium recognition protein from white<br />

clover. Biochim Biophys Acta 536:276–286<br />

Dazzo FB, Urbano MR, Brill WJ (1979) Transient appearance of lectin receptors on Rhizobium<br />

trifolii. Curr Microbiol 2:15–20<br />

Dazzo FB, Truchet GL, Sherwood JE, Hrabak EM, Gardiol AE (1982) Alteration of the trifoliin<br />

A-binding capsule of Rhizobium trifolii 0403 by enzymes released from clover<br />

roots. Appl Environ Microbiol 44:478–490<br />

Dazzo FB, Truchet G, Sherwood J, Hrabak E, Abe M, Pankratz HS (1984) Specific phases<br />

of root hair attachment in the Rhizobium trifolii-clover symbiosis. Appl Environ<br />

Microbiol 48:1140–1150<br />

Dazzo FB, Hollingsworth RI, Abe M, Smith KB, Welsch M, Morris PJ, Philip-Hollingsworth<br />

S, Salzwedel JL, Castillo RM (1987) Rhizobium trifolii polysaccharides,<br />

oligosaccharides, and other metabolites affecting development and symbiotic infection<br />

of clover root hairs. In: Steffens G, Rumsey T (eds) Biomechanisms regulating<br />

growth and development: keys to progress. Beltsville Symposium XII on Agricultural<br />

Research. Kluwer, Dordrecht, pp 343–355


546<br />

Frank B. Dazzo<br />

Dazzo FB, Hollingsworth R, Philip-Hollingsworth S, Robeles M, Olen T, Salzwedel J,<br />

Djordjevic M, Rolfe B (1988) Recognition processes in the Rhizobium trifolii-white<br />

clover symbiosis. In: Bothe H, de Bruijn F, Newton W (eds) Nitrogen fixation: hundred<br />

years after. Gustav Fischer, Stuttgart, Germany, pp 431–436<br />

Dazzo F, Truchet G, Hollingsworth R, Hrabak E, Pankratz H, Philip-Hollingsworth S,<br />

Salzwedel J, Chapman K, Appenzeller L, Squartini A, Gerhold D, Orgambide G (1991)<br />

Rhizobium lipopolysaccharide modulates infection thread development in white<br />

clover root hairs. J Bacteriol 173:5371–5384<br />

Dazzo FB, Mateos P, Orgambide G, Philip-Hollingsworth S, Squartini A, Subba-Rao NS,<br />

Pankratz HS, Baker D, Hollingsworth R,Whallon J (1993) The infection process in the<br />

Rhizobium-legume symbiosis and visualization of rhizoplane microorganisms by<br />

laser scanning confocal microscopy. In: Guerrero R, Pedros-Alio C (eds) Trends in<br />

microbial ecology. Spanish Society for Microbiology, Barcelona, pp 259–262<br />

Dazzo FB, Orgambide G, Philip-Hollingsworth S, Hollingsworth RI, Ninke K, Salzwedel<br />

JL (1996a) Modulation of development, growth dynamics, wall crystallinity, and<br />

infection thread formation in white clover root hairs by membrane chitolipooligosaccharides<br />

from Rhizobium leguminosarum bv. trifolii. J Bacteriol 178:3621–3627<br />

Dazzo FB, Orgambide G, Philip-Hollingsworth S, Hollingsworth RI, Ninke K, Smith D,<br />

Mateos PF, Squartini A, Bjourson AJ, Cooper JE, Wopereis J (1996b) Involvement of<br />

membrane chitolipo-oligosaccharides in the Rhizobium-white clover symbiosis. In:<br />

Chordi-Corbo A, Martinez-Molina E, Mateos P, Carpio-Santos M (eds) Advances in<br />

the investigation on biological nitrogen fixation. Excma Diputacion Provincal De<br />

Salamanca, Salamanca, Spain, pp 29–33<br />

Dazzo FB, Joseph AR, Gomma AB, Yanni YG, Robertson GP (2003) Quantitative indices<br />

for the autecological biogeography of a Rhizobium endophyte of rice at macro and<br />

micro spatial scales. Symbiosis 35:147–158<br />

Diaz CL, Melchers LS, Hooykaas PJ, Lugtenberg BJ, Kijne JW (1989) Root lectin as a<br />

determinant of host-<strong>plant</strong> specificity in the Rhizobium-legume symbiosis. Nature<br />

338:579–581<br />

Diaz CL, Spaink HP, Kijne JW (2000) Heterologous rhizobial lipochitin and chitin<br />

oligomers induced cortical cell divisions in red clover roots transformed with the pea<br />

lectin gene. Molec Plant-Microbe Interact 13:268–276<br />

Djordjevic MA, Gabriel DW, Rolfe BG (1987) Rhizobium: the refined parasite of legumes.<br />

Annu Rev Phytopathol 25:145–168<br />

Fåhraeus G (1957) The infection of clover roots by nodule bacteria studied by a simple<br />

glass slide technique. J Gen Microbiol 16:374–381<br />

Fernandez A, Hashsham S, Dollhopf D, Raskin L, Glagoleva O, Dazzo FB, Hickey R, Criddle<br />

C, Tiedje JM (2000) Flexible community structure correlates with stable community<br />

function in methanogenic bioreactor communities perturbed by glucose. Appl<br />

Environ Microbiol 66:4058–4067<br />

Gerhold DL, Dazzo FB, Gresshoff PM (1985) Selective removal of seedling root hairs for<br />

studies of the Rhizobium-legume symbiosis. J Microbiol Meth 4:95–102<br />

Hashsham S, Fernandez A, Dollhopf S, Dazzo FB, Hickey R, Tiedje JM, Criddle CS (2000)<br />

Parallel processing of substrate correlates with greater functional stability in<br />

methanogenic bioreactor communities perturbed by glucose. Appl Environ Microbiol<br />

66:4050–4057<br />

Hirsch A (1999) Role of lectins (and rhizobial exopolysaccharides) in legume nodulation.<br />

Curr Opin Plant Biol 2:320–326<br />

Hollingsworth RI, Abe M, Sherwood JE, Dazzo FB (1984) Bacteriophage-induced acidic<br />

heteropolysaccharide lyases that convert acidic heteropolysaccharides of Rhizobium<br />

trifolii into oligosaccharide units. J Bacteriol 160:510–516


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 547<br />

Hollingsworth RI, Dazzo FB, Hallenga K, Musselman B (1988) The complete structure of<br />

the trifoliin A lectin-binding capsular polysaccharide of Rhizobium trifolii 843. Carbohydr<br />

Res 172:97–112<br />

Hollingsworth RI, Squartini A, Philip-Hollingsworth S, Dazzo FB (1989) Root hair<br />

deforming and nodule initiating factors from Rhizobium trifolii. In: Lugtenberg B<br />

(ed) Signal molecules in <strong>plant</strong>s and <strong>plant</strong>-microbe interactions. Springer, Berlin Heidelberg<br />

New York, pp 387–393<br />

Holt JJ, Krieg NR, Sneath PH, Staley JT, Williams ST (1994) Bergey’s manual of determinative<br />

bacteriology 9th edn. Williams and Wilkins, Baltimore, 787 pp<br />

Hrabak EM, Urbano MR, Dazzo FB (1981) Growth-phase dependent immunodeterminants<br />

of Rhizobium trifolii lipopolysaccharide which bind trifoliin A, a white clover<br />

lectin. J Bacteriol 148:697–711<br />

Hrabak EM, Truchet GL, Dazzo FB, Govers F (1985) Characterization of the anomalous<br />

infection and nodulation of subterranean clover roots by Rhizobium leguminosarum<br />

1020. J Gen Microbiol 131:3287–3302<br />

Jiminez-Zurdo J, Mateos P, Dazzo FB, Martinez-Molina E (1996) Cell-bound cellulase<br />

and polygalacturonase production by Rhizobium and Bradyrhizobium species. Soil<br />

Biol Biochem 28:917–921<br />

Leigh J, Reed J, Hanks J, Hirsch A, Walker G (1987) Rhizobium meliloti mutants that fail<br />

to succinylate their calcofluor-binding exopolysaccharide are defective in nodule<br />

invasion. Cell 51:579–587<br />

Lerouge P, Roche P, Faucher C, Maillet F, Truchet G, Prome J, Denarie J (1990) Symbiotic<br />

host-specificity of Rhizobium meliloti is determined by a sulfated and acylated glucosamine<br />

oligosaccharide signal. Nature 344:781–784<br />

Li D, Hubbell DH (1969) Infection thread formation as a basis for nodulation specificity<br />

in Rhizobium-strawberry clover associations. Can J Microbiol 15:1133–1136<br />

Liu J, Dazzo FB, Glagoleva O, Yu B, Jain A (2001) CMEIAS „ : a computer-aided system for<br />

image analysis of microbial communities. Microbial Ecology 41:173–194, 42:215<br />

Lopez-Lara I, Orgambide G, Dazzo FB, Olivares J, Toro N (1993) Characterization and<br />

symbiotic importance of acidic extracellular polysaccharides of Rhizobium sp. strain<br />

GRH2 isolated from Acacia nodules. J Bacteriol 175:2826–2832<br />

Lopez-Lara I, Orgambide G, Dazzo FB, Olivares J, Toro N (1995) Surface polysaccharide<br />

mutants of Rhizobium sp. (Acacia) strain GRH2: major requirement of lipopolysaccharide<br />

and acidic exopolysaccharide for successful invasion of Acacia nodules and<br />

host range determination. Microbiology (UK) 141:573–581<br />

Mateos P, Jiminez J, Chen J, Squartini A, Martinez-Molina E, Hubbell DH, Dazzo FB<br />

(1992) Cell-associated pectinolytic and cellulolytic enzymes in Rhizobium trifolii.<br />

Appl Environ Microbiol 58:1816–1822<br />

Mateos P, Baker D, Philip-Hollingsworth S, Squartini A, Peruffo A, Nuti M, Dazzo FB<br />

(1995) Direct in situ identification of cellulose microfibrils associated with Rhizobium<br />

leguminosarum biovar trifolii attached to the root epidermis of white clover.<br />

Can J Microbiol 41:202–207<br />

Mateos PF, Zurdo J, Molina-Blanco J, Velazquez A, Dazzo FB, Martinez-Molina E (1996)<br />

Implication of cellulase production by Rhizobium in the establishment of the symbiosis<br />

with legumes. In: Chordi-Corbo A, Martinez-Molina E, Mateos P, Capri-Santos<br />

M (eds) Advances in the investigation on biological nitrogen fixation, Excma Diputacion<br />

Provincal De Salamanca, Salamanca, Spain, pp 45–48<br />

Mateos P, Baker DL, Petersen M, Velázquez E, Jiménez-Zurdo JI, Martínez-Molina E,<br />

Squartini A, Orgambide G, Hubbell DH, Dazzo FB (2001) Erosion of root epidermal<br />

cell walls by Rhizobium polysaccharide-degrading enzymes as related to primary<br />

host infection in the Rhizobium-legume symbiosis. Can J Microbiol 47:475–487<br />

McDermott TR, Dazzo FB (2002) Use of fluorescent antibodies for studying the ecology<br />

of soil- and <strong>plant</strong>-associated microbes. In: Hurst C, Crawford RC, Knudsen GR, McIn-


548<br />

Frank B. Dazzo<br />

erney MJ, Stetzenbach LD (eds), Manual of environmental <strong>microbiology</strong>, Chap. 28,<br />

American Society for Microbiology Press, Washington, DC, pp 615–626<br />

McKhann HI, Hirsch AM (1993) In situ localization of specific mRNAs in <strong>plant</strong> tissues.<br />

In: Thompson J, Glick B (eds) Methods in <strong>plant</strong> molecular biology and biotechnology.<br />

CRC Press, Boca Raton, pp 173–205<br />

Munoz J, Coronado C, Perez-Hormeache J, Kondorosi A, Ratet P, Palomares AJ (1998)<br />

MsPG3, a Medicago sativa polygalacturonase gene expressed during the alfalfa-Rhizobium<br />

meliloti interaction. Proc Natl Acad Sci USA 95:9686–9692<br />

Murray CJ (2002) Sampling and data analysis for environmental <strong>microbiology</strong>. In: Manual<br />

of environmental <strong>microbiology</strong>, American Society for Microbiology Press, Washington,<br />

DC, pp 166–177<br />

Napoli C, Hubbell DH (1976) Ultrastructure of Rhizobium-induced infection threads in<br />

clover root hairs. Appl Microbiol 30:1003–1009<br />

Napoli C, Dazzo FB, Hubbell DH (1975a) Production of cellulose microfibrils by Rhizobium.<br />

Appl Microbiol 30:123–131<br />

Napoli CA, Dazzo FB, Hubbell DH (1975b) Ultrastructure of infection and common antigen<br />

relationships in the Rhizobium-Aeschynomene symbiosis. In: Vincent J (ed) Proceedings<br />

of the 5th Australian Legume Nodulation Conference. Brisbane, Australia,<br />

pp 35–37<br />

Nutman P, Doncaster C, Dart P (1973) Infection of Clover by Root-Nodule Bacteria.<br />

British Film Institute, London<br />

Orgambide G, Philip-Hollingsworth S, Cargill L, Dazzo FB (1992) Evaluation of acidic<br />

heteropolysaccharide structures in Rhizobium leguminosarum biovars altered in<br />

nodulation genes and host range. Mol Plant-Microbe Interact 5:482–488<br />

Orgambide GG, Philip-Hollingsworth S, Hollingsworth RI, Dazzo FB (1994) Flavoneenhanced<br />

accumulation and symbiosis-related biological activity of a diglycosyl diacylglycerol<br />

membrane glycolipid from Rhizobium leguminosarum biovar trifolii. J<br />

Bacteriol 176:4338–4347<br />

Orgambide G, Lee J, Hollingsworth R, Dazzo FB (1995) Structurally diverse chitolipooligosaccharide<br />

Nod factors accumulate primarily in membranes of wild type<br />

Rhizobium leguminosarum bv. trifolii. Biochemistry 34:3832–3840<br />

Orgambide G, Philip-Hollingsworth S, Mateos P, Hollingsworth RI, Dazzo FB (1996) Subnanomolar<br />

concentrations of membrane chitolipooligosaccharides from Rhizobium<br />

leguminosarum biovar trifolii are fully capable of eliciting symbiosis-related<br />

responses on white clover. Plant Soil 186:93–98<br />

Parniske M, Zimmermann C, Cregan PB, Werner D (1990) Hypersensitive reaction of<br />

nodule cells in the Glycine max sp./Bradyrhizobium japonicum symbiosis occurs at<br />

the genotype-specific level. Botanica Acta 103:143–148<br />

Parniske M, Ahlborn B, Werner D (1991) Isoflavonoid inducible resistance to the phytoalexine<br />

glyceollin in soybean rhizobia. J Bacteriol 173:3432–3439<br />

Pellock BJ, Cheng HP, Walker GC (2000) Alfalfa root nodule invasion efficiency is dependent<br />

on Sinorhizobium meliloti polysaccharides. J Bacteriol 182:4310–4318<br />

Philip-Hollingsworth S, Hollingsworth RI, Dazzo FB (1989a) Host-range related structural<br />

features of the acidic extracellular polysaccharides of Rhizobium trifolii and<br />

Rhizobium leguminosarum. J Biol Chem 264:1461–1466<br />

Philip-Hollingsworth S, Hollingsworth RI, Dazzo FB, Djordjevic M, Rolfe BG (1989b)<br />

The effect of interspecies transfer of Rhizobium host-specific nodulation genes on<br />

acidic polysaccharide structure and in situ binding by host lectin. J Biol Chem<br />

264:5710–5714<br />

Philip-Hollingsworth S, Hollingsworth RI, Dazzo F (1991) N-acetylglutamic acid: an<br />

extracellular Nod signal of Rhizobium trifolii ANU843 which induces root hair<br />

branching and nodule-like primordia in white clover roots. J Biol Chem 266:16854–<br />

16858


27 Applications of Quantitative Microscopy in Plant Surface Microbiology 549<br />

Philip-Hollingsworth S, Orgambide G, Bradford J, Smith D, Hollingsworth R, Dazzo FB<br />

(1995) Mutation or increased copy number of nodE has no effect on the spectrum of<br />

chitolipooligosaccharide Nod factors made by Rhizobium leguminosarum bv. trifolii.<br />

J Biol Chem 270:20968–20977<br />

Philip-Hollingsworth S, Hollingsworth RI, Dazzo FB (1997) Structural requirements of<br />

chitolipooligosaccharides from Rhizobium leguminosarum bv. trifolii for uptake and<br />

mitogenic activity in legume roots as revealed by synthetic analogs and bioreactive<br />

fluorescent probes. J Lipid Res 38:1229–1241<br />

Reddy PM, Ladha JK, So R, Hernandez R, Dazzo FB, Angeles O, Ramos M, de Bruijn F<br />

(1997) Rhizobial communication with rice: induction of phenotypic changes, mode<br />

of invasion and extent of colonization. Plant Soil 194:81–98<br />

Reddy C, Liu J, Wadekar M, Prabhu A, Trione D, Marshall E, Zurdo J, Liu F-I, Urbance J,<br />

Dazzo FB (2002a) New features of CMEIAS innovative software for computer-assisted<br />

microscopy of microorganisms and their ecology. 2002 Ann. Mtg., Long-Term Ecological<br />

Research in Row-Crop Agriculture, KBS-LTER Site. Abstract at <br />

Reddy C, Liu F-I, Zurdo J, Dazzo FB (2002b) A new CMEIAS color recognition program<br />

for digital microbial ecology. 2002 Ann. Mtg., Long-Term Ecological Research in Row-<br />

Crop Agriculture, KBS-LTER Site. Abstract at <br />

Robertson P (2002) GS+ Geostatistics for the environmental sciences. Gamma Design<br />

Software, http://www.gammadesign.com<br />

Rolfe BG, Carlson RW, Ridge RW, Dazzo FB, Mateos PF, Pankhurst CE (1996) Defective<br />

infection and nodulation of clovers by exopolysaccharide mutants of Rhizobium leguminosarum<br />

bv. trifolii. Aust J Plant Physiol 23:285–303<br />

Salzwedel J, Dazzo FB (1993) pSym nod gene influence on elicitation of peroxidase activity<br />

from white clover and pea roots by Rhizobia and their cell-free supernatants. Mol<br />

Plant-Microbe Interact 6:127–134<br />

Sanchez B, Coronado C, Philip-Hollingsworth S, Dazzo FB, Palomares A (1997) Structure<br />

and role in symbiosis of the exoB gene of Rhizobium leguminosarum bv. trifolii. Mol<br />

Gen Genet 255:131–140<br />

Schloter M, Borlinghaus R, Bode W, Hartmann A (1993) Direct identification and localization<br />

of Azospirillum in the rhizosphere of wheat using fluorescence-labeled monoclonal<br />

antibodies and confocal scanning laser microscopy. J Microsc 171:173–177<br />

Sherwood JE, Truchet GL, Dazzo FB (1984a) Effect of nitrate supply on in vivo synthesis<br />

and distribution of trifoliin A, a Rhizobium-trifolii binding lectin, in Trifolium repens<br />

seedlings. Planta 162:540–547<br />

Sherwood JE, Vasse JM, Dazzo FB, Truchet GL (1984b) Development and trifoliin Abinding<br />

ability of the capsule of Rhizobium trifolii. J Bacteriol 159:145–152<br />

Smit G, Swart S, Lugtenberg B, Kijne JW (1992) Molecular mechanisms of attachment of<br />

bacteria to <strong>plant</strong> roots. Mol Microbiol 6:2897–2903<br />

Subba-Rao NS, Mateos PF, Baker D, Pankratz HS, Palma J, Dazzo FB, Sprent JI (1995) The<br />

unique root-nodule symbiosis between Rhizobium and the aquatic legume, Neptunia<br />

natans (L. f.) Druce. Planta 196:311–320<br />

Towner H (1999) EcoStat ecological analysis program for windows, Ver. 1.03, Trinity<br />

Software,Campton,NH<br />

Truchet GL, Sherwood JE, Pankratz HS, Dazzo FB (1986) Clover root exudate contains a<br />

particulate form of the lectin, trifoliin A, which binds Rhizobium trifolii. Physiol Plant<br />

66:575–582<br />

Vance CP (1983) Rhizobium infection and nodulation: A beneficial <strong>plant</strong> disease? Annu<br />

Rev Microbiol 37:399–424<br />

van Rhijn P, Goldberg R, Hirsch A1 (1998) Lotus corniculatus nodulation specificity is<br />

changed by the presence of a soybean lectin gene. Plant Cell 10:1233–1249


550<br />

Frank B. Dazzo<br />

van Workum WAT, van Slogeren S, van Brussel AA, Kijne JW (1998) Role of exopolysaccharides<br />

of Rhizobium leguminosarum bv. viciae as host-specific molecules required<br />

for infection thread formation during nodulation of Vicia sativa. Molec Plant-<br />

Microbe Interact 11:1233–1241<br />

Vega-Hernández MC, Pérez-Galdona R, Dazzo FB, Jarabo-Lorenzo A,Alfayate MC, León-<br />

Barrios M (2001) Novel infection process in the indeterminate root nodule symbiosis<br />

between tagasaste (Chamaecytisus proliferus) and Bradyrhizobium sp. (Chamaecytisus).<br />

New Phytol 150:707–721<br />

Vernoud V, Journet EP, Barker DG (1999) MtENOD20, a Nod factor-inducible molecular<br />

marker for root cortical cell activation. Mol Plant-Microbe Interact 12:604–614<br />

Wilcox CD, Dove SB, Doss-McDavid W, Greer DB (1997) UTHSCSA ImageTool „ Ver. 1.27,<br />

http://www.uthscsa.edu/dig/itdesc.html, Univ. Texas Health Science Center, San Antonio,<br />

TX<br />

Yanni Y, Rizk R, Corich V, Squartini A, Ninke K, Philip-Hollingsworth S, Orgambide G,<br />

deBruijn F, Stoltzfus J, Buckley D, Schmidt T, Mateos P, Ladha JK, Dazzo FB (1997) Natural<br />

endophytic association between Rhizobium leguminosarum bv. trifolii and rice<br />

roots and assessment of its potential to promote rice growth. Plant Soil 194:99–114<br />

Yanni YG, Rizk RY, Abd El-Fattah FK, Squartini A, Corich V, Giacomini A, de Bruijn F,<br />

Rademaker J, Maya-Flores J, Ostrom P, Vega-Hernandez M, Hollingsworth RI, Martinez-Molina<br />

E, Mateos P,Velazquez E,Wopereis J, Triplett E, Umali-Garcia M,Anarna<br />

JA, Rolfe BG, Ladha JK, Hill J, Mujoo R, Ng PK, Dazzo FB (2001) The beneficial <strong>plant</strong><br />

growth-promoting association of Rhizobium leguminosarum bv. trifolii with rice<br />

roots. Aust J Plant Physiol 28:845–870<br />

Yao Y,Vincent JM (1976) Factors responsible for the curling and branching of clover root<br />

hair by Rhizobium. Plant Soil 45:1–16


28 Analysis of Microbial Population Genetics<br />

Emanuele G. Biondi, Alessio Mengoni and Marco Bazzicalupo<br />

1 Introduction<br />

The knowledge of genetic diversity in bacterial population has increased considerably<br />

over the last 15 years, due to the application of molecular techniques<br />

to microbial ecological studies. Quantitative resolution has improved as a<br />

large number of haplotypic markers are found within each sample and as a<br />

large number of samples can be simultaneously investigated.<br />

Among the molecular methods, the PCR-based techniques provide a powerful<br />

and high throughput approach for the study of genetic diversity in bacterial<br />

populations. PCR fingerprinting methods for the analysis of biodiversity<br />

are numerous and usually very effective. Some of the most commons are<br />

the PCR-RFLP of specific sequences (16S rDNA, intergenic transcribed<br />

spacer, ITS) (Laguerre et al. 1996), the Repetitive Extragenic Palindromic-PCR<br />

(Woods et al. 1992) and the BOX-PCR (Louws et al. 1994) based on the presence<br />

of repetitive elements within the bacterial genome, the DNA amplification<br />

fingerprintings (DAF; Caetano-Anollés and Bassam 1993), RAPDs (random<br />

amplified polymorphic DNA; Williams et al. 1990,Welsh and McClelland<br />

1990) and AFLPs (amplified fragment length polymorphism; Vos et al. 1995).<br />

Each method has advantages and disadvantages and the choice of the appropriate<br />

one depends on the expected degree of polymorphism within the population,<br />

the selection of the specific genomic region and the possibility of<br />

automation for screening of large samples. ITS, RAPD and AFLP have been<br />

shown to be particularly relevant for the study of genetic diversity within<br />

populations of bacteria belonging to the same or closely related species.<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


552<br />

Emanuele G. Biondi<br />

2 Materials for RAPD, AFLP and ITS<br />

Equipment:<br />

– Thermal cycler<br />

– Gel electrophoresis apparatus with power supply, agarose and polyacrylamide<br />

(sequencing)<br />

– Automated sequencer for capillary electrophoresis equipped with discrete<br />

band analysis software<br />

– UV transilluminator and gel documentation system<br />

Caution: UV rays are dangerous. Protect eyes with a plastic shield<br />

Reagents and solutions:<br />

– double distilled water (ddH 2 O) sterilised by autoclaving. Prepare 100 ml<br />

aliquots before sterilisation and keep at –20 °C. Discard the aliquot after<br />

each use<br />

– 50 mM MgCl 2 stock solution usually supplied with the Taq enzyme<br />

– dNTPs stock solution (2 mM of each dNTP in ddH 2O)<br />

– Taq DNA polymerase<br />

– Restriction enzymes: EcoRI and MseI, a single restriction buffer compatible<br />

with both enzymes<br />

– T4 DNA ligase and specific ligation buffer, 5x stock solution supplied with<br />

the ligase enzyme<br />

– Double-stranded adapters for AFLP (use 50 pmol for each adapter in the<br />

ligation mixture). The sequence of two single stranded oligonucleotides<br />

(5¢–3¢) corresponding to double-stranded adapters are reported. To prepare<br />

the double-strand molecule, incubate 100 pmol/ml of each oligonucleotide<br />

at 94 °C for 10 min and then slowly decrease the temperature down<br />

to 4 °C. Keep at –20 °C<br />

EcoRI adapter oligonucleotides:<br />

5¢–CTC GTA GAC TGC GTA CC–3¢<br />

5¢–AAT TGG TAC GCA GTC TAC–3¢<br />

EcoRI double stranded adapter:<br />

5¢–CTC GTA GAC TGC GTA CC–3¢<br />

5¢–AAT TGG TAC GCA GTC TAC–3¢<br />

MseI adapter oligonucleotides:<br />

5¢–GAC GAT GAG TCC TGA G–3¢<br />

5¢–TAC TCA GGA CTC AT–3¢<br />

MseI double stranded adapter:<br />

5¢–GAC GAT GAG TCC TGA G–3¢<br />

5¢–TC TCA GGA CTC TA–3¢<br />

– Primers for AFLP (without selective bases): pEcoRI-T (5¢–GAC TGC GTA<br />

CCA ATT C-T–3¢), 5¢ labelled with 6-carboxifluorescein (6-FAM); pMseI-A<br />

(5¢–GAT GAG TCC TGA GTA-A–3¢), 5¢ labelled with 4,7,2¢,4¢,5¢,7¢-hexachloro-6-carboxyfluorescein<br />

(HEX). Prepare 10 mM stock solution


– Primers for ITS amplification: FGPS1490 (5¢–TGCGGCTGGATCACCTC-<br />

CTT–3¢) and FGPS132¢ (5¢–CCGGGTTTCCCCATTCGG–3¢), 10 mM stock<br />

solution in ddH 2O. These primers are selected for Rhizobia and may apply<br />

to other bacterial groups, however specific primers for particular genera<br />

can be designed on known 16S and 23S sequences retrieved from GenBank<br />

or Ribosomal Database (RDP)<br />

– 10-base random primers for RAPD (series OP from Operon Technologies),<br />

80 mM stock solution in ddH 2O. The choice of the primers is highly relevant<br />

for the usefulness of the results obtained (see the RAPD principles section<br />

for details)<br />

– DNA size marker: good examples are a 100-bp ladder for agarose gel electrophoresis<br />

and TAMRA 500 (Applied Biosysem, PE) for capillary electrophoresis<br />

– Genomic DNA: for RAPD and ITS concentrations at 10 ng/ml in ddH 2 O. For<br />

the AFLP, use concentrations at 50 ng/ml. For general extraction protocols,<br />

see Bazzicalupo and Fancelli (1997).Alternatively, use BIO 101 DNA extraction<br />

kit<br />

Note: All the above reagents should be kept at –20 °C<br />

– TAE buffer: 40 mM Tris/Acetate, 1 mM EDTA, pH 8. Prepare a 50x stock<br />

solution<br />

– ethidium bromide stock solution: 10 mg/ml; store in a dark bottle<br />

– agarose<br />

– 10x loading buffer: 70 % (w/v) glycerol, 0.5 % (w/v) bromophenol blue;<br />

store at 4 °C<br />

Caution: Ethidium bromide is a powerful mutagen: wear gloves when handling<br />

this compound; wear mask when weighing it<br />

3 RAPD<br />

28 Analysis of Microbial Population Genetics 553<br />

Principle<br />

The RAPD assay (Welch and McClelland 1990; Williams et al. 1990) is a PCR<br />

amplification performed on genomic DNA template using a single short, arbitrary<br />

oligonucleotide primer and low annealing temperature, conditions that<br />

ensure the generation of several discrete DNA products. Each of these fragments<br />

is derived from a region of the genome that contains two primer binding<br />

sites on opposite strands and at an amplifiable distance. Polymorphism<br />

between strains results from sequence differences which inhibit or enhance<br />

primer binding or otherwise affect amplification. Single base mutations,<br />

insertions and deletions are molecular events that produce RAPD polymorphism.<br />

The large number of bands amplified with a single arbitrary primer<br />

generates a complex fingerprinting that can be utilised to detect relative differences<br />

in the random amplified DNA sequences from two different<br />

genomes. RAPDs have been applied to bacterial population genetics for sev-


554<br />

Emanuele G. Biondi<br />

eral species which live in association with <strong>plant</strong>s, such as Sinorhizobium<br />

meliloti (Paffetti et al. 1996; Carelli et al. 2000), Burkholderia cepacia (Di Cello<br />

et al. 1997) and Pseudomonas (Picard et al. 2000).<br />

Although the sequence of RAPD primers is arbitrarily chosen, two basic<br />

criteria must be met: a minimum of 40 % G+C content (50–80 % G+C content<br />

is generally used) and the absence of palindromic sequences. Primers can also<br />

be purchased as a specific set for RAPD reactions from Operon Technologies<br />

(http://www.operon.com/).<br />

Experimental procedure<br />

In order to minimise the risk of contamination, the reaction should be prepared<br />

with a set of pipettes and tips used exclusively for this purpose<br />

in a clean environment (laminar flow hood being optimal) and wearing<br />

gloves.<br />

1. Prepare a master mix in ice of those reagents common to all the programmed<br />

reactions, i.e. dNTPs, MgCl 2 , primer, buffer and Taq DNA polymerase.<br />

Mix all reagents well. Prepare a quantity sufficient for the samples<br />

and for control reactions in which template DNA is omitted. Usually RAPD<br />

reactions are carried out in 25 ml total volume in the 0.2-ml PCR tube. The<br />

following concentrations are required:<br />

– Template DNA: 1 ng/ml<br />

– dNTPs: 200 mM<br />

– Primer: 6.4 mM<br />

– MgCl 2 :3mM<br />

– Taq buffer: 1x strength<br />

– Taq DNA polymerase: 0.032 U/ml<br />

For 10 samples, 11 reactions should be prepared using the following volumes:<br />

– ddH 2O: 149.6 ml<br />

– 10x Taq buffer: 27.5 ml<br />

– 2 mM dNTPs: 27.5 ml<br />

– 80mM primer: 22 ml<br />

– 50 mM MgCl 2: 16.5 ml<br />

– 2 U/ml Taq DNA polymerase: 4.4 ml<br />

2. Aliquot the DNA (25 ng=2.5 ml) in the PCR tubes and then add the required<br />

volume of master mix (22.5 ml per tube)<br />

3. Place the tubes in the thermal cycler and perform an initial denaturation<br />

step at 94 °C for 5 min<br />

4. Cycle the reactions 45 times with the following temperature profile: denaturation<br />

at 94 °C for 1 min, annealing at 36 °C for 1 min and extension at<br />

72 °C for 2 min. After the last cycle perform an extension step of 10 min<br />

5. Store samples at 4 °C for a few hours (or –20 °C if longer)<br />

6. Prepare a 2 % agarose gel in TAE buffer with 1 mg/ml of ethidium bromide.<br />

It is advisable to use a comb with teeth as thin as possible: the thinner the


28 Analysis of Microbial Population Genetics 555<br />

teeth, the sharper the bands will appear. Caution: ethidium bromide is a<br />

mutagen! Wear gloves when handling<br />

7. Add 1 ml of 10x loading buffer to 9 ml of each sample<br />

8. Load the samples and the required amount of size marker<br />

9. Run the gel at 10 V/cm for 1 h 30 min<br />

10. Document the gel on UV transilluminator<br />

Results and Comments<br />

RAPD is a fast, cheap and powerful technique, which generates a high amount<br />

of polymorphism, being able to distinguish among isolates of the same populations<br />

in a very effective manner. The RAPD assay, according to the described<br />

protocol, generates reproducible fingerprints. Usually the size of RAPD products<br />

ranges from a few hundreds to about 2000 bp (Fig. 1). As a rule, the highest<br />

and lowest bands should be avoided as they are less reproducible. Before<br />

starting the analysis, a collection of primers, usually 20–30, should be tested<br />

on a selected subsample of strains in order to choose those that appear more<br />

suitable for the purpose and exclude those that did not show polymorphism.<br />

In general, four to six primers with different degrees of polymorphism are<br />

used for population analysis.<br />

Troubleshooting<br />

– Low polymorphism: use different primers.<br />

– Reproducibility: use the same enzyme brand, the same thermal cycler for<br />

all the experiments. Poor quality or insufficient amounts of template DNA<br />

are most likely involved for low reproducibility. Perform RAPD reactions<br />

twice on at least some of the samples to check the reproducibility of all the<br />

recorded bands.<br />

– Smearing: an excessive amount of template or primer or Taq DNA polymerase<br />

has most likely been used. Perform test reaction with reduced<br />

amount of each of these components at a time.<br />

Fig. 1. RAPD pattern of different isolates of Sinorhizobium meliloti. M Ladder 100 bp<br />

(Life Technologies)


556<br />

– Low intensity of the bands: insufficient amount of primer or dNTPs. Try<br />

increasing the amount of each one of these components at a time.<br />

3 AFLP<br />

Emanuele G. Biondi<br />

Principle<br />

Amplified Fragment Length Polymorphism (AFLP) is a recently developed<br />

technique based on restriction and amplification (Zabeau et al, 1993; Vos et al.<br />

1995; Fig. 2). Using this method it is possible to generate up to 100 genomic<br />

markers with a single combination of restriction enzyme and primers. In particular,<br />

the application to microbial population analysis has been used to differentiate<br />

bacteria at strain and species levels from the taxonomic, phylogenetic<br />

or the population genetics point of view (Biondi et al. 2003). Moreover,<br />

adapter<br />

GAATT<br />

C<br />

GAATT<br />

C<br />

primer<br />

genomic DNA<br />

EcoRI MseI<br />

GAATTC<br />

CTTAAG<br />

C<br />

TTAAG<br />

C<br />

TTAAG<br />

GAATTC<br />

CTTAAG<br />

AATT<br />

TTAA<br />

Digestion with EcoRI and MseI<br />

T<br />

AAT<br />

Ligation with Adapters<br />

T<br />

AAT<br />

TTAA<br />

AATT<br />

TAA<br />

T<br />

TAA<br />

T<br />

Annealing of Selective primers<br />

adapter adapter<br />

Amplification<br />

primer<br />

adapter<br />

Fig. 2. Outline of AFLP technique. In dark grey the EcoRI restriction site and in light<br />

grey the MseI restriction site. See text for details


28 Analysis of Microbial Population Genetics 557<br />

the AFLP analysis can be applied to map phenotypic traits in eukaryotic<br />

organisms.<br />

The genomic DNA is digested with restriction enzymes chosen to obtain<br />

fragments whose size is less than 1 Kb. After the digestion, all the fragments<br />

are ligated with adapters which recognise the digested ends. During this passage<br />

all DNA molecules acquire the same sequence at the ends. The ligated<br />

DNA is used as a template for PCR amplification; the primers used in this<br />

amplification are complementary to the adapter’s sequence. Moreover, by<br />

adding one or two bases to the 3¢ end of the primers sequence, it is possible to<br />

obtain different numbers of genetic markers: more selective bases result in<br />

the reduction of the number of amplified fragments. Finally, the detection of<br />

the amplified fragments and the estimation of their size can be obtained by<br />

two methods: polyacrylamide gel electrophoresis and capillary electrophoresis.<br />

This second method is more powerful and easier to handle and, therefore<br />

we will discuss only this method to analyse AFLP results.<br />

Experimental procedures<br />

1. Prepare the DNA using an extraction method that preserves the integrity of<br />

high molecular weight molecules (see material for reference)<br />

2. Digest 200 ng aliquots of extracted genomic DNA in 25 ml final volume with<br />

5U ofEcoRI and 5 U of MseI using as enzyme buffer the MseI buffer supplied<br />

by the manufacturer, incubate 2 h at 37 °C. Heat-inactivate the<br />

enzymes at 70 °C for 15 min<br />

3. Ligate the adapters to the restriction products by adding 25 ml of the 2x ligation<br />

solution (1 unit of T4 DNA ligase, 50 pmoles of each adapter) to the<br />

digestion mixture (50 ml final volume) using double-stranded adapters<br />

with single-stranded overhang complementary to 5¢ and 3¢ ends generated<br />

during digestion. The ligation solution is incubated for 2 h at 20 °C<br />

4. Perform the amplification reactions in a 50 ml total volume containing, 10x<br />

reaction buffer, 2.5 mM MgCl 2, 0.2 mM of each dNTP, 1.6 U of Taq DNA<br />

polymerase, 10 pmoles of each primer, 1 ml of template DNA (corresponding<br />

to approximately 4 ng of digested and ligated genomic DNA). For example:<br />

for 10 samples, consider a master mix solution for 11 single PCR reactions;<br />

add 1ml of template derived from the AFLP ligation to a 0.2-ml PCR<br />

tube; prepare a master mix (MM) with 408.1 ml of ddH 2 O, 11ml of each<br />

primer solution, 55ml PCR buffer 10x, 27.5 ml of a 50 mM MgCl 2 solution<br />

and finally 9.9 ml of Taq DNA polymerase solution (3.5 U/ml); mix gently<br />

and aliquot 50 ml of the MM solution in each tube. The PCR conditions have<br />

been optimised in a Perkin-Elmer 9600 thermocycler (Perkin-Elmer, Norfolk,<br />

CT, USA), using the following amplification program: (94 °C for 30 s +<br />

65 °C for 30 s’ + 72 °C for 60 s) repeated for 13 cycles, decreasing the annealing<br />

temperature by 0.7 °°C each cycle and 23 cycles as follows: 94 °C for 30 s<br />

+ 56 °C for 30 s + 72 °C for 60 s. Several combinations of primers can be<br />

selected, but good results were obtained with: pEcoRI-T (5¢–GAC TGC GTA


558<br />

Emanuele G. Biondi<br />

CCA ATT CT–3¢), labelled with 5¢-6-carboxifluorescein (6-FAM) and<br />

pMseI-A (5¢–GAT GAG TCC TGA GTA AA–3¢), labelled with 5¢-<br />

4,7,2¢,4¢,5¢,7¢-hexachloro-6-carboxyfluorescein (HEX) (in bold the selective<br />

bases)<br />

5. Check the amplification by running a 5 ml aliquot in a 1.5 % agarose gel<br />

6. Size the product on an automatic capillary electrophoresis sequencer<br />

(Perkin-Elmer ABI 310 analyser). Load the capillary with 1.5 ml volume of<br />

AFLP PCR product and 0.5 ml of GenScan internal size standard TAMRA-<br />

500 (PE Biosystems) with 12.5 ml of deionised formamide and perform the<br />

electrophoresis as recommended by the manufacturer for fragment sizing.<br />

Results and Comments<br />

The AFLP technique usually produces a large amount of data (up to 100 different<br />

molecular markers) and for this reason it is recommended to use a<br />

computer-based system to manage the results. In this section we will discuss<br />

only the DNA sequencer output data analysis which gives data corresponding<br />

to fragments between 50 and 500 bp (range of TAMRA 500 molecular<br />

marker). The first step is the selection of useful data from the raw results.<br />

First, reject all peaks derived from single fluorochromes and analyse only the<br />

signals derived from both fluorochromes. After that, a threshold has to be<br />

introduced in order to continue only with real amplification signals. Usually<br />

only peaks having an intensity higher than 50 Fluorescence Units will be<br />

selected.After the selection, signals will be used to compute a distance matrix<br />

from which the genetic structure of the population can be analysed.<br />

Troubleshooting<br />

– Low intensity of the amplified AFLP bands: check the purity and the<br />

amount of DNA (100–300-ng range), try a different extraction method and<br />

different amount of DNA, test the reagents and the procedure with control<br />

DNA and control primers. Check that the primers used are correctly<br />

labelled. For the PCR reaction try a different amount of ligated DNA<br />

(1–4 ml) as template. Magnesium chloride concentration and annealing<br />

temperature are most likely involved in poor amplification, perform test<br />

reactions modifying the amount/value of these variables. Load different<br />

amounts of the PCR product on the automatic sequencer to optimise the<br />

fluorescent signal.<br />

– Too many or too few bands: test different combinations of primers. If the<br />

bands are fewer than expected, remove the extra bases from the adapter<br />

complementary primers. On the contrary, if the bands are too many, add<br />

selective bases of up to two for each primer.


6 ITS-RFLP analysis<br />

28 Analysis of Microbial Population Genetics 559<br />

Principle<br />

The 16S–23S rRNA intergenic transcribed spacer (Fig. 3; ITS, the spacer<br />

sequence between 16S and 23S rRNA bacterial genes synonymous with IGS,<br />

inter genic spacer) is a sequence that exhibits large variability useful in identifying<br />

genomic groups at the intraspecific level (Barry et al. 1991; Jensen et al.<br />

1993; Laguerre et al. 1996; Doignon-Bourcier et al. 2000). The genetic variability<br />

of this particular region derives from: (1) the presence of t-RNA genes<br />

inside the ITS and (2) the mutation rate of ITS higher than that of ribosomal<br />

genes. Restriction fragment length polymorphism of PCR-amplified ITS<br />

(ITS-RFLP) is a fingerprinting method for the characterisation of bacterial<br />

strains with a higher discriminating power than the 16S rDNA RFLP (ARDRA<br />

method). For the amplification of the ITS, different primer pairs, designed on<br />

the coding regions of 16SrRNA and 23SrRNA genes, can be chosen depending<br />

on the bacterial group to be analysed. In general, the forward primer corresponds<br />

to the internal region of the 16S gene while the reverse primer corresponds<br />

to the beginning of the 23S gene. Information on primers for specific<br />

bacterial groups can be retrieved from the specific literature or from Gen-<br />

Bank or RibosomalDataBase (http://www.ncbi.nlm.nih.gov/ or http://rdp.<br />

cme.msu.edu/html/). For the amplification of the ITS region of rhizobia we<br />

used primers FGPS1490 (Navarro et al., 1992) and FGPS132¢ (Ponsonnet and<br />

Nesme 1994). FGPS1490 is designed on conserved sequences of the 3¢ end of<br />

the 16S rRNA gene (corresponding to Eschericha coli numbering positions<br />

1525–1541), and reverse primer FGP132¢ is designed on the 5¢ end of the 23S<br />

rRNA gene (corresponding to the E. coli numbering positions 115–132). For<br />

ITS-RFLP the amplified intergenic region is digested with four-base recognition<br />

site restriction enzymes in order to generate specific patterns of bands.<br />

Depending on the type of the samples and on the aim of the study, from two<br />

to five or more different restriction enzymes are used. The more enzymes<br />

used, the higher the number of bands, i.e. molecular markers produced. The<br />

restriction of the amplification product should be performed using enzymes<br />

which cut several times in the intergenic spacer, thus, before starting to<br />

analyse the IGS of a particular species, a number of enzymes should be tested<br />

to select the best combination. Some restriction enzymes frequently used are:<br />

AluI, MseI, HhaI, TaqI, Sau3A.<br />

16S rDNA ITS/IGS<br />

23S rDNA<br />

Fig. 3. Structure of the bacterial ribosomal operon showing the position of ITS region.<br />

Primers are indicated by arrows


560<br />

Emanuele G. Biondi<br />

Experimental procedures:<br />

1. Perform a PCR amplification reaction in a 50 ml total volume containing,<br />

10x reaction buffer, 2.5 mM MgCl 2, 0.2 mM of each dNTP, 1.6 U of Taq DNA<br />

polymerase, 10 pmoles of each primer (FGPS1490 and FGPS132¢), 25 ng of<br />

template DNA concentrated to 25 ng/ml. For nine samples consider a master<br />

mix solution for ten single PCR reactions with the following volumes:<br />

– ddH 2O: 337ml<br />

– 10x Taq buffer: 50 ml<br />

– 2 mM dNTPs: 50 ml<br />

– 10mM primer FGPS1490 : 10 ml<br />

– 10mM primer FGPS132¢:10ml<br />

– 50 mM MgCl 2 :25ml<br />

– 2U/ml Taq DNA polymerase: 8 ml<br />

– 1ml of template in each tube before aliquoting 49 ml of the master mix<br />

2. Cycle the reactions through the following temperature profiles: initial melting<br />

at 94 °C for 5 min followed by 35 cycles at 94 °C for 1 min, 55 °C for 55 s,<br />

72 °C for 2 min. Perform a final extension step at 72 °C for 10 min<br />

3. Analyse 5 ml of each amplification mixture by agarose gel (1.2 % w/v) electrophoresis<br />

in TAE buffer containing 1 mg/ml (w/v) of ethidium bromide.<br />

Caution: ethidium bromide is mutagenic: wear gloves when handling. The<br />

result of the electrophoresis will ensure that amplification has been successful<br />

and will also help to quantify the amount of amplified DNA<br />

4. Digest approximately 500–600 ng (5–6 ml) of the amplified IGS, with 2 units<br />

of the restriction enzyme in a total volume of 15 ml for 2 h. Use the buffer<br />

and incubation conditions recommended by the manufacturer of the<br />

restriction enzyme. Inactivate the enzyme. Make a separate digestion for<br />

each restriction enzyme to be used<br />

5. Resolve the reaction products (15 ml) by agarose gel (2.5 % w/v) electrophoresis<br />

in TAE buffer run at 10 V/cm and stained with 1 mg/ml (w/v) of<br />

ethidium bromide.<br />

Caution: ethidium bromide is mutagenic: wear gloves when handling<br />

Troubleshooting<br />

– Low intensity of the amplified ITS: check PCR reaction conditions. Magnesium<br />

chloride concentration and annealing temperature are most likely<br />

involved, perform test reactions modifying these variables.<br />

– Partially digested products: excessive amount of amplified ITS, low restriction<br />

enzyme concentration, incubation time too short. Perform test reactions<br />

with a reduced amount of DNA, or add more restriction enzyme or<br />

incubate for a longer time.


7 Statistical analysis<br />

28 Analysis of Microbial Population Genetics 561<br />

Introduction<br />

The studies of microbial population genetics with molecular methods are<br />

often characterised by an extremely high number of samples and by a high<br />

number of molecular markers. As a consequence, an immediate interpretation<br />

of the results can be difficult unless powerful statistical techniques are<br />

used in order to describe the structure of the populations and to highlight the<br />

contributions of its components (Mengoni and Bazzicalupo 2002).<br />

Methods and Procedure<br />

Statistical treatment of data in microbial population genetics include at least<br />

four different levels of analysis:<br />

1. Quantification of the genetic diversity within the population<br />

2. Measurement of genetic distances between strains<br />

3. Analysis of the genetic structure<br />

4. Analysis of the genetic relationships among populations.<br />

Several methods can be used to address each of these points. Here, a brief<br />

summary of the principal parameters and software used is provided.<br />

The molecular data obtained from RAPD,AFLP, and ITS-RFLP analyses are<br />

usually bands in a gel or peaks in a chromatogram. These data are transformed<br />

into a matrix of state binary vectors (molecular haplotype) for each<br />

individual isolate using a compiler such as Microsoft Excel or similar. Bands<br />

and peaks of equal sizes are interpreted as identical and intensity is not considered<br />

as a difference. The molecular haplotype of each isolate is expressed<br />

as a vector of zeroes (for the absence of the band) or ones (for its presence),<br />

assuming that bands represent independent loci.<br />

1. The quantification of genetic diversity within the population can be done<br />

using several parameters. The most commonly used are the gene diversity,<br />

the average gene diversity over loci and the mean number of pairwise differences<br />

between haplotypes.<br />

The gene diversity is equivalent to the expected heterozygosity for diploid<br />

data. It is defined as the probability that two randomly chosen molecular<br />

haplotypes are different in the sample.<br />

The average gene diversity over loci is defined as the probability that two<br />

individuals are different for a randomly chosen locus. These two parameters<br />

vary from 0 (all isolates identical) to 1 (maximum diversity).<br />

The mean number of pair-wise differences simply calculates the mean<br />

number of differences between all pairs of molecular haplotypes in the<br />

population. The computation of these three parameters is performed with<br />

Arlequin software (Schneider et al. 1997).<br />

2. For the measurement of the genetic distances between strains, several<br />

methods can be applied. The basic principle is the ratio of bands shared by


562<br />

Emanuele G. Biondi<br />

two strains with respect to the total ones. One commonly used parameter is<br />

the Euclidean distance whose formalisation is E=n(1–2n xy /2n), where n is<br />

the total number of bands of strain x and y and n xy the number of bands<br />

shared by strains x and y. Another widely used parameter is the Nei’s<br />

distance which, using the same notation, can be formalised as<br />

D=1–[2n xy/(n x+n y)], where n x and n y are the number of bands present in the<br />

strains x and y,respectively.<br />

3. Examples of techniques for ordering the genetic diversity to analyse the<br />

genetic structure of a population are the analysis of molecular variance<br />

(AMOVA) and the principal component analysis (PCA). The AMOVA is a<br />

methodology for the analysis of variance which makes use of molecular<br />

data. AMOVA allows us to uncover the structure of the population and to<br />

test the validity of the hypotheses on the subdivision of the analysed population.<br />

AMOVA was designed by Excoffier, Smouse and Quattro in 1992<br />

(Excoffier et al. 1992) as “an alternative methodology that makes use of<br />

available molecular information gathered in population surveys, while<br />

remaining flexible enough to accommodate different types of assumptions<br />

about the evolutionary genetic system” (Excoffier et al. 1992). Assuming<br />

that a set of samples belongs to different populations and that these populations<br />

could be arranged in genetically distinguishable groups, the aim of<br />

AMOVA is to perform statistical tests on the hypothesised genetic structure.A<br />

hierarchical analysis of variance splits the total genetic variance into<br />

components due to intra-population differences among individual samples,<br />

inter-population differences and inter-group differences.<br />

The PCA is an analysis in which a data set is searched for some significant<br />

independent variables, with respect to all possible variables. These variables<br />

are termed ‘components’ and interest attaches especially to the principal,<br />

or most important, components, hence the name ‘principal component<br />

analysis’. The output of the analysis is a plot in which the samples are<br />

dispersed in a two- or three-dimensional space allowing the recognition of<br />

the clusterisation pattern with respect to one of the dimensions (components).<br />

4. The genetic relationships among populations can be estimated as the<br />

results of AMOVA with respect to the variance between populations. The<br />

parameter of the genetic separation between populations is F ST (Wright<br />

1965) which derives directly from the analysis of variance. The F ST values<br />

can be used to construct a matrix of distances whose representation takes<br />

the form of a dendrogram or tree. Two tree-building methods are applicable<br />

to the distance matrix: UPGMA and Neighbor-Joining (Saitou and Nei<br />

1987). The UPGMA is based on a simple mathematical algorithm in which<br />

a step-wise clusterisation is made. The Neighbor-Joining method is a simplified<br />

version of a minimal evolution method; a star-like tree is made and<br />

then the topology is reconstructed on the basis of the minimisation of the<br />

overall length of the tree.


Software requirements<br />

– Scoring of the bands: MICROSOFT EXCEL or similar.<br />

– Quantification of genetic diversity within population: ARLEQUIN (Schneider<br />

et al. 1997).<br />

– Measurement of the genetic distances between strains: ARLEQUIN<br />

(Schneider et al. 1997);<br />

NTSYS-pc (Rohlf 1990)<br />

RAPDistance (freely downloaded from http://life.anu.edu.au/molecular/<br />

software/rapd.html)<br />

– Analysis of the genetic structure:<br />

(1) AMOVA: ARLEQUIN (Schneider et al. 1997);<br />

(2) PCA: NTSYS-pc (Rohlf 1990)<br />

– Estimation of genetic relationships among populations: ARLEQUIN<br />

(Schneider et al. 1997)<br />

MEGA (Kumar et al. 1993)<br />

NTSYS-pc (Rohlf 1990)<br />

PHYLIP (Freely downloaded from<br />

http://evolution.genetics.washington.edu/phylip.html)<br />

Many of these softwares exist either as DOS/Windows and as Mac versions.<br />

For ARLEQUIN a Linux version also has been developed.<br />

8 Concluding Remarks<br />

28 Analysis of Microbial Population Genetics 563<br />

Several techniques for the analysis of genetic diversity of bacterial populations<br />

have been proposed. RAPD, ITS and AFLP are effective technologies able<br />

to show intra-population polymorphism and to detect phylogenetic relationships<br />

among strains belonging to the same or closely related bacterial species.<br />

RAPD is a suitable technique in that it is fast, cheap and the amount of polymorphism<br />

displayed is high. RAPD has the disadvantage of requiring accurate<br />

setting up of the conditions to obtain high reproducibility. ITS-RFLP analysis<br />

on the contrary, shows less polymorphism, which is linked to a defined DNA<br />

region, being more suitable to define phylogenetic relationships among<br />

strains.<br />

AFLP shows some advantages over the other methods: (1) the high stringency<br />

of the PCR conditions gives robust reproducibility; (2) easy application<br />

to <strong>plant</strong>, animal and bacterial genomic DNA. AFLP requires more DNA than<br />

RAPD and ITS-RFLP and a more laborious procedure. Nevertheless,AFLP has<br />

a high informational content per single reaction, in fact, up to 100 different<br />

bands can be displayed in a single lane and the scoring can be done with an<br />

automatic sequencer.


564<br />

Emanuele G. Biondi<br />

References and Selected Reading<br />

Barry T, Colleran G, Glennon M, Dunican LK, Gannon F (1991) The 16 S/23 S ribosomal<br />

spacer region as a target for DNA probes to identify eubacteria. PCR Methods Appl<br />

1:51–56<br />

Bazzicalupo M, Fancelli S (1997) DNA extraction from bacterial colonies. In: Micheli<br />

MR, Bova R (eds) Fingerprinting methods based on arbitrary primed PCR. Springer,<br />

Berlin Heidelberg New York, pp 41–46<br />

Biondi EG, Pilli E, Giuntini E, Roumiantseva ML, Andronov EE, Onichtchouk OP, Kurchak<br />

ON, Simarov BV, Dzyubenko NI, Mengoni A, Bazzicalupo M (2003) Evolutionary<br />

relationship of Sinorhizobium meliloti and Sinorhizobium medicae strains isolated<br />

from Caucasian region. FEMS Lett 220:207–213<br />

Caetano-Anollés G, Bassam BJ (1993) DNA amplification fingerprinting using arbitrary<br />

oligonucleotide primers. Appl Biochem Biotech 42:189–200<br />

Carelli M, Gnocchi S, Fancelli S, Mengoni A, Paffetti D, Scotti C, Bazzicalupo M (2000)<br />

Genetic diversity and dynamics of Sinorhizobium meliloti populations nodulating<br />

different alfalfa varieties in Italian soils. Appl Environ Microbiol 66:4785–4789<br />

Di Cello F, Bevivino A, Chiarini L, Fani R, Paffetti D, Tabacchioni S, Dalmastri C (1997)<br />

Biodiversity of a Burkholderia cepacia population isolated from the maize rhizosphere<br />

at different <strong>plant</strong> growth stages. Appl Environ Microbiol 63:4485–4493<br />

Doignon-Bourcier F, Willems A, Coopman R, Laguerre G, Gillis M, De Lajudie P (2000)<br />

Genotypic characterization of Bradyrhizobium strains nodulating small Senegalese<br />

legumes by 16S-23S rRNA intergenic gene spacers and amplified fragment length<br />

polymorphism fingerprint analyses. Appl Environ Microbiol 66:3987–3997<br />

Ellsworth DL, Rittenhouse KD, Honeycutt EL (1993) Artifactual variation in randomly<br />

amplified polymorphic DNA banding patterns. BioTechniques 14:214–217<br />

Excoffier L, Smouse PE, Quattro JM (1992) Analysis of molecular variance inferred from<br />

metric distances among DNA haplotypes: application to human mitochondrial DNA<br />

restriction data. Genetics 131:479–491<br />

Jensen MA, Webster JA, Strauss N (1993) Rapid identification of bacteria on the basis of<br />

polymerase chain reaction-amplified ribosomal DNA spacer polymorphisms. Appl<br />

Environ Microbiol 59:945–952<br />

Kumar S, Tamura K, Nei M (1993) MEGA: Molecular Evolutionary Genetics Analysis,<br />

version 2.0. The Pennsylvania State University, University Park, PA 16802. Freely<br />

downloadable from: http://www.megasoftware.net/<br />

Laguerre G, Mavingui P, Allard MR, Charnay MP, Louvrier P, Mazurier SI, Rigottier-Gois<br />

L,Amarger N (1996) Typing of rhizobia by PCR DNA fingerprinting and PCR-restriction<br />

fragment length polymorphism analysis of chromosomal. Appl Environ Microbiol<br />

62:2029–2036<br />

Louws FJ, Fulbright DW, Stephens CT, de Bruijn FJ (1994) Specific genomic fingerprints<br />

of phytopathogenic Xanthomonas and Pseudomonas pathovars and strains generated<br />

with repetitive sequences and PCR. Appl Environ Microbiol 60:2286–2295<br />

Mengoni A, Bazzicalupo M (2002) The statistical treatment of data and the analysis of<br />

molecular variance (AMOVA) in molecular microbial ecology. Ann Microbiol<br />

52:95–101<br />

Navarro E, Simonet P, Normand P, Bardi R (1992) Characterization on natural populations<br />

of Nitrobacter spp. Using PCR/RFLP analysis of the ribosomal intergenic spacer.<br />

Arch Microbiol 157:107–115<br />

Paffetti D, Scotti C, Gnocchi S, Fancelli S, Bazzicalupo M (1996) Genetic diversity of an<br />

Italian Rhizobium meliloti population from different Medicago sativa varieties. Appl<br />

Environ Microbiol 62:2279–85


28 Analysis of Microbial Population Genetics 565<br />

Paffetti D, Daguin F, Fancelli S, Gnocchi S, Lippi F, Scotti C, Bazzicalupo M (1998) Influence<br />

of <strong>plant</strong> genotype on the selection of nodulating Sinorhizobium meliloti strains<br />

by Medicago sativa. Antonie Van Leeuwenhoek 73:3–8<br />

Picard C, Di Cello F, Ventura M, Fani R, Guckert A (2000) Frequency and biodiversity of<br />

2,4-diacetylphloroglucinol-producing bacteria isolated from the maize rhizosphere<br />

at different stages of <strong>plant</strong> growth. Appl Environ Microbiol 66:948–955<br />

Ponsonnet C, Nesme X (1994) Identification of Agrobacterium strains by PCR-RFLP<br />

analysis of pTi and chromosomal regions. Arch Microbiol 161:300–309<br />

Rohlf FJ (1990) NTSYS-pc. Numerical Taxonomy and Multivariate Analysis System. Version<br />

2.0. Exeter Software, New York<br />

Saitou N, Nei M (1987) The neighbour-joining method: A new method for reconstructing<br />

phylogenetic trees. Molec Biol Evol 4:406–425<br />

Schneider S, Kueffer JM, Roessli D, Excoffier L (1997) ARLEQUIN: a software for population<br />

genetics data analysis. Version 1.1. University of Geneva. Freely downloadable<br />

from http://lgb.unige.ch/arlequin/<br />

Vos P, Hogers R, Bleeker M, Reijans M, van de Lee T, Hornes M, Frijters A, Pot J, Peleman<br />

J, Kuiper M, Zabeau M (1995) AFLP: a new technique for DNA fingerprinting. Nucleic<br />

Acids Res 23:4407–4414<br />

Welsh J, McClelland M (1990) Fingerprinting genomes using PCR with arbitrary<br />

primers. Nucleic Acids Res 18:7213–7218<br />

Williams JGK, Kubelik AR, Livak KJ, Rafalski JA, Tingey SV (1990) DNA polymorphism<br />

amplified by arbitrary primers are useful as genetic markers. Nucleic Acids Res<br />

18:6531–6535<br />

Woods CR Jr, Versalovic J, Koeuth T, Lupski JR (1992) Analysis of relationships among<br />

isolates of Citrobacter diversus by using DNA fingerprints generated by repetitive<br />

sequence-based primers in the polymerase chain reaction. J Clin Microbiol 30:2921–<br />

2929<br />

Wright S (1965) The interpretation of population structure by F-statistics with special<br />

regards to systems of mating. Evolution 19:395–420<br />

Zabeau M, Vos P (1993) Selective restriction fragment amplification: a general method<br />

for DNA fingerprinting. Publication no. 0 534 858 A1. European Patent Office, Munich,<br />

Germany


29 Functional Genomic Approaches for Studies of<br />

Mycorrhizal Symbiosis<br />

Gopi K. Podila and Luisa Lanfranco<br />

1 Introduction<br />

Mycorrhizal fungi, one of the principal biological components of the rhizosphere,<br />

interact with the roots of about 90 % of land <strong>plant</strong>s to form different<br />

types of symbiotic associations (Smith and Read 1997). On the basis of the<br />

colonization pattern of host cells, two main types of mycorrhizas can be identified:<br />

ectomycorrhizas and arbuscular mycorrhizas. In the ectomycorrhizas,<br />

the fungus does not penetrate the host cells, whereas in endomycorrhizas the<br />

fungal hyphae form intracellular structures like coils or arbuscules (Smith<br />

and Read 1997). Mycorrhizal fungi are commonly beneficial due to a wide<br />

network of external hyphae that extend beyond the depletion zone, allowing<br />

host <strong>plant</strong>s to have improved access to limited soil resources. On the other<br />

hand, mycorrhizal fungi receive carbon compounds from host <strong>plant</strong>s to sustain<br />

their metabolism and complete the life cycle and this may lead to reductions<br />

in <strong>plant</strong> growth under some circumstances (Graham and Eissenstat<br />

1998; Graham 2000).<br />

While there is a considerable amount of knowledge based on the ecology<br />

and physiology of mycorrhizal fungi and their uses, the knowledge about cellular<br />

and molecular aspects leading to the growth and the development of a<br />

mycorrhizal fungus as well as the establishment of a functioning symbiosis is<br />

still limited (Harrison 1999; Martin et al. 2001; Podila et al. 2002). The development<br />

of molecular techniques has offered new opportunities: automatic highthroughput<br />

sequencing methods has made it possible to determine the complete<br />

sequence of even eucaryotic genomes. While many ectomycorrhizal<br />

fungal genomes are supposedly of reasonable size (Doudrick 1995),some mycorrhizal<br />

fungi including arbuscular mycorrhizal fungi (AMF), have a large<br />

genome size (Bianciotto and Bonfante 1992; Hosny et al. 1998; for a review<br />

Gianinazzi-Pearson 2001). The presence of repetitive DNA, regulative regions<br />

and introns makes the analysis of genomic sequences relatively complex. The<br />

sequencing of complete genomes for mycorrhizal fungi is still years away until<br />

better methods for application towards mycorrhizal fungi are available.<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


568<br />

Gopi K. Podila and Luisa Lanfranco<br />

An appropriate approach to the study of mycorrhizal fungi is to understand<br />

the molecular process leading to the host recognition, development and<br />

functioning of mycorrhiza through the analysis of expressed sequences. With<br />

the advent of many high throughput techniques that have been successfully<br />

applied to the functional analysis of genes from many organisms, it is now<br />

possible to apply similar strategies to study the various aspects of the mycorrhizal<br />

symbiosis. In this chapter, we describe protocols leading to (1)<br />

expressed sequence tags (EST) and (2) macroarray techniques.<br />

The EST methods allow for rapid identification of sequences through single-run<br />

sequencing of 250–700 bases on randomly picked cDNA clones. Comparisons<br />

with sequence databases frequently allow the assignation of potential<br />

functions to the corresponding gene products. Since its introduction<br />

(Adams et al. 1991), this technique has been successfully applied to several<br />

organisms to provide an overview of the gene repertoire expressed in a particular<br />

stage of development or in a particular tissue (Hofte et al. 1993; Nelson<br />

et al. 1997; Kamoun et al. 1999; Lee et al. 2002). The macroarray or membrane<br />

array methods allows the study of genome-wide expression patterns.<br />

Macroarrays require considerably less RNA for target preparation compared<br />

to microarrays and do not involve costly set-ups.With more refined protocols<br />

macroarrays can be as sensitive as microarrays and also are more easily accessible<br />

for academic laboratories (see Bertucci et al. 1999; Jordan 1998).<br />

Macroarrays are also more suitable for gene expression studies, where only<br />

small subsets of genes (unigene sets) need to be tested for their expression, for<br />

example, genes involved in carbon or nitrogen metabolism, signal transduction,<br />

ion transport, etc.<br />

In this chapter,we describe the experimental procedures for the establishment<br />

of EST collections from mycorrhizal fungi and also macroarray-based techniques<br />

for gene expression profiling of symbiosis process.These procedures can<br />

be applied even in cases of limited amount of biological starting material.<br />

2 Material and Methods<br />

2.1 Equipment<br />

Micro-centrifuge and high-speed centrifuge with proper rotors<br />

Sterile hood<br />

Chemical hood<br />

–20 and –80 °C freezers<br />

VP scientific 384 pin multiblot replicator<br />

Thermal Cycler with a heated lid (Hybaid or Eppendorf Master Cycler or similar)<br />

37 °C shaking incubator<br />

37 °C gravity convection incubator


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 569<br />

Boekel cooler or similar to maintain 16 °C temperature<br />

Gel electrophoresis equipment and power supplies<br />

Hybridization oven (Amersham or Fisher biotech or similar)<br />

Variable volume pipettes<br />

High resolution scanner with transparency adapter<br />

Phosphorimager (Bio-Rad Personal Imager FX)<br />

2.2 Biological Material<br />

Biological material used for the RNA extraction is collected as quickly as possible,<br />

immediately frozen in liquid nitrogen and stored at –80 °C.<br />

2.3 RNA Extraction<br />

The protocol is modified from the one described by Chomoczynski and Sacchi<br />

(1987).<br />

Reagents<br />

Extraction buffer:<br />

4 M Guanidinium thiocyanate<br />

25 mM Na-citrate pH 7<br />

0.5 % Na-laurylsarcosine<br />

0.1 M b-mercaptoethanol (added just prior to use)<br />

2 M Na-acetate pH 4<br />

Phenol water-saturated<br />

Chloroform:isoamylalcohol (49:1, v/v)<br />

Isopropanol<br />

8M LiCl<br />

80 % ethanol<br />

Procedure<br />

Grind the material (100 mg) in liquid nitrogen (with a pestle in a microfuge<br />

tube or in a mortar)<br />

1. Add 200 ml of extraction buffer and place on ice.<br />

2. Add 40 ml of 2 M Na-acetate pH 4, mix thoroughly by inversion.<br />

3. Add 400 ml of phenol and mix by inversion.<br />

4. Add 150 ml of chloroform/isoamylalcohol, mix by inversion and place on<br />

ice 10 min. Centrifuge at 10,000 g for 20 min at 4 °C.<br />

5. Transfer the aqueous phase into a new tube and extract with an equal volume<br />

of chloroform/isoamylalcohol.<br />

6. Transfer the aqueous phase into a new tube and add 1 volume of isopropanol.


570<br />

Gopi K. Podila and Luisa Lanfranco<br />

7. Incubate 2 h at –20 °C.<br />

8. Centrifuge at 10,000 g for 20 min at 4 °C. Remove the supernatant.<br />

9. Wash the pellet with 80 % ethanol.<br />

10. Resuspend in 50–100 ml of sterile water and store at –80 °C.<br />

Note: As an alternative to the RNA extraction protocol explained above, commercial<br />

kits from several companies are available. These usually have no need<br />

of phenol:chloroform manipulations and are relatively rapid. RNA obtained<br />

with these kits often needs to be treated with RNase-free DNase to remove DNA.<br />

DNase Treatment<br />

Incubate the RNA sample in DNase 1¥ buffer (100 mM Tris-HCl pH 7.5,<br />

10 mM MgCl 2 , 1 % BSA) with units of DNase (RNase-free; Promega, Madison,<br />

WI, USA) for 30 min at 37 °C.Add EDTA for 2 mM final concentration. Extract<br />

with an equal volume of phenol/chloroform/isoamylalcohol (25/24/1; v/v/v).<br />

Precipitate the RNA with Na-acetate (0.3 M final concentration) and ethanol<br />

(2.5 volumes). As an alternative, to remove contaminant DNA, a precipitation<br />

with LiCl (final concentration 2 M, overnight at 4 °C) can be performed.<br />

3 RNA Quantification<br />

RNA quantification can be determined with a spectrophotometer (A 260/280)<br />

or fluorometer (Amersham Pharmacia Biotech). The quality of RNA should<br />

be checked on a denaturing agarose gel (Sambrook and Russel 2001) to make<br />

sure that the integrity of RNA is good.<br />

3.1 Construction of a cDNA Library<br />

There are many kits available for the construction of a cDNA library. If there<br />

is plenty of total RNA available to purify poly-A RNA, standard cDNA synthesis<br />

kits can be used such as lambda zap kits (Stratagene, CA, USA). However,<br />

if the availability of the amounts of RNA is limited, it is advisable to use a kit<br />

that can use either a small amount of total RNA or poly-A RNA to synthesize<br />

the cDNA library. Because the amount of tissue and RNA available from mycorrhizal<br />

tissues or mycorrhizal fungi is often limited, we describe here the<br />

method of synthesizing a cDNA library using the SMART cDNA library construction<br />

kit (Clontech, CA, USA). This kit can work on as little as 50 ng of<br />

total RNA since it uses an amplification step after the first strand cDNA synthesis<br />

that compensates for small amounts of starting RNA material.


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 571<br />

3.1.1 cDNA Synthesis (Total volume: 10 ml)<br />

1. Combine the following reagents:<br />

1–3 ml of RNA (50 ng–1 mg)<br />

1 ml SMART III Oligonucleotide (10 mM)<br />

5¢AAGCAGTGGTATCAACGCAGAGTGGCCATTATGGCCGGG 3¢<br />

1 ml CDS III/3¢ PCR Primer (10 mM)<br />

5¢¢ATTCTAGAGGCCGAGGCGGCCGACATG –d(T) 30 N*N 3¢<br />

(N*:A, G or C; N: A, G, C or T)<br />

2. Incubate at 70 °C for 2 min, snap cool the tube on ice for 2 min<br />

3. Add<br />

2 ml x5 First strand buffer<br />

(250 mM Tris-HCl pH 8.3, 30 mM MgCl 2 , 375 mM KCl)<br />

1 ml DTT (20 mM)<br />

1 ml SuperScript II 200 U/ml (Invitrogen, CA, USA)<br />

4. Incubate at 42 °C for 1 h<br />

5. First strand cDNA can be stored at –20 °C for up to 3 months<br />

3.1.2 Long-Distance PCR and Synthesis of Double-Stranded cDNA<br />

1. Combine the following reagents:<br />

2 ml first strand cDNA<br />

80 ml sterile H 2O<br />

10 ml cDNA PCR buffer<br />

2 ml dNTPs (10 mM)<br />

2 ml 5¢PCR Primer (10 mM) 5¢ AAGCAGTGGTATCAACGCAGAGT 3¢<br />

2 ml CDS/3¢ PCR primer<br />

2 ml 50x Advantage cDNA Polymerase Mix (Clontech, CA, USA)<br />

100 ml total volume<br />

2. Run a PCR program on a thermal cycler (Perkin Elmer 2400/9600 with a<br />

heated lid) following these parameters:<br />

1 cycle: 5 °C 20 s<br />

18–26 cycles:<br />

95 °C 5 s<br />

68 °C 6 min<br />

Note: The number of cycles depends on the amount of RNA starting material. If<br />

1 mg of RNA is used, usually 10–15 cycles should be enough. If you start with<br />

0.05–0.25 mg total RNA, 25 cycles are recommended. It is critical not to overcycle<br />

in order to retain the proportion of rare cDNAs. Over-cycling will result in<br />

a disproportionate amplification of abundant cDNAs.<br />

3. Check an aliquot (5 ml) of the PCR product (double-stranded cDNA) on a<br />

1 % agarose gel: a smear of DNA fragments of molecular weight between<br />

0.1 and 4 kbp should appear (Fig. 1). At this stage, the ds cDNA can be<br />

stored at –20 °C up to 3 months.


572<br />

Kbp<br />

5.1 -<br />

2 -<br />

1.3 -<br />

Gopi K. Podila and Luisa Lanfranco<br />

MW dscDNA<br />

Fig. 1. Analysis of double stranded cDNA synthesis products.<br />

Lane MW is molecular weight markers in kilobase<br />

pairs. The bright smear ranging from 4–1 kb in lane<br />

dscDNA shows a good spread of cDNA fragment sizes<br />

3.1.3 Reparation of cDNAs for Ligation: Proteinase K Treatment and SfiI<br />

Digestion<br />

1. Transfer 50 ml of the ds cDNA into a new tube, add<br />

2 ml of proteinase K (20 mg/ml)<br />

Incubate at 45 °C for 20 min.<br />

2. Add 50 ml of H 2O.<br />

3. Mix contents and spin the tube briefly.<br />

4. Incubate at 45 °C for 20 min. Spin the tube briefly.<br />

5. Add 50 ml of deionized H 2O to the tube.<br />

6. Add 100 ml of phenol:chloroform:isoamyl alcohol (25:24:1;v/v/v) and mix<br />

by continuous gentle inversion for 1–2 min.<br />

7. Centrifuge at 10,000 g for 5 min to separate the phases.<br />

8. Remove the top (aqueous) layer to a clean 0.5-ml tube.<br />

9. Add 100 ml of chloroform:isoamylalcohol (24:1, v/v) to the aqueous layer.<br />

Mix by continuous gentle inversion for 1–2 min.<br />

10. Centrifuge at 10,000 g for 5 min to separate the phases.<br />

11. Remove the top (aqueous) layer to a clean 0.5-ml tube.<br />

12. Add 10 ml of 3 M sodium acetate, 1.3 ml of glycogen (20 mg/ml) and 260 ml<br />

of room-temperature 95 % ethanol. Immediately centrifuge at 10,000 g for<br />

20 min at room temperature.<br />

13. Carefully remove the supernatant with a pipette. Do not disturb the pellet.<br />

14. Wash pellet with 100 ml of 80 % ethanol.<br />

15. Air-dry the pellet (~10 min) to evaporate residual ethanol.<br />

16. Add 79 ml of deionized H 2 O to resuspend the pellet.<br />

Note: Proteinase K treatment is necessary to inactivate the DNA polymerase<br />

activity.


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 573<br />

17. SfiI I digestion<br />

Combine the following components in a fresh 0.5-ml tube:<br />

79 ml cDNA (Step 15, above)<br />

10 ml 10x SfiI I buffer<br />

10 ml SfiI I enzyme<br />

1 ml 100x BSA<br />

100 ml total volume<br />

18. Mix well. Incubate the tube at 50 °C for 2 h.<br />

Note: SfiI I-digested cDNA should be fractionated to remove small fragments<br />

which would otherwise compromise the quality of the cDNA library.<br />

3.1.4 cDNA Size Fractionation by CHROMA SPIN-400<br />

1. Label 16 1.5-ml tubes and arrange them in a rack in order.<br />

2. Prepare the CHROMA SPIN-400 column (Clontech, CA, USA) for drip<br />

procedure:<br />

CHROMA SPIN column should be warmed to room temperature before<br />

use. Invert the column several times to completely resuspend the gel<br />

matrix. Remove air bubbles from the column. Use a 1000-ml pipette to<br />

resuspend the matrix gently; avoid generating air bubbles. Remove the<br />

bottom cap and let the column drip.<br />

3. Attach the column to a ring stand. Let the storage buffer drain through the<br />

column by gravity flow until you can see the <strong>surface</strong> of the gel beads in the<br />

column matrix. The top of the column matrix should be at the 1.0-ml<br />

mark on the wall of the column. The flow rate should be approximately 1<br />

drop/40–60 s. The volume of 1 drop should be approximately 40 ml.<br />

4. When the storage buffer stops dripping out, carefully and gently (along<br />

the column inner wall) add 700 ml of column buffer to the top of the column<br />

and allow it to drain out.<br />

5. When this buffer stops dripping (~15–20 min), carefully and evenly apply<br />

~100 ml mixture of SfiI I-digested cDNA mixed with 2 ml xylene cyanol<br />

dye (1 %) to the top-center <strong>surface</strong> of the matrix.<br />

6. Allow the sample to be fully absorbed into the <strong>surface</strong> of the matrix (i.e.,<br />

there should be no liquid remaining above the <strong>surface</strong>).<br />

7. With 100 ml of column buffer, wash the tube that contained the cDNA and<br />

gently apply this material to the <strong>surface</strong> of the matrix.<br />

8. Allow the buffer to drain out of the column until there is no liquid left<br />

above the resin.<br />

9. Place the rack containing the collection tubes under the column, so that<br />

the first tube is directly under the column outlet.<br />

10. Add 600 ml of column buffer and immediately begin collecting singledrop<br />

fractions in tubes #1–16 (approximately 35 ml per tube). Cap each<br />

tube after each fraction is collected. Recap the column after fraction #16<br />

has been collected.


574<br />

Kbp<br />

5.1 -<br />

2 -<br />

0.9 -<br />

Gopi K. Podila and Luisa Lanfranco<br />

MW 1 2 3 4 5 6 7 8 9 10 11 12 13 14<br />

Fig. 2. Analysis of cDNA fractions on an agarose gel. In this particular case, fractions 6,<br />

7, and 8 are collected as they seem to represent a good spread of cDNA sizes. Lane MW<br />

is the molecular weight markers in kilobase pairs.<br />

11. Check the profile of the fractions before proceeding with the experiment<br />

on a 1.1 % agarose/EtBr gel; run 3 ml of each fraction in adjacent wells,<br />

alongside a 1-kb DNA size marker (0.1 mg). Run the gel at 150 V for 10 min<br />

(running the gel longer will make it difficult to see the cDNA bands).<br />

Determine the peak fractions by visualizing the intensity of the bands<br />

under UV (see Fig. 2).<br />

12. Collect the fractions containing cDNA fraction that matches your desired<br />

size distribution. Pool the above fractions in a clean 1.5-ml tube.<br />

13. Add the following reagents to the tube with 3–4 pooled fractions containing<br />

the cDNA: (105–140 ml, respectively):<br />

1/10 vol sodium acetate (3 M; pH 4.8)<br />

1.3 ml glycogen (20 mg/ml)<br />

2.5 vol 95 % ethanol (–20 °C)<br />

14. Mix by gently rocking the tube back and forth.<br />

15. Store the tube at –20 °C overnight.<br />

16. Centrifuge the tube at 10,000 g for 20 min at room temperature.<br />

17. Carefully remove the supernatant with a pipette. Do not disturb the pellet.<br />

18. Briefly centrifuge the tube to bring all remaining liquid to the bottom.<br />

19. Carefully remove all liquid and allow the pellet to air-dry for ~10 min.<br />

20. Resuspend the pellet in 7 ml of deionized H 2O and mix gently. The SfiI Idigested<br />

cDNA is now ready to be ligated to the SfiI I-digested, dephosphorylated<br />

lTriplEx2 vector provided with the kit or the cDNA can be<br />

stored at –20 °C until the ligation step.<br />

3.1.5 Ligation of cDNA to lTriplEx2 vector<br />

Note: The ratio of cDNA to vector in the ligation reaction is a critical factor in<br />

determining transformation efficiency, and ultimately the number of independent<br />

clones in the library. The optimal ratio of cDNA to vector in ligation reactions<br />

must be determined empirically for each vector/cDNA combination. To


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 575<br />

ensure that you obtain the best possible library from your cDNA, set up three<br />

parallel ligations using three different ratios of cDNA to vector, as shown below.<br />

1. Label three 0.5-ml tubes and add the indicated reagents. Mix the reagents<br />

gently; avoid producing air bubbles. Spin tubes briefly to bring contents to<br />

the bottom of the tube.<br />

Ligations using three different ratios of cDNA to phage vector<br />

Component 1st ligation 2nd ligation 3rd ligation<br />

cDNA 0.5 1.0 1.5<br />

Vector (500 ng/ml) 1.0 1.0 1.0<br />

10¥ Ligation buffer* 0.5 0.5 0.5<br />

ATP (10 mM) 0.5 0.5 0.5<br />

T4 DNA Ligase 0.5 0.5 0.5<br />

Deionized H 2O 2.0 1.5 1.0<br />

Total volume (ml) 5.0 5.0 5.0<br />

*x10 ligation buffer: 300 mM Tris-HCl, pH 7.8, 100 mM MgCl 2, 100 mM<br />

DTT<br />

2. Incubate tubes at 16 °C overnight.<br />

3.1.6 Packaging of Ligated cDNA and Preparation of cDNA Library<br />

Perform a separate, l-phage packaging reaction for each of the ligations as<br />

per manufacturer’s instructions.We used Gigapack packaging extracts (Stratagene,<br />

CA, USA) and also MaxPlaq packaging extracts (Epicenter, WI, USA)<br />

with very good success.<br />

1. Thaw three packaging extracts (50 ml per extract) on ice.<br />

2. Immediately after the extracts have thawed, add 5 ml of each ligation mixture<br />

to one tube, mix gently.<br />

3. Incubate at 22 °C for 4 h and add phage buffer (20 mM Tris-HCl, pH 7.4;<br />

100 mM NaCl; 10 mM MgSO 4) to 250 ml and 10 ml of chloroform. Gently mix<br />

well and allow the chloroform to settle down. This packaged mix can be<br />

stored at 4 °C up to 4 weeks.<br />

4. Titer each of the resulting libraries. From the three ligations combined, you<br />

should obtain 1–2x10 6 independent clones.<br />

Note:Ifyou obtained


576<br />

Gopi K. Podila and Luisa Lanfranco<br />

1. To recover the frozen cells, streak a small portion (~5 ml) of the frozen<br />

stock onto an LB agar plate containing the appropriate antibiotic. This is<br />

the primary streak plate. Use LB/tet for XL1-Blue stock plates.<br />

2. Incubate at 37 °C overnight.Wrap plate in Parafilm and store at 4 °C for up<br />

to 2 weeks.<br />

To prepare a working stock plate, pick a single isolated colony from the<br />

primary streak plate and streak it onto another LB/MgSO 4 agar plate<br />

(with antibiotics).<br />

3. Inoculate one colony into 5 ml of LB medium supplemented with 50 ml of<br />

20 % maltose (filter-sterilized) and 50 ml of 1 M MgSO 4 solution. Shake at<br />

160 rpm at 37 °C for 6–9 h or until OD 600=0.6.<br />

4. Dilute each packaged library sample 1000¥, 5000¥, or 10,000¥ with phage<br />

buffer.<br />

5. Add 20 ml of MgSO 4 solution and 2.8 ml of melted top agar to sterile glass<br />

tubes in a sterile laminar flow hood. Cap the tubes and keep them in a<br />

water bath at 50 °C for at least 30 min.<br />

6. Mix 0.1 ml of the diluted phage with 0.1 ml of fresh bacterial cells in a<br />

microfuge tube and allow the phage to adsorb to the cells in an incubator<br />

at 37 °C for 30 min.<br />

7. After incubation add the phage/bacterial mixture to the specified tubes of<br />

top agar in the water bath. Vortex gently to mix the contents and pour<br />

immediately onto LB agar plates. Rotate the plates and gently spread the<br />

top agar uniformly on the <strong>surface</strong> of LB agar.<br />

8. Cool the plates at room temperature for 10 min to allow the top agar to<br />

harden. Invert the plates and incubate them at 37 °C for 6–18 h. Periodically<br />

check the plates to ensure that plaques are developing.<br />

9. Count the plaque forming units (pfu) and calculate the titer of the phage<br />

pfu / ml = number of plaques per plate¥ dilution factor ¥10<br />

Determining the percentage of recombinant clones<br />

10. In lTriplEx2, as in many other l expression vectors, the cloning site is<br />

embedded in the coding sequence for the a-polypeptide of b-galactosidase<br />

(lacZ). This makes it possible to use lacZ a-complementation (Sambrook<br />

and Russel 2001) to easily identify insert-containing phage by<br />

transducing an appropriate host strain (such as E. coli XL1-Blue) and<br />

screening for blue on medium containing IPTG and X-gal.<br />

11. To perform blue/white screening in E. coli XL1-Blue, follow the procedure<br />

for titering on LB/MgSO 4 plates, but add IPTG and X-gal to the melted top<br />

agar before plating the phage + bacteria mixtures. For every 2 ml of<br />

melted top agar, use 50 ml each of the IPTG (10 mM stock) and X-gal (2 %<br />

stock). Aim for 500–1000 plaques/90-mm plate. Incubate plates at 37 °C<br />

for 6–18 h, or until plaques and blue color develop.


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 577<br />

12. The ratio of white (recombinant) to blue (nonrecombinant) plaques will<br />

give a quick estimate of recombination efficiency. A successful ligation<br />

will result in at least 80 % recombinants.<br />

13. Single plaques are isolated in 500 ml of SM buffer+20 ml of chloroform and<br />

stored at 4 °C.<br />

Note: lTriplEx2 phages from isolated single plaques can be converted into<br />

pTriplEx2 plasmids by in vivo excision using the E. coli strain BM25.8 and following<br />

the manufacturer’s instructions (Clontech, CA, USA). A brief protocol is<br />

given below.<br />

4 Conversion Protocol<br />

1. Pick a single, isolated colony from the working stock plate of BM25.8 host<br />

cells (prepared similarly to XL-blue cells as described in phage titration<br />

protocol above) and use it to inoculate 10 ml of LB broth in a 50-ml test<br />

tube or Erlenmeyer flask. Incubate at 31 °C overnight with shaking (at<br />

150 rpm) until the OD 600 of the culture reaches 1.1–1.4.<br />

2. Add 100 ml of 1 M MgCl 2 to the 10-ml overnight culture of BM25.8 (10 mM<br />

final concentration of MgCl 2).<br />

3. Pick each well-isolated plaque from the titration plates and place it in<br />

350 ml of phage buffer in 96-well plates. Mix the contents thoroughly using<br />

an 8 or 12 channel pipette and allow phage to elute at 4 °C overnight.<br />

4. In a deep 96-well plate combine 100 ml of overnight cell culture with 100 ml<br />

of the eluted plaque from each well. (Save the remainder of the eluted<br />

plaques in case you need to repeat the conversion.)<br />

5. Incubate at 31 °C for 30 min without shaking.<br />

6. Add 200 ml of LB broth and cover the plate with the lid provided.<br />

7. Incubate at 31 °C for an additional 1 h with shaking (225 rpm).<br />

8. Using a multichannel pipette transfer 1–5 ml of infected cell suspension<br />

into a 96-well LB/carbenicillin plate to obtain colonies and grow at 31 °C.<br />

9. Pick bacterial growth from each clone and prepare plasmid DNA. For high<br />

throughput processing use Qiagen (Qiagen, CA, USA) or Eppendorf<br />

(Eppendorf, MA, USA) 96 format plasmid Miniprep kits. The isolated plasmid<br />

DNA should be pure enough for direct sequencing. The pTriplEx2<br />

sequencing primers provided may be used with standard ds-DNAsequencing<br />

protocols.<br />

4.1 Evaluation of the Quality of the cDNA Library<br />

To check the quality of the cDNA library two factors must be considered: (1) the<br />

number of primary recombinants (at least 10 5 –10 6 ) and (2) the insert length.<br />

Insert size can be estimated by PCR with primers flanking the insertion site.


578<br />

Gopi K. Podila and Luisa Lanfranco<br />

1. Insert DNAs can be PCR-amplified directly from bacterial colonies with<br />

oligonucleotides designed on sequences flanking the cloning site 5T (5¢<br />

CTCGGGAAGCGCGCCATTGTGTTGG 3¢) and 3T (5¢ ATACGACTCAC-<br />

TATAGGGCGAATTGGCC 3¢). PCR reactions carried out in a final volume<br />

of 50 ml containing 10 mM Tris-HCl pH 8.3, 50 mM KCl, 1.1 mM MgCl 2,<br />

0.01 % gelatin, 200 mM dNTPs, 50 pmol of each primer and 2 U of RedTaq<br />

DNA polymerase (Sigma, St. Louis, MO, USA). The following amplification<br />

program is run in a Hybaid thermal cycler: 3 min at 95 °C (1 cycle); 45 s at<br />

92 °C, 45 s at 55 °C, 2 min at 72 °C (30 cycles).<br />

2. PCR products should be separated by gel electrophoresis (Sambrook and<br />

Russel 2001).<br />

5 Troubleshooting<br />

Possible contaminations by ribosomal sequences (18S and 28S rRNAs might<br />

contain stretches of A that can complement oligo d-T).<br />

Remedial actions: when a sufficient amount of total RNA is available, purify the<br />

poly-A RNA by using oligo d-T affinity columns (Sambrook and Russel 2001).<br />

Inserts with small size<br />

Remedial actions: enrichment of high molecular weight cDNAs through fractionation<br />

into a column.<br />

6 Sequencing Strategies<br />

Single run sequences of 250–700 bases are determined in most cases using an<br />

automated sequencer such as ABI Prism or Beckman CEQ 8 or other<br />

sequencers, whose accuracy has been estimated to be greater than 95 %.<br />

Because of the large number of sequences that needs to be processed in a relatively<br />

short time, it is advisable to outsource the sequencing process (private<br />

companies can do the service for a high number of sequences for a relatively<br />

low price). For people who have access to their own automated sequencers, we<br />

found Big Dye cycle sequencing kit from ABI (ABI, Foster City, CA, USA) or<br />

the DynamicET cycle sequencing kit from Amersham (Amersham Pharmacia<br />

Biotech, Piscataway, NJ, USA) give very good results. Both kits can be used to<br />

scale down the reactions to quarter reactions and still produce very good<br />

sequence reads, and makes it very economical. Since cDNAs are cloned into<br />

the pTriplex vector in a defined orientation, it is advisable to carry out the<br />

sequencing from the 5¢-end first so as to obtain coding region information<br />

from each EST.<br />

Note: If you use quarter reactions, purity of plasmid DNA and quantity are<br />

critical. Sequencing of the 3¢-end could help to identify cDNAs derived from the<br />

same mRNA, but which are truncated at different positions at the 5¢-end.


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 579<br />

6.1 Data Analysis<br />

Sequence similarity of each EST can be detected by BLASTX alignment<br />

(Altschul et al. 1997) of amino acid translations of the six possible open reading<br />

frames against the NCBI (National Center for Biotechnology Information)<br />

databank. Sequences of less than 100 bp should be removed from data analysis<br />

as these are usually not very useful in finding matches. Also, vector<br />

sequences or linker sequences should be filtered from the EST sequence<br />

before performing a similarity analysis. There are many programs such as<br />

DNA sequencher (Gene Codes, Ann Arbor, MI, US) that can automatically<br />

remove vector or linker sequences and clean up the EST sequences before<br />

thorough analysis.<br />

6.2 Sequence Homology Comparisons<br />

Organize all EST sequences into batches in Microsoft Word (Microsoft Corp.,<br />

CA) in FASTA format. Batch nucleotide-protein searches can be done using<br />

BLASTX against all protein databases at GenomeNet http://www.blast.<br />

genome.ad.jp (Japan). cDNA sequences yielding an E value=10 –5 can be considered<br />

to classify known genes or have partial similarity to known genes.<br />

BLAST scores should be further analyzed visually to confirm significant similarity<br />

and not solely determined by numerical value. In order to remove<br />

redundancies from EST sequences analysed, one can use Multalin, a multiple<br />

sequence alignment with hierarchical clustering, http://www.prodes.toulose.<br />

inra.fr/multalin/multalin.html (Corpet 1988). DNA sequences with approximately<br />

90 % identity to another clone may be eliminated as redundant<br />

sequences. The sequence with the most accurate information should be kept<br />

for further analysis. All nonredundants (sequences or ESTs) may be then submitted<br />

to GenBank at the National Center for Biotechnology Information<br />

(http://www.ncbi.nih.gov/Genbank).<br />

6.3 Examples of Expressed Sequence Tag Data Analysis<br />

6.3.1 Expressed Sequence Tags from the Asymbiotic Phase of an<br />

Arbuscular Mycorrhizal Fungus<br />

A cDNA library was constructed from about 100 germinated spores of the<br />

endomycorrhizal fungus Gigaspora margarita (BEG 34). Insert lengths<br />

ranged from 100 to 800 bp with an average of 500 bp. Randomly selected<br />

cDNAs were characterized by sequencing at the 5¢ end and comparison with<br />

databases. BLASTX searches were performed through the NCBI and clones<br />

were grouped on the basis of the E value (Table 1; Fig. 3).


580<br />

Gopi K. Podila and Luisa Lanfranco<br />

Table 1. Selected list of EST clones from G. margarita and their similarity to known<br />

genes and E values to indicate the level of similarity.<br />

Clone BLASTX similarity Species E value<br />

1 14–3-3 Protein Lentinula edodes e –101<br />

2 Endopeptidase Arabidopsis thaliana 1e –88<br />

3 Heat shock protein HSS1 Puccinia graminis 1e –71<br />

4 Cell cycle switch protein Medicago sativa 7e –68<br />

5 Protein involved in phosphate metabolism Saccharomyces cerevisiae 2e –52<br />

6 Cu-Zn Superoxide dismutase Ovis aries 7e –51<br />

7 Pre-mRNA cleavage factor Homo sapiens 4e –49<br />

8 Spliceosome-associated protein Schizosaccharomyces pombe 6e –47<br />

9 Aldehyde dehydrogenase Caenorhabditis elegans 3e –46<br />

10 Polyubiquitine Neurospora crassa 5e –44<br />

11 Histone H4 Styela plicata 3e –39<br />

12 Maleylacetate isomerase 2 Drosophila melanogaster 1e –37<br />

13 Cytidindeaminase Homo sapiens 1e –30<br />

14 Glutathione S transferase Naegleria fowleri 7e –24<br />

15 Ornithine carbamoyltransferase Aspergillus terreus 7e –21<br />

16 Transcriptional factor StuA Aspergillus nidulans 4e –21<br />

17 Subunit G of vacuolar ATP synthase Neurospora crassa 5e –20<br />

18 Isocitrate lyase Dendrobium crumenatum 1e –20<br />

Fig. 3. Distribution of G. margarita EST clones based on the level of BLASTX similarity<br />

ESTs presenting an E value


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 581<br />

Fig. 4. Distribution of G. margarita EST clones based on matches to various organisms<br />

Fig. 5. Distribution of G. margarita EST clones based on similarity to functional groups<br />

using BLASTX analysis<br />

showed similarity to proteins involved in defence responses to stresses. This<br />

result could suggest that the in vitro growth conditions are not favorable and<br />

could mimic a stress situation.<br />

6.3.2 Expressed Sequence Tags from Early Symbiotic Interactions Between<br />

the Ectomycorrhizal Fungus Laccaria bicolor and Red Pine<br />

Over 500 random EST clones from a cDNA library made from pooled RNA<br />

samples from various stages of interaction were sequenced. Out of these, over<br />

400 nonredundant clones were obtained based on sequence analysis. Based on<br />

the BLAST analysis (Altschul et al. 1997), 33 % of the clones showed no significant<br />

similarity to any sequences in the NCBI database. The sequences of the


582<br />

Gopi K. Podila and Luisa Lanfranco<br />

remaining 67 %,however,suggested that they were homologues of genes previously<br />

identified in other systems. These were classified into groups based on<br />

their probable function.About 13 % were related to signal transduction,15 % to<br />

metabolism, 10 % to cellular protein synthesis/processing and turnover, 9 % to<br />

transport and movement of ions/peptides and amino acids, 7 % to structural<br />

proteins, 5 % to RNA/DNA processing, 6 % to transcriptional regulation, and<br />

about 10 % to hypothetical proteins with no known function. In addition, one<br />

clone sequence suggested it was related to apoptosis. The majority of the<br />

matches for L. bicolor ESTs came from animal systems rather than fungi.<br />

7 Macroarrays<br />

7.1 PCR Amplification of cDNA Inserts<br />

cDNA inserts from plasmid templates of EST clones need to be amplified,<br />

purified and quantified before used for printing macroarrays. The following<br />

protocol describes the general methods to obtain PCR products for printing<br />

macroarrays. Due to the large numbers of clones to be amplified, it is best to<br />

use 96-well formatted PCR plates, which will also facilitate printing macroarrays<br />

using either 96 or 384 pin manual or robotic arrayer. Conversely, 8 or 12<br />

PCR strip tubes can also be used for rapid manipulation in setting up the PCR<br />

reactions.<br />

1. For each 96-well plate to be amplified, prepare a PCR reaction mixture containing<br />

the following ingredients:<br />

1000 ml 10¥ PCR buffer<br />

20 ml dATP (100 mM)<br />

20 ml dGTP (100 mM)<br />

20 ml dCTP (100 mM)<br />

20 ml dTTP (100 mM)<br />

5 ml forward primer* (1 mM)<br />

5 ml reverse primer* (1 mM)<br />

100 ml Red-Taq polymerase (1 U/ml)<br />

8800 ml H 2O<br />

Note: * primers used for PCR amplification depend on the vector in which the<br />

cDNA inserts are. Keep all reagents on ice and return the enzyme tube promptly<br />

to the freezer.<br />

2. Label 96-well PCR plates and aliquot 100 ml of PCR reaction mix to each<br />

well. Gently tap plates to insure that no air bubbles are trapped at the bottom<br />

of the wells.<br />

3. Add 1 ml (10 ng) of purified EST plasmid template to each well. Mix well<br />

with pipette.<br />

Note: Mark the donor and recipient plates at the corner near the A1 well to facilitate<br />

correct orientation during transfer of the template. It is important to watch


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 583<br />

that the pipette tips are all submerged in the PCR reaction mix when delivering<br />

the template. Mixing the liquid is easier when multi-channel pipettes are used.<br />

Always use sterile filtered tips to avoid contamination.<br />

4. Replace PCR plate covers and centrifuge the plates at 2700 rpm for 1 min.<br />

5. Place the PCR plates in a thermal cycler (such as Eppendorf Master Cycler)<br />

and run the following cycling program.<br />

Initial denaturation 96 °Cx¥2min<br />

Denaturation 94 °Cx30 s¥30 cycles<br />

Primer annealing 55 °Cx30 s¥30 cycles<br />

Primer extension 72 °Cx2 min¥30 cycles<br />

Final extension 72 °Cx5 min<br />

Note: After PCR, plates can be held at 4 °C while quality controls are performed<br />

on PCR products. To check the quality of the amplified products, analyze 2 ml<br />

each of PCR products on 2% TAE agarose gel as described (Sambrook and Russel<br />

2001). Take a digital photo of the gel on a UV table and store the image for future<br />

reference. The gels should show bands of fairly uniform brightness distributed in<br />

size between 600 and 2000 base-pairs depending on the sizes of cDNAs amplified.<br />

Further computer analysis of such images can be carried out with image<br />

analysis packages to provide a list of the number and size of bands. Ideally this<br />

information can be made available during analysis of the data from hybridizations<br />

involving these PCR products<br />

7.2 Purification and Quantification of PCR Products<br />

1. Spin down PCR reaction plates and then transfer the PCR products (100 ml)<br />

to a Multiscreen filter plate and place the filter on a vacuum manifold filtration<br />

system (e.g., Millipore; Cat # MAVM0960R).<br />

2. Apply a vacuum pressure of approx. 10–15 in. Hg (250–380 mm Hg) for<br />

10 min or until plate is dry.<br />

3. Remove plate from manifold filtration system and add 100 ml of MilliQ<br />

water to each well. Place filter plate on a shaker and shake vigorously for<br />

20 min to resuspend the DNA.<br />

4. Pipette the purified PCR product to a new U-bottom 96 well plate. Seal PCR<br />

storage plates with a plastic cap mat or adhesive foil lid and store at –20 °C<br />

until needed for printing macroarrays.<br />

7.3 Printing of Macroarrays<br />

1. Transfer PCR products to 384-well source plate at a concentration of<br />

100 ng/ml.<br />

Note: The concentration of the source plate is critical, we have shown spot intensity<br />

is directly related to source plate concentrations.


584<br />

Gopi K. Podila and Luisa Lanfranco<br />

2. Include a group of negative and positive control clones with the other<br />

clones to be printed onto the membrane.<br />

3. Denature the PCR products in 5¥ denaturation solution (2 N NaOH, 50 mM<br />

EDTA) diluted to 1x final concentration at 37 °C for 30 min (Jordan 1998).<br />

4. Presoak Hybond N+ nylon membrane filters (Amersham Pharmacia<br />

Biotech, Piscataway, NJ, USA) in 0.1 M NaOH for 1 min, then place on 3-mm<br />

filter paper.<br />

Note: When using a manual arrayer, one layer of filter paper on top of a mouse<br />

pad seems to be an optimal <strong>surface</strong> for printing. Spotting can be done with a<br />

384-pin dot-blot tool (V&P Scientific, San Diego, CA, USA) with 0.9–0.5 mm<br />

diameter flat tip pins. If you are going to print many copies, use a multi-print<br />

replication device (V&P Scientific, San Diego, CA, USA) to give consistent<br />

alignment between membranes and to allow spacing for printing up to 4x384<br />

spots on the same membrane (Schummer et al. 1997).<br />

5. Dip the pins into the 384-well plate containing the denatured DNA. The<br />

pins will deliver ~50 nl of sample yielding spots consisting of approximately<br />

5 ng (the linearity of delivery can be tested by using different concentrations<br />

of PCR products in the same volume).<br />

6. Cross-link membranes for 30 s in UV-crosslinker at optimal setting (Fisher<br />

Scientific, Pittsburgh, PA).<br />

7. Neutralize the array for 5 min in a solution of 0.5 M Tris pH 7.8, 1.5 M NaCl<br />

followed by rinsing in ddH 2O for 5 min.<br />

8. Air dry the arrays on filter paper and wrap in Saran wrap until use.<br />

7.4 Generation of Exponential cDNA Probes from RNA for Macroarrays<br />

and Hybridization Analysis<br />

We found the protocols described by Gonzalez et al. (1998) work very well.We<br />

use components from SMART cDNA synthesis kit (Clontech, CA, USA) for<br />

this purpose.<br />

1. Assemble the following in a 0.2-ml PCR tube<br />

0.5–1 mg RNA 1 ml<br />

10 mM oligo dT primer (CDS from SMART cDNA kit) 1 ml<br />

10 mM of SMART IV oligonucleotide 1 ml<br />

ddH 2O 2ml<br />

Heat the mixture to 70 °C for 2 min, spin briefly and cool at room temperature<br />

to anneal the primers.<br />

2. Add<br />

5x Reverse transcription buffer 2 ml<br />

20 mM DTT 1 ml<br />

10 mM dNTPs 1 ml<br />

Powerscript Reverse Transcriptase 200 U (Clontech, USA) 1 ml<br />

Total volume 10 ml


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 585<br />

Incubate at 42 °C for 1 h.<br />

Add 40 ml of TE buffer (10 mM Tris-HCl, pH 7.2, 1 mM EDTA) to stop the<br />

reaction. All reactions can be done in a PCR machine.<br />

3. To determine the number of cycles required to obtain a population of representative<br />

dscDNAs, 1 ml from each sscDNA reaction should be amplified<br />

following the protocol given below.<br />

sscDNA 1 ml<br />

ddH 2O 41ml<br />

10¥ PCR buffer 5 ml<br />

10 mM PCR anchor primer 1 ml<br />

10 mM dNTPs 1 ml<br />

Advantage Taq 2 U (Clontech, USA) 1 ml<br />

Total volume 50 ml<br />

4. Set up three reactions and amplify for 17, 20, and 23 cycles (95 °C for 15 s,<br />

65 °C for 30 s, and 68 °C for 6 min).<br />

5. Run aliquots from each reaction on an agarose gel and stain with ethidium<br />

bromide. Select the cycle number before the reaction’s plateau.<br />

7.5 Exponential Amplification of the sscDNAs<br />

1. Amplify 2 ml of sscDNA from the RT reactions using the number of cycles<br />

selected from above. Use same conditions for amplification.<br />

2. Clean the PCR products using QIAquick columns (Qiagen, Chatsworth,<br />

CA, USA) into a final volume of 50 ml.<br />

8 Generation of Radiolabeled Probes<br />

1. Denature the dscDNAs prepared above by heating the tube in boiling water<br />

for 5 min and snap-cool the tube on ice.<br />

2. Add to Prime-A-Gene (Promega, Madison,WI, USA) random primer labeling<br />

mixture containing 50 mCi each of 32 P-dATP and 32 Pd-CTP in a 50 ml<br />

reaction volume with twice as much Klenow DNA polymerase than the kit<br />

recommends.<br />

3. Incubate at 37 °C for 2 h and purify the probes using Qiagen nucleotide<br />

removal kit (Qiagen, Chatsworth, CA, USA) as per manufacturer’s instructions.<br />

Note: The double labelling with 32 P-dATP and 32 P-dCTP not only helps in getting<br />

high specific activity targets, but also eliminates problems associated with<br />

labeling GC-rich sequences.


586<br />

Gopi K. Podila and Luisa Lanfranco<br />

9 Hybridization of Macroarrays to Radiolabeled Probes<br />

1. Prehybridize membranes in 10 ml of prehybridization solution (5¥SSC,<br />

10¥Denhardt’s solution, 0.5 % SDS, 100 mg/ml sheared salmon sperm<br />

DNA), at 65 °C for 4 h.<br />

2. Add denatured probe and continue incubation for 22 h at 65 °C.<br />

3. Wash the hybridized membranes successively for 3x5 min in 2xSSC at room<br />

temperature, 2x20 min in 2¥SSC containing 0.5 % SDS, 2x20 min in 1xSSC<br />

containing 0.1 % SDS, and 2x20 min in 0.1xSSC containing 0.1 % SDS, all at<br />

65 °C.<br />

Note: All washes are recommended even if signal intensity seems to drop.<br />

4. Wrap the membranes in Saran wrap and expose to X-ray film (Kodak Biomax<br />

MR) at –80 °C for varying periods (7 h to 3 days).<br />

Note: Exposing membranes to film without an intensifying screen yields clearer<br />

spots.<br />

5. Alternatively, capture the image on a Kodak storage phosphor screen (Eastman<br />

Kodak Company, Rochester, NY, USA) and scan the screens using a<br />

Bio-Rad FX Phosphorimager (Bio-Rad, Hercules, CA) at 100 mM resolution.<br />

Note: It is preferable to use a Phosphorimager as it produces better resolution<br />

and automates the acquisition of data from macroarrays for downstream processing.<br />

10 Data Analysis<br />

Using Phosphorimager:<br />

1. Transfer the raw image data obtained with the phosphorimager imaging<br />

system into a computer.<br />

2. Define each spot on the image by making a grid using QUANTITY ONE<br />

software (Bio-Rad, Hercules, CA, USA).<br />

3. For each image, determine the average pixel intensity (representing the<br />

hybridized DNA) within each spot in each grid square.<br />

4. Generate a data table using QUANTITY ONE and export the data to Excel<br />

worksheet (Microsoft Corporation, Redmond, WA, USA).<br />

5. Calculate background for each membrane by averaging over ten positions<br />

on the image where there are no DNA spots.<br />

6. Calculate net signal for each spot by subtracting the average background<br />

value from the spot intensity.<br />

Note: If any spot values fall below the set threshold value (twofold less than the<br />

background) assign a arbitrary value of 0.1.<br />

7. Probe to probe variance can be filtered out using signal intensities of positive<br />

or negative controls used in the macroarray. In addition, to take into<br />

account experimental variations in specific activity of the cDNA probe


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 587<br />

preparations or exposure time that might alter the signal intensity, normalize<br />

the data obtained from different hybridizations by dividing the intensity<br />

for each spot by the average of the intensities of all the spots present on<br />

the filter, to obtain a centered, normalized value (Eisen et al. 1998).<br />

8. It is important to take average values from multiple experiments to reduce<br />

the variation from experiment to experiment. The final data can be analyzed<br />

using Cluster and Treeview software (http://rana.lbl.gov) to obtain<br />

more normalized data.<br />

Note: You can also use k-means analysis and hierarchical clustering on the net<br />

via the website: http://ep.ebi.ac.uk/EP/EPCLUST/ dedicated to statistical analysis<br />

of gene expression data from macroarrays.<br />

10.1 Data Analysis Autoradiography Images on X-ray Films<br />

Note: If X-ray film is used to capture the image, it is important to do multiple<br />

time exposures to obtain more reliable spot intensities for further analysis. It is<br />

also important not to overexpose the X-ray film where the signal intensities are<br />

not saturated. This will prevent the calculation of any subtle differences in the<br />

expression levels.<br />

1. Scan X-ray films at high resolution (1200x1200 dpi) using a scanner with<br />

transparency adapter and save the images as TIFF files.<br />

Note: These images take up a substantial amount of hard disk space (on average<br />

25–30 MB).<br />

2. Open the image in a quantification program such as One D-scan (Scanalytics<br />

Inc., Fairfax,VA, USA).<br />

3. Scale down the image to fit the screen using the scale and rotate option.<br />

4. Draw a grid over the image using a preset size of 16 rows x 24 columns and<br />

a numbering scheme to match the EST database.<br />

5. Place the grid such that all spots are in the center of each cell. It is possible<br />

to remove segments if artifacts or defects or over-intensity occur on<br />

the image where the neighboring spots may overlap.<br />

6. Calculate the spot intensity values using the volumes option in the analyzing<br />

tool bar.<br />

7. Calculate background using the boundary of each segment option, this<br />

takes a value from each pixel bordering the cell and averages them to yield<br />

the background value.<br />

8. Using the volumes tool create a spread sheet of the data containing the<br />

segment number with it’s background value and volume, along with high<br />

and low values found within each cell.<br />

9. Calculate the volume, or intensity, by adding the intensity of each pixel<br />

within a given segment.<br />

10. Transfer these data to Microsoft Excel (Microsoft Corp., CA) for further<br />

manipulation.


588<br />

Gopi K. Podila and Luisa Lanfranco<br />

11. Calculate a normalized value for each segment by dividing each spot by<br />

the average intensity of all spots on the film to account for probe–probe<br />

variance (Eisen et al. 1998).<br />

12. Correct all values by subtracting the average intensity of the four negative<br />

controls on the membrane.<br />

13. Calculate fold increase or decrease in expression and create another column<br />

for these data.<br />

14. Now the data can be incorporated into a graphical form between the control<br />

and treatments by giving the control a value of 1. Data for a treatment<br />

can then be combined into one spreadsheet along with control data and<br />

imported into GeneCluster at:<br />

http://www-genome.wi.mit.edu/cancer/software/software.html.<br />

This output shows genes that are expressed at approximately the same<br />

ratios, thus creating a cluster of genes that are most likely to be linked in some<br />

biochemical pathway or genes that are relevant to interaction response.<br />

11 Example of Laccaria bicolor Macroarrays<br />

We examined quantitative changes in the expression of ESTs from L. bicolor<br />

using the membrane array technique. We prepared a membrane array consisting<br />

of 384 EST clones selected from L. bicolor interaction cDNA library<br />

(Kim et al. 1999) and probed it with control free-living mycelium mRNA<br />

probes and mRNA probes prepared from various time points of preinfection<br />

stage interaction with red pine. A typical membrane array image obtained<br />

after hybridization with control mRNA and 72-h interaction mRNA probes is<br />

A B<br />

Fig. 6. Example of L. bicolor EST macroarray prepared using the 384 pin manual unit<br />

and expression profiling of interaction-related gene expression in L. bicolor. Macroarrays<br />

were printed using 0.9-mm diameter pins containing 384 ESTs and hybridized to<br />

probes prepared from RNA from free living L. bicolor (A) or from 72 h interaction (B).<br />

Image obtained from X-ray films exposed to the hybridized membrane. Image is captured<br />

using Saphir high-resolution scanner (Linotype-Hell, Heidelberg Inc. NY. USA) at<br />

1200x1200 dpi. Quantification of signal intensities from spots and gene expression levels<br />

is determined using Mac 1-D software (Scanalytics,VA, USA)


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 589<br />

Fig. 7. Histogram showing<br />

clustering of genes from<br />

macroarray analysis based<br />

on levels of expression. The<br />

interaction (72 h)-related<br />

expression is shown on the<br />

x-axis and the number of<br />

genes that are upregulated<br />

at a given expression ratio<br />

is shown on the y-axis.Only<br />

genes that are upregulated<br />

are shown from the<br />

macroarray in Fig. 6A. For<br />

identification of some of<br />

the genes upregulated by<br />

interaction see Table 2<br />

Fig. 8. Scatter plot analysis of interaction-specific genes between free-living Laccaria<br />

bicolor and L. bicolor interacted 72 h with Pinus resinosa seedling roots. For each gene,<br />

transcript levels were calculated for the free-living mycelium and the mycelium that<br />

interacted with red pine seedling root signals. Solid lines indicate an expression level of<br />

onefold or above the free-living mycelium, dashed lines 2.5–6-fold increase and dotted<br />

line eightfold or higher levels of expression. Only nonredundant clones are represented<br />

in the plot. Selected clones that showed significant differential expression are highlighted


590<br />

Gopi K. Podila and Luisa Lanfranco<br />

Table 2. Differential expression of selected interaction clones from L. bicolor. Clones are<br />

selected from macroarray analysis. The E ratio was calculated by comparing the expression<br />

levels in the interacted fungal tissue with those in the free-living fungus using<br />

macroarrays<br />

GenBank no. E value Database match Expression<br />

ratio<br />

BI094576 3e –69 BiP protein (Aspergillus nidulans) 4.10<br />

BI094582 2e –69 PF6.2.1 (Laccaria bicolor) 8.00<br />

BI094583 2e –48 a-tubulin (Ustilago maydis) 4.51<br />

BI094587 1e –10 Homeobox genes Hox-2.6 (Mus musculus) 3.30<br />

BI094592 2e –10 PEP carboxykinase (Mus musculus) 4.33<br />

BI094601 2e –63 LbAut7 (L. bicolor) 3.73<br />

BI094606 3e –09 b-importin (Schizosaccharomyces pombe) 4.43<br />

BI094612 2e –35 Malate synthase (L. bicolor) 3.82<br />

BI094615 9e –36 TEF (EF1a) (Schizophyllum commune) 3.61<br />

BI094619 1e –06 IRS 1-like protein (Xenopus laevis) 3.13<br />

BI094621 1e –31 Ras related protein (L. bicolor) 3.81<br />

BI094622 3e –86 AAD (Phanerochate. chrysosporium) 2.61<br />

BI094623 1e –06 LZK protein kinase (Homo sapiens) 2.90<br />

BI094629 7e –13 Lactonohydrolase (Fusarium oxysporum) 3.91<br />

BI094632 9e –27 E-MAP-115 (H. sapiens) 4.21<br />

BI094635 1e –26 SUG1 subunit 8 (S. cerevisiae) 4.61<br />

BI094639 1e –16 Septin Spn3 (S. pombe) 3.92<br />

BI094653 5e –62 Rho GTPase (S. cerevisiae) 3.21<br />

BI094657 1e –10 Clathrin adapter protein (A. thaliana) 2.08<br />

BI094660 6e –21 AcetylCoA acetyltransferase (L. bicolor) 4.25<br />

BI094667 1e –20 b-transducin (S. pombe) 4.11<br />

BI094676 2e –37 Chitin synthase I (U. maydis) 3.71<br />

shown in Fig. 6.An E ratio that indicates the relative increase in the expression<br />

of each gene in the interaction over the free-living state is used to quantitate<br />

differential expression. There is an overall increase in levels of expression of<br />

several clones tested (Fig. 7). The scatter plot of the normalized data from signal<br />

analysis of the membranes is presented in Fig. 8, which shows global<br />

changes in the expression of interaction related genes. Levels of expression of<br />

selected genes from 72-h interaction are listed in Table 2.<br />

12 Conclusions<br />

The EST and macroarray approaches provide efficient tools for mycorrhizal<br />

symbiosis research. These approaches have the resolution and ability to<br />

obtain a more comprehensive view of various stages of mycorrhiza development<br />

or treatment effects due to nutritional changes or differences due to host


29 Functional Genomic Approaches for Studies of Mycorrhizal Symbiosis 591<br />

responses. In addition, since they require a relatively modest budget, compared<br />

to genome sequencing or microarray-based methods, they are easily<br />

accessible for many academic research groups. With increased use of these<br />

techniques using a variety of mycorrhizal symbiosis models, data can be<br />

exchanged and compared between different laboratories and eventually will<br />

provide a platform to understand the key players (genes) that are markers for<br />

ectomycorrhizal or AM fungal symbioses. In the last couple of years, several<br />

laboratories have begun using these approaches to unravel the mycorrhizal<br />

symbiosis (Martin et al. 2001; Voiblet et al. 2001; Podila et al. 2002; Polidori et<br />

al. 2002).<br />

References and Selected Reading<br />

Adams MD, Kelley JM, Gocayne JD, Dubnick M, Polymeropoulos M, Xiao H, Merril C,<br />

Wu A, Olde B, Moreno R (1991) Complementary DNA sequencing, expressed<br />

sequence tags and humane genome project. Science 252:1651–1656<br />

Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ (1997)<br />

Gapped Blast and PSI-BLAST: a new generation of protein database search programs.<br />

Nucl Acids Res 25:3389–3402<br />

Bertucci F, Bernard K, Loriod B, Chang YC, Granjeaud S, Birnbaum D, Nguyen C, Peck K,<br />

Jordan BR. (1999) Sensitivity issues in DNA array-based expression measurements<br />

and performance of Nylon microarrays for small samples. Hum Mol Genet 8(9):1715–<br />

22<br />

Bianciotto V, Bonfante P (1992) Quantification of the nuclear content of two arbuscular<br />

mycorrhizal fungi. Mycol Res 96:1071–1076<br />

Chomoczynski P, Sacchi N (1987) Single-step method of RNA isolation by acid guanidinium<br />

thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156–159<br />

Corpet F (1988) Multiple sequence alignment with hierarchical clustering. Nucleic Acids<br />

Res 16:10881–10890<br />

Doudrick RL, Raffle VL, NelsonCD, Fournier GR (1995) Genetic analysis of homokaryons<br />

from a basidiome of Laccaria bicolor using random amplified polymorphic<br />

DNA (RAPD) markers. Mycol Res 99:1361–1365<br />

Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster analysis and display of<br />

genome-wide expression patterns. Proc Natl Acad Sci USA 95:14863–14868<br />

Gianinazzi-Pearson V, van Tuinen D, Dumas-Gaudot E, Dulieu H (2001) Exploring the<br />

genome of Glomalean fungi. In: Hock B (ed) The Mycota, vol IX. Fungal Associations.<br />

Springer, Berlin Heidelberg New York, pp 3–17<br />

Gonzalez P, Zigler S, Epstein DL, Borras T (1998) Identification and isolation of differentially<br />

expressed genes from very small tissue samples. Biotechniques 26:884–892<br />

Graham JH (2000) Assessing costs of arbuscular mycorrhizal symbiosis in agroecosystems.<br />

In: Podila GK, Douds DD (eds) Current advances in mycorrhizae research. APS<br />

Press, St. Paul, MN, pp 127–140<br />

Graham JH, Eissenstat DM (1998) Field evidence for carbon costs of citrus mycorrhizas.<br />

New Phytol 140:103–110<br />

Harrison MJ (1999) Molecular and cellular aspects of the arbuscular mycorrhizal symbiosis.<br />

Ann Rev Plant Physiol Plant Mol Biol 50:361–389<br />

Hofte H, Desprez T, Amselem J, et al. (1993) An inventory of 1152 expressed sequence<br />

tags obtained by partial sequencing of cDNA from Arabidopsis thaliana. Plant J<br />

4:1051–1061


592<br />

Gopi K. Podila and Luisa Lanfranco<br />

Hosny M, Gianinazzi-Pearson, V, Dulieu H (1998) Nuclear DNA contents of 11 fungal<br />

species in Glomales. Genome 41:422–429<br />

Jordan BR (1998) Large-scale expression measurement by hybridization methods: from<br />

high-density membranes to “DNA chips”. J Biochem (Tokyo) 124(2):251–258<br />

Kamoun S, Hraber P, Sobral B, Nuss D, Govers F (1999) Initial assessment of gene diversity<br />

for the oomycete pathogen Phytophthora infestans based on expressed sequence<br />

tags. Fungal Genet Biol 28:94–106<br />

Lee SH, Kim BG, Kim KJ, Lee JS, Yun DW, Hahn JH, Kim GH, Lee KH, Suh DS, Kwon ST,<br />

Lee CS, Yoo YB (2002) Comparative analysis of sequences expressed during the liquid-cultured<br />

mycelia and fruit body stages of Pleurotus ostreatus. Fungal Genet Biol<br />

35(2):115–134<br />

Martin F, Duplessis S, Ditengou F, Lagrange H,Voiblet C, Lapeyrie F (2001) Development<br />

of cross talking in the ectomycorrhizal symbiosis: Signals and communication genes.<br />

New Phytol 151:145–154<br />

Nelson MA, Kang S, Braun EL, Crawford ME, Dolan PL, Leonard PM, Mitchell J, Armijo<br />

AM et al. (1997) Expressed sequences from conidial, mycelial and sexual stages of<br />

Neurospora crassa. Fungal Genet Biol 21:348–363<br />

Podila GK, Zheng, J, Balasubramanian S, Sundaram S, Hiremath S, Brand J, Hymes M<br />

(2002) Molecular interactions in ectomycorrhizas: identification of fungal genes<br />

involved in early symbiotic interactions between Laccaria bicolor and red pine. Plant<br />

Soil 244:117–128<br />

Polidori E,Agostini D, Zeppa S, Potenza L, Palms F, Sisti D, Stocchi V (2002) Identification<br />

of differentially expressed cDNA clones in Tilia platyphyllos – Tuber borchii ectomycorrhizae<br />

using a differential screening approach. Mol Gen Genomics 266:858–864<br />

Sambrook J, Russel DW (2001) Molecular Cloning. A Laboratory Manual. 3rd Edition.<br />

Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York<br />

Schummer M, Ng, WL, Nelson PS, Bumgarner RB, Hood L (1997) A simple high-performance<br />

DNA arraying device for comparative expression analysis of a large number of<br />

genes. BioTechniques 23:1087–1092<br />

Smith SE, Read DJ (1997) Mycorrhizal symbiosis, 2nd edn. Academic Press, London<br />

Voiblet C, Duplessis D, Encelot N, Martin F (2001) Identification of symbiosis-regulated<br />

genes in Eucalyptus globulus-Pisolithus ectomycorrhiza by differential hybridization<br />

of arrayed cDNAs. Plant J 25:181–191


30 Axenic Culture of Symbiotic Fungus<br />

Piriformospora indica<br />

Giang Huong Pham, Rina Kumari, Anjana Singh, Rajani Malla,<br />

Ram Prasad, Minu Sachdev, Michael Kaldorf, François Buscot,<br />

Ralf Oelmüller, Rüdiger Hampp, Anil Kumar Saxena,<br />

Karl-Heinz Rexer, Gerhard Kost and Ajit Varma<br />

1 Introduction<br />

A large number of media compositions are available in the literature for the<br />

cultivation of various groups of fungi, but almost no literature is available for<br />

axenic cultivation of symbiotic fungi. In this chapter, we have made efforts to<br />

provide the documentary evidence for growth and multiplication of Piriformospora<br />

indica (see Chap. 15, this Vol. for characteristic features of the fungus).<br />

This new fungus named P. indica, due to its characteristic spore morphology,<br />

improves the growth and overall biomass production of different<br />

<strong>plant</strong>s, herbs and trees, etc., and can easily be cultivated on a number of complex<br />

and synthetic media (Varma et al. 1999, 2001; Singh An et al. 2003a, b).<br />

Significant quantitative and morphological changes were detected when the<br />

fungus was grown on different nutrient compositions with no apparent negative<br />

effect on <strong>plant</strong>s. It is relevant to mention here that different media can be<br />

used to understand the morphological and functional properties, or to test<br />

possible biotechnological applications.<br />

2 Morphology<br />

Young mycelia were white and almost hyaline, but inconspicuous zonations<br />

were recorded in other cultures. The mycelium was mostly flat and submerged<br />

into the substratum. Hyphae were thin-walled and of different diameters<br />

ranging from 0.7 to 3.5 mm. The hyphae were highly interwoven, often<br />

adhered together and gave the appearance of simple intertwined cords. The<br />

hyphae often showed anastomoses and were irregularly septated. They often<br />

intertwined and overlapped each other. In older cultures and on the root <strong>surface</strong>,<br />

hyphae were often irregularly inflated, showing a nodose to coralloid<br />

Plant Surface Microbiology<br />

A.Varma, L. Abbott, D. Werner, R. Hampp (Eds.)<br />

© Springer-Verlag Berlin Heidelberg 2004


594<br />

Giang Huong Pham et al.<br />

Fig. 1. An overall view of P. indica, grown on solidified MYP medium for 7 days. Note<br />

the distinct hyphal coils (h) and pear-shaped chlamydospores (c) Bar = 20 µm. By courtesy<br />

of Oliver Blechert<br />

Fig. 2. P. indica colonized<br />

maize root segment<br />

covered by numerous<br />

chlamydospores on<br />

the <strong>surface</strong> and scattering<br />

away from the root.<br />

Enlarged view of<br />

chlamydospores showing<br />

nuclei. Chlamydospores<br />

were stained<br />

with DAPI and observed<br />

in epifluorescence. Different<br />

optical planes<br />

were assembled in one<br />

picture using the<br />

IMPROVISION software<br />

package (IMPROVI-<br />

SION, Govenny, UK)


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 595<br />

shape and granulated dense bodies were observed. Many cells contained more<br />

than one nucleus. Chlamydospores were formed from thin-walled vesicles at<br />

the tips of the hyphae. The chlamydospores appeared singly or in clusters and<br />

were distinctive due to their pear-shaped appearance (Fig. 1). The chlamydospores<br />

were (14-) 16–25 (-33) mm in length and (9-) 10–17 (-20) mm in<br />

width. Figure 2 shows the maize root colonization. The cytoplasm of the<br />

chlamydospores was densely packed with granular material and usually contained<br />

8–25 nuclei (Fig. 2, inset).Very young spores had thin, hyaline walls. At<br />

maturity, these spores had walls up to 1.5 mm thick, which appeared two-layered,<br />

smooth and pale yellow. Neither clamp connections nor sexual structures<br />

could be observed.<br />

3 Taxonomy of the Fungus<br />

Different kinds of substrates were tested to induce sexual development, such<br />

as young and mature leaves of Cynodon dactylon and pollen grains, oat meal,<br />

potato, carrot or tomato dextrose agar and soil-on-agar culture methods. No<br />

apparent adverse affect was seen on cultivation in light. It is not necessary to<br />

grow the fungus in the dark. Growth under light and dark conditions did not<br />

promote sexuality. May be the fungus is heterothallic in nature and one has to<br />

work for compatible strains. Since all these efforts did not lead to the desired<br />

results, there were only a few features to characterize the fungus morphologically<br />

and group it according to the classical species concept. In order to obtain<br />

more information about the systematic position of the new fungus, the ultrastructures<br />

of the septal pore and the cell wall were examined. The cell walls<br />

were very thin and multilayered structures. The septal pores consisted of<br />

dolipores with continuous parenthosomes. The dolipores were very prominent,<br />

with a multilayered cross wall and a median swelling mainly consisting<br />

of electron-transparent material. The electron-transparent layer of the cross<br />

walls extended deep into the median swellings, but did not fan out. In median<br />

sections of the septal pores, the parenthosomes were always straight and had<br />

the same diameter as the corresponding dolipore. Parenthosomes were flat<br />

discs without any detectable perforation. The parenthosomes consisted of an<br />

electron-dense outer layer, which showed an inconspicuous dark line in the<br />

median region. The parenthosomes were in contact with the ER membranes,<br />

which were mostly found near the dolipore (Verma et al. 1998).<br />

The ultrastructural data proof that P. indica is a menber of the Hymenomycetes<br />

(Basidiomycota). Studies on the moleclar phylogeny will help to<br />

reveal the closest relatives of this species (Fig. 3).<br />

Interestingly, immunological characterization showed a strong cross-reactivity<br />

with the members of Zygomycota (Glomerales) instead of species of<br />

Basidiomycota (Table 1). This aspect needs further critical appraisal.


596<br />

Giang Huong Pham et al.<br />

Table 1. Cross-reactivities of polyclonal antisera raised against P. indica (total hyphal<br />

homogenate) as determined by ELISA. Optical density (OD 405 nm ) values are given as the<br />

mean of three replicates after correction of control (OD 405 nm )±SD. Statistical analysis<br />

was done by ANOVA. (n.d., not detectable)<br />

Antigens OD 405 nm Source<br />

1:1600<br />

Piriformospora indica<br />

Nonmycorrhizal fungi<br />

0.49±0.005 Ajit Varma, JNU, New Delhi<br />

Agaricus bisporus 0.08±0.002 AK Sarbhoy, IARI, New Delhi<br />

Beauvaria sp. 0.003±n.d. AK Sarbhoy, IARI, New Delhi<br />

Candida albicans 0.11±0.004 R Prasad, JNU, New Delhi<br />

Cladosporium sp. 0.004±n.d. AK Sarbhoy, IARI, New Delhi<br />

Cunninghamella echinulata 0.03±0.001 G Kost, Marburg, Germany<br />

Fusarium solani 0.03±0.002 AK Sarbhoy, IARI, New Delhi<br />

Rhizoctonia bataticola 0.04±0.002 G Kost, Marburg, Germany<br />

Rhizoctonia solani 0.013±0.001 A K Sarbhoy, IARI, New Delhi<br />

Rhizopus sp. 0.06±0.001 AK Sarbhoy, IARI, New Delhi<br />

Saccharomyces cerevisiae 0.17±0.020 R Prasad, JNU, New Delhi<br />

Schizophyllum commune 0.005±0.004 G Kost, Marburg, Germany<br />

Sclerotinia sclerotiorum 0.16±0.006 AK Sarbhoy, IARI, New Delhi<br />

Sclerotium solani 0.05±0.001 G Kost, Marburg, Germany<br />

Ustilago maydis 0.08±0.007 AK Sarbhoy, IARI, New Delhi<br />

Ectomycorrhizal fungi<br />

Amanita muscaria 0.18±0.007 T Satyanarayana, South Campus, Delhi University<br />

Lactarius torminosus 0.03±0.004 Erika Kothe, Jena, Germany<br />

Lentinus edodes 0.02±0.001 T Satyanarayana, South Campus, Delhi University<br />

Paxillus involutus 0.02±0.001 T Satyanarayana, South Campus, Delhi University<br />

Pisolithus tinctorius 0.15±0.007 Erika Kothe, Jena, Germany<br />

Rhizopogon roseolus 0.12±0.019 T Satyanarayana, South Campus, Delhi University<br />

Rhizopogon vulgaris 0.01±0.001 T Satyanarayana, South Campus, Delhi University<br />

Suillus variegatus<br />

Endomycorrhizal fungi<br />

0.003±0.004 Erika Kothe, Jena, Germany<br />

Gigaspora margarita 0.41±0.005 Alok Adholeya, TERI, New Delhi<br />

Gi. gigantia 0.46±0.002 KVBR Tilak, IARI, New Delhi<br />

Glomus caledonium 0.20±0.039 François Buscot, Jena, Germany<br />

G. coronatium 0.07±0.011 François Buscot, Jena, Germany<br />

G. geosporura 0.16±0.019 François Buscot, Jena, Germany<br />

G. intraradices 0.003±0.003 François Buscot, Jena, Germany<br />

G. lamellosum 0.02±0.004 François Buscot, Jena, Germany<br />

G. mosseae 0.15±0.010 François Buscot, Jena, Germany<br />

G. mosseae 376 0.10±0.027 François Buscot, Jena, Germany<br />

G. proliferum 0.24±0.023 François Buscot, Jena, Germany<br />

Scutellospora gilmorei<br />

AMF-like<br />

0.40±0.002 Ajay Shanker, JNU, New Delhi<br />

Sebacina vermifera var sensu 0.39±0.049 Karl-Hein Rexer, Marburg, Germany<br />

Sebacina sp. 0.23±0.013 Karl-Hein Rexer, Marburg, Germany<br />

Statistical analysis of the data shows the P values, which are significant (P


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 597<br />

Fig. 3. An overall view of the molecular taxonomic position of P. indica (modified<br />

after Schüßler et al. 2001)<br />

4 Chlamydospore Formation and Germination<br />

The fungus produces chlamydospores at the apex of hyphae, which were<br />

mostly irregular undulated in shape. These chlamydospores can be easily germinated<br />

on various synthetic media (Verma et al. 1998). On solidified agar<br />

(2 %) medium, a tendency for cluster formation of chlamydospores was<br />

observed. Temperature (low to high and/or vice versa), pH (alkaline to acid or<br />

vice versa), and shock treatment also induced excessive sporulation. These<br />

spores were viable for over a year when preserved at room temperature. Loss<br />

in viability of the dormant spores was the least when germinated after 1 year.<br />

Dormant spores germinated within 1 day of their placement on nutrient agar<br />

medium and incubated at 40 °C under high humidity (>90 %). The first step of<br />

germination was the formation of germ tubes at the protruded zone of the<br />

spore, followed by hyphal emergence. Most of the nuclei followed the hyphae<br />

and seldom were one or two nuclei left behind in the spore. Soon branching<br />

appeared with a short and long branch (Fig. 4).<br />

5 Cultivation<br />

Fungi are heterotrophic for carbon compounds and these serve two essential<br />

functions in fungal metabolism. The first function is to supply the carbon<br />

needed for the synthesis of compounds which comprise living cells. Proteins,


598<br />

Giang Huong Pham et al.<br />

nucleic acids, reserve and food materials, etc., would be included here. Second,<br />

the oxidation of carbon compounds produces appreciable amounts of<br />

energy. Fungi can utilize a wide range of carbon sources such as monosaccharides,<br />

disaccharides, oligosaccharides, polysaccharides, organic acids and<br />

lipids. Carbon dioxide can be fixed by some fungi, but cannot be used as an<br />

exclusive source of carbon for metabolism. P. indica can be successfully cultivated<br />

on a wide range of synthetic solidified and broth media, e.g., MMN1/10,<br />

modified aspergillus, M4N, MMNC, MS, WPM, MMN, Malt-Yeast Extract,<br />

MYP, PDA and aspergillus (Fig. 5). Among the tested media, aspergillus (Kae-<br />

Fig. 4. Chlamydospores of P.<br />

indica. a Germinating chlamydospore<br />

showing initial branching<br />

after 12 h, b mature chlamydospores<br />

were germinated on a<br />

glass slide coated with thin<br />

nutrient agar, photographed<br />

after 24 h, c scattered spores and<br />

thin, irregular, undulating<br />

hyphae


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 599<br />

fer 1977) was the best. However, other media were helpful in carrying out several<br />

physiological and molecular experiments (see Chap. 15, this Vol.).<br />

Figure 6 shows typical growth on solidified aspergillus medium after 28 days.<br />

Rhythmic growth was often recorded. The mycelium stopped its growth for<br />

some time and produced a large number of chlamydospores of different<br />

dimensions. After 24–48 h, the mycelium started its growth again, producing<br />

normal amount of chlamydospores. This resulted in the formation of rythmic<br />

rings. The physiological reason for this phenomenon is not yet known,<br />

although this tendency has been recorded for several other members of<br />

Basidiomycetes. The fungus grows profusely upon shaking broth aspergillus<br />

medium. The temperature range of the fungal growth is 25–35 °C; the optimum<br />

temperature and pH being 30 °C and 5.8 (4.8–6.8), respectively. Figure 7<br />

gives a view of the cultivation on broth media. Colonies were large and small<br />

depicting sea urchin-like radial growth. The maximum <strong>surface</strong> growth was<br />

recorded after 10 days. The colony diameter is indicated in Fig. 8. The fungal<br />

biomass is indicated in Table 2. The optimum growth was recorded after 5<br />

Fig. 5. P. indica was<br />

grown on the following<br />

solidified media. a MS,<br />

b WPM, c MMN, d M4N,<br />

e PDA, f aspergillus


600<br />

Giang Huong Pham et al.<br />

days with a gradual decrease in fresh and dry biomass after prolong incubation.<br />

Linear growth of the fungus on different solidified agar media is represented<br />

in Table 3. On modified Melin-Norkrans (MMN) medium sparsely<br />

running hyaline hyphae on the agar <strong>surface</strong> were seen, while on Potato Dextrose<br />

Agar deep furrows with strong adhesion to the agar <strong>surface</strong> were apparent.<br />

This sharp mode of growth was not observed when fortified with malt<br />

extract and normal aspergillus medium. In contrast to aspergillus medium,<br />

shaking conditions on MMN broth medium invariably inhibited the growth.<br />

The explanation for this observation is not known. Fungal growth acidifies<br />

the medium within 10 days to pH 4.4. Buffered medium prevented the reduction<br />

of pH (Table 4). 2-(N-morpholine) ethane sulfonic acid (MES) in the<br />

range of 25–100 mM was used.<br />

6 Carbon and Energy Sources<br />

Fig. 6. An overall view of P.<br />

indica grown on solidified<br />

aspergillus medium. Inset<br />

shows enlarged view of a small<br />

portion. On an agar concentration<br />

of 2 % w/v concentric rings<br />

often appeared (arrows) indicating<br />

the rhythmic growth of<br />

the fungus. The black arrows<br />

point on the regions with slow<br />

growth and high amount of<br />

chlamydospores, the white<br />

arrows point on thin mycelial<br />

mats resulting from fast growth<br />

of the hyphae<br />

Individual sugars were uniformly added to the minimal broth at a rate of 1.0 %<br />

(w/v) in all treatments.They were included in the medium separately after sterilization.<br />

In all the sugar-supplied media, growth was better than the control<br />

(Table 5). There were not many changes in the growth except for rafinose and<br />

fructose.There were no changes in the color of the mycelium.Good growth was<br />

recorded in media containing maltose followed by xylose, sucrose, rhamnose,<br />

arabinose, glucose, lactose and mannose, respectively. The final pH was not<br />

altered significantly, but was lower than that of the control (Table 5).<br />

In a further study, fungal growth was best when glucose (1 % w/v) was<br />

used as a carbon source as compared to sucrose, and followed by fructose. A


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 601<br />

Table 2. The data represent an average of P. indica biomass of 5 replicates grown in 100<br />

ml aspergillus broth medium in 250 ml capacity Erlenmeyer flasks. Incubation was done<br />

on a rotary shaker (GFL 3.19, Germany) at 144 rpm at 30 °C<br />

Days Biomass (g)<br />

Fresh Dry<br />

5 3.67±0.84 0.06±0.02<br />

7 2.99±0.38 0.06±0.01<br />

10 2.39±0.01 0.07±0.01<br />

Fig. 7. Growth of P. indica on aspergillus broth medium under constant shaking condition<br />

at 25 °C for 7 days. Colonies of different developmental stages were shown. Mature<br />

colonies have the appearance of sea urchins<br />

5 7 10<br />

5 7 10<br />

LM A LM A LM A<br />

3.6 ± 0.15 10.2 5.4 ± 0.36 22.9 7.5 ± 0.16 43.9<br />

Fig. 8. A comparative linear growth of P. indica on aspergillus solidified medium. Measurements<br />

were made after 5, 7 and 10 days, respectively. Incubation was conducted in<br />

dark at 25 °C. Parameter selected was the diameter of 5 replicates of the linear measurement<br />

(LM). Readings are given in cm standard deviation and area (A) on agar medium.<br />

Statistical analysis of the data showed P


602<br />

Giang Huong Pham et al.<br />

Table 3. Comparative linear growth of P. indica on different<br />

solidified agar media. The data represent an average<br />

colony diameter of five replicates, measured after<br />

5 days of incubation<br />

Media Linear measurement of the growth (cm)<br />

MMN 4.2±0.05<br />

M4N 2.8±0.09<br />

PDA 3.5±0.11<br />

aspergillus 3.6±0.15<br />

Table 4. Change of pH of the aspergillus broth medium incubated with P. indica<br />

Medium conditions (pH) Incubation days<br />

0 3 5 7 10<br />

Unbuffered 6.5 6.0 5.7 5.1 4.4<br />

Buffered 6.5 6.5 6.5 6.5 6.3<br />

Initial pH was adjusted to 6.5. 25–100 mM MES was used as buffering agent<br />

Table 5. End pH and biomass of P. indica grown on minimal<br />

aspergillus broth medium containing different sugars (each 1 %<br />

w/v)<br />

Sugars End pH Biomass (mg/10 ml)<br />

Control (no addition) 5.18 7.5<br />

Glucose 4.39 10.0<br />

Fructose 5.25 8.0<br />

Maltose 4.46 12.0<br />

Rhamnose 4.60 10.3<br />

Mannose 4.30 9.5<br />

Lactose 4.44 10.0<br />

Sucrose 4.39 10.0<br />

Xylose 4.31 11.0<br />

Arabinose 4.28 10.0<br />

Raffinose 4.43 9.0<br />

One agar disc (1 cm in diameter loaded with hyphae and<br />

chlamydospores) was transferred to individual test tubes containing<br />

10 ml minimal broth. Sterile sugar solution (microsyringe-filtered,<br />

0.22 mm Schleicher & Schuell) was included. Incubation<br />

was done under constant shaking conditions (GFL, 3026,<br />

Germany) for 7 days at 25 °C. Fungal biomass was removed and<br />

end pH was measured


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 603<br />

Table 6. Growth of P. indica on unbuffered aspergillus broth medium supplemented<br />

with different carbon sources<br />

Sugars (w/v) Fresh weight Remarks<br />

(g/l)<br />

Sucrose(0.5 %) 148.8 Compact and numerous chlamydospores<br />

Glucose (1 %) 184.8 Loose, peg-like bodies, few chlamydospores<br />

Fructose (1 %) 78.0 Compact, numerous tiny chlamydospores<br />

Glucose + fructose 109.6 Loose, a few chlamydospores, turned slimy<br />

(0.5 % each)<br />

Data represent an average of three replicates; biomass measured after 7 days.<br />

combination of glucose and fructose (each 0.5 % w/v) led to a medium<br />

increase in the biomass of P. indica (Table 6). On supplementation of glucose<br />

by a mixture of glucose, fructose and sucrose (each 0.5 % w/v), the former<br />

was consumed completely and then the sucrose was metabolized by production<br />

of invertase. This led to an increase of the fructose concentration of the<br />

medium. After the complete consumption of free glucose there was a slow<br />

utilization of fructose.<br />

Fig. 9. P. indica colonies produced in aspergillus broth medium fortified with glucose,<br />

sucrose or fructose. An enlarged view of a colony showing protuberances and peg-like<br />

structures on glucose medium


604<br />

Giang Huong Pham et al.<br />

The morphology of the colonies differed according to the sugar supply. In<br />

fructose and sucrose, the colonies were roundish and compact, in glucose they<br />

were large and irregular with short and long protrusions (Fig. 9).<br />

7 Biomass on Individual Amino Acids<br />

The addition of glycine, methionine, serine, alanine promoted fungal growth<br />

to different extents (Table 7). Not much difference in mycelial growth was<br />

observed in media containing glutamine, asparagine and histidine, although a<br />

substantial difference in the end pH of these amino acid-fortified culture<br />

broths was recorded (Table 7).<br />

8 Growth on Complex Media<br />

P. indica grown in minimal broth was transferred onto one set of fresh minimal<br />

media containing agar. In the minimal broth, the complex mixtures such as<br />

soil-extract, malt-extract, peptone, beef-extract, yeast-extract and caseinhydrolysate<br />

were added individually to an amount of 1 % (w/v). Before autoclaving,<br />

the pH of the media was adjusted to 6.5. Compared to all other media<br />

used,excellent growth of mycelium was recorded in the incubation broth fortified<br />

with casamino hydrolysate-HCl. Growth in beef, yeast, malt extracts and<br />

peptone was moderate. Soil extracts did not support fungal growth (Table 8).<br />

Table 7. End pH and fungal biomass grown on minimal broth medium supplemented<br />

with amino acids (each 0.5 % w/v)<br />

Amino acids End pH Biomass (mg/10 ml)<br />

Control (no addition) 5.14 3.8<br />

Alanine 6.94 6.2<br />

Phenyl alanine 4.31 5.8<br />

Methionine 4.71 7.0<br />

Serine 5.82 6.9<br />

Asparagine 5.86 4.2<br />

Glutamine 4.76 4.3<br />

Cysteine 1.90 3.8<br />

Glycine 5.01 7.8<br />

Aspartic acid 2.91 3.8<br />

Arginine 8.93 3.8<br />

Histidine 7.05 4.0<br />

pH was re-adjusted after the addition of microsyringe-filtered amino acids to 6.5. Incubation<br />

was done under constant shaking condition (GFL, 3026, Germany) for 7 days at<br />

25 °C. Fungal biomass was removed and end pH was measured


9 Phosphatic Nutrients<br />

30 Axenic Culture of Symbiotic Fungus Piriformospora indica 605<br />

Phosphorus is an essential mineral for the growth of P. indica. Optimum<br />

growth was obtained on supplementing the modified aspergillus medium<br />

with Di-potassium hydrogen phosphate in equimolar concentrations (Table<br />

9). Interestingly, the fungus utilized tri-poly phosphate and solubilized insoluble<br />

calcium-hydrogen phosphate. Acid phosphatases were observed to be<br />

active in P. indica mycelium (Varma et al. 2001). The fungus was able to utilize<br />

a variety of inorganic and organic phosphate sources which is in accordance<br />

with the broad range of the substrates utilized by the acid phosphatases of<br />

many fungi. Moreover, phosphate starvation of P. indica led to an overall<br />

(27 %) increase in the intracellular acid phosphatase activity. This increase<br />

was probably due to the appearance of a P-repressible isoform of acid phos-<br />

Table 8. Mycelial biomass of P. indica grown on complex modified aspergillus medium<br />

Complexes Mycelial biomass (mg/10 ml)<br />

Control (no addition) 10<br />

Soil-extract 6<br />

Malt-extract 8<br />

Peptone 9<br />

Beef-extract 12<br />

Yeast-extract 11.5<br />

Casein hydrolysate 5<br />

Complex chemicals (obtained from Difco or Hi media) were included at the rate of 1 %<br />

(w/v); and soil-extract 15 % v/v. pH was readjusted to 6.5. Incubation conditions were the<br />

same as described earlier<br />

Table 9. Biomass of P. indica after 24 days of growth on modified aspergillus medium<br />

supplemented with equimolar (10 mM) concentrations of phosphatic nutrient sources<br />

Phosphate source Dry biomass Final pH of the<br />

g/1000 ml medium<br />

Control (P-) 2.9±0.001 4.10±0.013<br />

Di-hydrogen potassium phosphate 6.9±0.009 4.62±0.05<br />

Di-potassium hydrogen phosphate 7.9±0.002 4.59±0.024<br />

Calcium-hydrogen phosphate 4.8±0.003 4.17±0.008<br />

Di-hydrogen sodium phosphate 6.3±0.005 4.47±0.068<br />

Di-potassium hydrogen phosphate 6.8±0.003 4.34±0.053<br />

Di-hydrogen ammonium phosphate 5.5±0.004 4.36±0.056<br />

Tetra-hydrogen ammonium phosphate 5.2±0.003 4.26±0.04<br />

Tri-polyphosphate 7.9±0.002 5.49±0.074<br />

Tri-metaphosphate 5.8±0.001 4.40±0.008<br />

Aspergillus medium was modified by reducing the concentration of peptone, yeast<br />

extract and casein hydrolysate to ten times the normals


606<br />

phatase in addition to the constitutive one observed in the enzyme staining of<br />

the native polyacrylamide gels. The significance of these enzymes in the phosphate<br />

transport needs to be further substantiated by the studies on the <strong>plant</strong><br />

roots colonized with P. indica.<br />

10 Composition of Media<br />

a Aspergillus (Kaefer 1977)<br />

Composition (g/l)<br />

Glucose 20.0<br />

Peptone 2.0<br />

Yeast extract 1.0<br />

Casein hydrolysate 1.0<br />

Vitamin stock solution 1.0 ml<br />

Macro-elements from stock 50.0 ml<br />

Micro-elements from stock 2.5 ml<br />

Agar 10.0<br />

CaCl2 0.1 M 1.0 ml<br />

FeCl3 0.1 M 1.0 ml<br />

pH<br />

Macro-elements<br />

6.5<br />

(Major elements) Stock (g/l)<br />

NaNO3 120.0<br />

KCl 10.4<br />

MgSO .<br />

4 7H2O 10.4<br />

KH2PO4 Micro-elements<br />

30.4<br />

Trace elements Stock (g/l)<br />

ZnSO .<br />

4 7H2O 22.0<br />

H 3 BO 3<br />

Giang Huong Pham et al.<br />

11.0<br />

MnCl 2 . 4H2O 5.0<br />

FeSO 4 . 7H2O 5.0<br />

CoCl 2 . 6H2O 1.6<br />

CuSO 4 . 5H2O 1.6<br />

(NH 4) 6 Mo 7O 27 4H 2O 1.1<br />

Na 2EDTA 50.0<br />

Vitamins % (w/v)<br />

Biotin 0.05<br />

Nicotinamide 0.5<br />

Pyridoxal phosphate 0.1<br />

Amino benzoic acid 0.1<br />

Riboflavin 0.25<br />

The pH was adjusted to 6.5 with 1 N HCl. All the stocks were stored at 4 °C<br />

except the vitamins which were stored at –20 °C


Modified aspergillus medium (Varma et al. 2001)<br />

The media composition was the same, except that yeast extract, peptone and<br />

casein hydrolysate were reduced to 1/10 in quantity<br />

c M4N (Mukerji et al. 1998)<br />

Composition (g/l)<br />

D-Glucose 10.0<br />

(NH4) 2HPO4 0.25<br />

KH 2 PO 4<br />

0.50<br />

MgSO 4 . 7H2O 0.15<br />

CaCl 2 . 2H2O 0.05<br />

Ferric citrate<br />

(2 % Ferric citrate,<br />

2 % Citric acid w/v) 7.0 ml<br />

NaCl 0.025<br />

Thiamine HCl 100.0 mg<br />

MES 2.5<br />

Malt extract 1.5<br />

Yeast extract 1.5<br />

Agar 15.0<br />

pH 5.6<br />

d Malt Extract (Galloway and Burgess 1952)<br />

Composition (g/l)<br />

Malt extract 30.0<br />

Mycological peptone 5.0<br />

Agar 15.0<br />

pH 5.4<br />

e MMN (Modified Melin-Norkrans) (Johnson et al. 1957)<br />

Composition (g/l)<br />

NaCl 0.025<br />

KH2PO4 0.5<br />

(NH4) 2HPO4 0.25<br />

CaCl2 0.05<br />

0.15<br />

MgSO4 FeCl3 30 Axenic Culture of Symbiotic Fungus Piriformospora indica 607<br />

0.001<br />

Thiamine HCl 83.0 ml<br />

Tryticase peptone 0.1 % (w/v)<br />

Glucose monohydrate 1.0 % (w/v)<br />

Malt extract 5.0 % (w/v)<br />

Trace elements from stock 10.0 ml/l<br />

Trace elements (stock) (g/l)<br />

KCl 3.73


608<br />

H3BO3 1.55<br />

MnSO .<br />

4 H2O 0.85<br />

ZnSO4 0.56<br />

CuSO4 0.13<br />

pH adjusted to 5.8 with 1 N HCl/NaOH. All stocks were stored at 4 °C except<br />

thiamine hydrochloride which was stored at –20 °C<br />

f MMN 1/10 (Herrmann et al. 1998)<br />

Composition (g/l)<br />

CaCl 2 . 2H2O 0.07<br />

MgSO 4 . 7H2O 0.15<br />

NaCl 0.03<br />

(NH4) 2HPO4 0.03<br />

KH2PO4 0.05<br />

Trace elements (stock) (mg/l)<br />

(NH 4 ) 6 Mo 7 O 24 . 4H2 O 0.09<br />

H 3BO 4<br />

1.55<br />

CuSO 4 . 5H2O 0.13<br />

KCl 3.73<br />

MnSO 4 . H2 O 0.84<br />

ZnSO 4 . 7H2O 0.58<br />

Fe-EDTA (mg/l)<br />

FeSO4 8.50<br />

EDTA 1.50<br />

Agar 20.0 g/l<br />

g MMNC (Marx 1969; Kottke et al. 1987)<br />

Composition (g/l)<br />

Glucose 10.0<br />

CaCl 2 . 2H2O 0.07<br />

MgSO 4 . 7H2O 0.15<br />

NaCl 0.03<br />

(NH4) 2HPO4 0.25<br />

KH2PO4 0.5<br />

Casein hydrolysate 1.0<br />

Malt extract 5.0<br />

Trace elements (mg/l)<br />

(NH 4 ) 6 Mo 7 O 24 . 4H2 O 0.02<br />

H 3BO 4<br />

Giang Huong Pham et al.<br />

1.55<br />

CuSO 4 . 5H2 O 0.13<br />

KCl 3.73<br />

MnSO 4 . H2 O 0.85<br />

ZnSO 4 . 7H2O 0.58<br />

Fe-EDTA (mg/l)


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 609<br />

FeSO4 8.5<br />

EDTA 1.5<br />

Vitamins (mg/l)<br />

Thiamine HCl 0.1<br />

Riboflavin 0.1<br />

pH 5.6<br />

Agar 20.0 g/l<br />

h Moser b (modified after Moser 1960)<br />

Macro-elements (g/l)<br />

Glucose 10<br />

Sucrose 10<br />

Maltose 10<br />

Malt extract 10<br />

Peptone 2<br />

K2HPO4 0.15<br />

KH2PO4 0.35<br />

NH4NO3 1<br />

0.3<br />

NaNO3 MgSO .<br />

4 7H2O 0.5<br />

CaCl2 0.1<br />

Micro-elements mg/l<br />

Thiamine 50<br />

Biotine 1<br />

Inositol 50<br />

ZnSO4 1<br />

FeCl3 10<br />

MnSO4 5<br />

Agar 20 g/l<br />

i MS (Murashige and Skoog 1962)<br />

Composition<br />

Macro-nutrients<br />

(mg/l)<br />

NH4NO3 0.5<br />

KNO3 1650.0<br />

CaCl .<br />

2 2H2O 900.0<br />

MgSO .<br />

4 7H2O 440.0<br />

KH2PO4 Micro-nutrients<br />

370.0<br />

KI 170.0<br />

H3BO3 0.83<br />

MnSO .<br />

4 H2O 6.20<br />

ZnSO .<br />

4 7H2O 15.60<br />

NaMoO .<br />

4 2H2O 8.60


610<br />

CuSO 4 . 5H2 O 0.25<br />

CoCl 2 . H2O 0.025<br />

Iron source<br />

Na 2EDTA 0.025<br />

FeSO 4 . 7H2O 37.30<br />

Vitamins<br />

Nicotinic acid 27.8<br />

Pyridoxine HCl 0.5<br />

Thiamine HCl 0.1<br />

Glycine 2.0<br />

Myo-inositol 100.0<br />

Agar 0.7 % (w/v)<br />

Sucrose 3.0 % (w/v)<br />

pH, 5.6–5.7<br />

Each chemical was dissolved in bidistilled water. The pH of the medium<br />

was adjusted using 1 N NaOH/HCl before autoclaving at 121 °C, for 20 min.<br />

Stock solutions were stored at 4 °C except organic supplements, which were<br />

stored at –20 °C<br />

j MYP (Bandoni 1972)<br />

Composition (g/l)<br />

Malt extract 7<br />

Soytone (Difco) 1<br />

Yeast extract 0.5<br />

Agar 15<br />

k Potato Dextrose Agar (PDA) (Martin 1950)<br />

Composition (g/l)<br />

Potato peel 200.0<br />

Dextrose 20.0<br />

Agar 15.0<br />

The periderm (skin) of potatoes (200 g) was peeled-off, cut into small<br />

pieces and boiled in 500 ml of water, until they were easily penetrated by a<br />

glass rod. After filtration through cheese cloth, dextrose was added. Agar was<br />

dissolved and the required volume (1 l) was made up by the addition of water.<br />

The medium was autoclaved at 121 °C for 20 min.<br />

l WPM (“Woody Plant Medium” for Populus) (Ahuja et al. 1986)<br />

Composition (g/l)<br />

Sucrose 20.0<br />

K 2SO 4<br />

Giang Huong Pham et al.<br />

1.00<br />

Ca (NO .<br />

3) 2 4H2O 0.73<br />

NH4NO3 0.40


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 611<br />

MgSO 4 . 7H2 O 0.37<br />

Myo-inositol 0.10<br />

Agar 7.00<br />

Add 700 ml H 2O, adjust pH to 5.8 using 3.7 % HCl (ca. 9.5 ml),<br />

Add after autoclaving sterile phosphate solution (0.17 g KH 2PO 4 dissolved in<br />

270 ml H 2 O+15 ml NaOH (saturated).<br />

10 ml of trace element stock solution (see below)<br />

10 ml Fe-EDTA (see below)<br />

10 ml glycine stock solution (100x: dissolve 20 mg in 100 ml)<br />

1 ml thiamine – stock solution (1000x: dissolve 10 mg in 100 ml)<br />

1 ml nicotinic acid – stock solution (1000x: dissolve 50 mg in 100 ml)<br />

1ml CaCl 2 – stock solution (1000¥: dissolve 3.6 g in 50 ml)<br />

250 ml Pyridoxine – stock solution (4000x: dissolve 40 mg in 100 ml)<br />

100 ml CuSO 4 – stock solution (10,000x: dissolve 25 mg in 100 ml)<br />

sterilize by filtration before adding 100¥ trace element stock solution (autoclave,<br />

store at 4 °C)<br />

MnSO .<br />

4 H2O 2.23<br />

ZnSO .<br />

4 7H2O 0.86<br />

H3BO4 0.62<br />

Ammonium molybdate 0.10<br />

KI 0.09<br />

100x Fe-EDTA stock solution: dissolve 0.128 g FeSO4 and 0.172 g EDTA at<br />

60 °C in 100 ml H2O, store at 4 °C<br />

CaCl .<br />

2 2H2O 0.07<br />

MgSO .<br />

4 7H2O 0.15<br />

NaCl 0.03<br />

(NH4) 2HPO4 0.03<br />

KH2PO4 0.05<br />

Trace elements (mg/l)<br />

(NH4 ) 6Mo7O .<br />

24 4H2O 0.018<br />

H3BO4 0.62<br />

The fungus grew on a wide range of synthetic and complex media. Significant<br />

quantitative and morphological changes were detected when the fungus<br />

was challenged to grow on different media. Shaking during incubation<br />

retarded growth in MMN broth cultures (7–12 g fresh wt./l, after 2 weeks at<br />

30 °C), whereas no such negative effect was ever observed during cultivation<br />

on any other substrates. There was practically no growth when mycelia were<br />

incubated under shaking conditions, whereas in stationary conditions, normal<br />

growth was obtained. Hyphae did not adjust to even a slow rate of shaking.<br />

In fact, the fungal biomass was considerably enhanced on shaking cultures<br />

with aspergillus medium, sometimes up to 50 g fresh wt./l after 2 weeks<br />

at 30 °C. On aspergillus and Moser b media, the colonies appeared compact,


612<br />

Giang Huong Pham et al.<br />

wrinkled with furrows and constricted. The mycelium produced fine zonation<br />

and a great amount of white aerial hyphae. Hyphae were highly interwoven,<br />

often adhered together and gave the appearance of simple cords. New<br />

branches emerged irregularly and the hyphal walls showed some external<br />

deposits at regular intervals, which stained deeply with toluidine blue. Since<br />

septation was irregular, the single compartment could contain more than one<br />

nucleus. The chlamydospores appeared singly or in clusters at the apex of<br />

hyphae. They were distinctive due to their pear-shaped habit.<br />

11 Conclusions<br />

Mycorrhiza does not always promote the growth of agricultural crops. In<br />

phosphorus-rich soils, they can parasitize <strong>plant</strong>s such as citrus, wheat and<br />

maize by tapping sugars from these <strong>plant</strong>s without giving anything back.<br />

Researchers ignore this darker side of the mycorrhiza. Theoretically, mycorrhiza<br />

can also harm biodiversity. In the long run, specific mycorrhizas can<br />

promote the growth of one <strong>plant</strong> at the expense of another.“What exactly happens<br />

probably depends on the system itself,” states Van der Heijden (2002). In<br />

any case, the interaction between <strong>plant</strong>s and mycorrhiza forming fungi clearly<br />

has at least as great an effect on the ecosystem’s species composition as the<br />

interaction/competition between <strong>plant</strong>s themselves.<br />

P. indica, the fungus treated in this chapter, acts as biofertilizer, bioregulator<br />

and bioprotector, and can be easily mass-multiplied on defined synthetic<br />

media. It is thus, an interesting model fungus with respect to studies on<br />

endomycorrhiza. In addition, commercial production of this fungus under<br />

aseptic conditions could support biological hardening of tissue-cultureraised<br />

<strong>plant</strong>s as well as <strong>plant</strong> survival in general on poor soils.<br />

Acknowledgements. The Indian authors are thankful to DBT, DST, CSIR, UGC, and the<br />

Government of India for partial financial assistance.<br />

References and Selected Reading<br />

Ahuja MR (1986) In: Evans DA, Sharp WR and Ammirato PJ (eds) Handbook of <strong>plant</strong><br />

cell culture 4, techniques and applications. Macmillan, New York, pp 626–651<br />

Badoni RJ (1972) Terrestrial occurrence of some aquatic Hyphomycetes. Can J Bot<br />

50:2283–2288<br />

Galloway LD, Burgess R (1952) Applied mycology and bacteriology, 3rd edn. Leonard<br />

Hill, London, pp 54–57<br />

Herrmann S, Munch JC, Buscot F (1998) A gnotobiotic culture system with oak microcuttings<br />

to study specific effects of mycobionts on <strong>plant</strong> morphology before, and in<br />

the early phase of, ectomycorrhiza formation by Paxillus involutus and Piloderma<br />

croceum. New Phytol 138:203–212


30 Axenic Culture of Symbiotic Fungus Piriformospora indica 613<br />

Johnson CN, Stout PR, Broyer RC, Carlton AB (1957) Comparative chlorine requirements<br />

of different <strong>plant</strong> species. Plant Soil 8:337–353<br />

Kaefer E (1977) Meiotic and mitotic recombination in Aspergillus and its chromosomal<br />

aberrations. Adv Genet 19:33–131<br />

Kottke I, Guttenberger M, Hampp R, Oberwinkler F (1987) An in vitro method for establishing<br />

mycorrhizae on coniferous tree seedlings. Trees 1:191–194<br />

Martin JP (1950) Use of acid, rose bengal and streptomycin in the plate method for estimating<br />

soil fungi. Soil Sci 69:215–232<br />

Marx, DH (1969) The influence of ectotrophic mycorrhizal fungi on the resistance of<br />

pine roots to pathogenic infections. I. Antagonism of mycorrhizal fungi to root pathogenic<br />

fungi and soil bacteria. Phytopathology 59:153–163<br />

Moser M (1960) Die Gattung Phlegmacium. J Klinkhardt, Bad Heilbrunn, Austria<br />

Mukerji KG, Mandeep, Varma A (1998) Mycorrhizosphere microorganisms: screening<br />

and evaluation. In: Varma A (ed) Mycorrhiza manual. Springer, Berlin Heidelberg<br />

New York, pp 85–98<br />

Murashige T, Skoog F (1962) A revised medium for rapid growth and bioassay with<br />

tobacco tissue cultures. Physiol Plant 15:431–487<br />

Schüßler A, Schwarzott D, Walker C (2001) A new fungal phylum, the Glomeromycota:<br />

phylogeny and evolution. Mycol Res 105:1413–1421<br />

Singh An, Singh Ar, Kumari M, Rai MK,Varma A (2003a) Biotechnological importance of<br />

Piriformospora indica Verma et al. a novel symbiotic mycorrhiza-like fungus: an<br />

overview. Indian J Biotechnol 2:65–75<br />

Singh An, Singh Ar, Kumari M, Kumar S, Rai MK, Sharma AP and Varma A (2003b)<br />

Unmassing the accessible treasures of the hidden unexplored microbial world. In:<br />

Prasad BN (ed) Biotechnology in sustainable biodiversity and food security. Science<br />

Publishers, Inc. Enfield, NH, USA, pp 101–124<br />

Van der Heijden MAG (2002) Arbuscular mycorrhizal fungi as a determinant of <strong>plant</strong><br />

diversity: in search for underlying mechanisms and general principles. In: Van der<br />

Heijden MGA and Sanders IR (eds) Mycorrhizal ecology. Ecological Studies 157.<br />

Springer, Berlin Heidelberg New York, pp 243–266<br />

Varma A,Verma S, Sudha, Sahay NS, Franken P (1999) Piriformospora indica,a cultivable<br />

<strong>plant</strong> growth promoting root endophyte with similarities to arbuscular mycorrhizal<br />

fungi. Appl Environ Microbiol 65:2741–2744<br />

Varma A, Singh A, Sudha, Sahay NS, Sharma J, Roy A, Kumari M, Rana D, Thakran S, Deka<br />

D, Bharati K, Hurek T, Blechert O, Rexer KH, Kost G, Hahn A, Hock B, Maier W, Walter<br />

M, Strack D, Kranner I (2001) Piriformospora indica: An axenically culturable<br />

mycorrhiza-like endosymbiotic fungus. In: Hock B (ed) Mycota IX. Springer, Berlin<br />

Heidelberg New York, pp 123–150<br />

Varma A, Singh A, Sudha, Sahay NS, Kumari M, Bharti K, Sarbhoy AK, Maier W, Walter<br />

MH, Strack D, Franken P, Singh An, Malla R, Hurek T (2002) Piriformospora indica:A<br />

<strong>plant</strong> stimulator and pathogen inhibitor arbuscular mycorrhizal-like fungus. In:<br />

Markandey DK, Markandey NR (eds) Microorganisms in bioremediation. Capital<br />

Publishing Company Ltd., New Delhi, pp 71–89<br />

Verma S, Varma A, Rexer KH, Hassel A, Kost G, Sarbhoy A, Bisen P, Bütehorn B, Franken<br />

P (1998) Piriformospora indica gen. et sp. nov., a new root-colonizing fungus. Mycologia<br />

90:895–909


Subject Index<br />

A<br />

AAD 590<br />

Abies (fir) 151<br />

Abiotic factors 26<br />

Abrus precatorius 243<br />

Abscisic acid 88<br />

ABTS 261<br />

Acacia sp. 113<br />

Acacia catechu 243<br />

A. holoseriaca 201<br />

A. nilotica 243<br />

ACC deaminase 133, 489, 494<br />

Acetobacter 83,198, 200<br />

Acetyl CoA acetyltransferase 590<br />

Achnatherum 158<br />

Acid phosphatase 337, 606<br />

Acidic heteropolysaccharide 505, 516<br />

Acremonium 89<br />

Actin 230<br />

Actin cap 304<br />

Actin genes 297<br />

Actin-GFP 318<br />

Actinomycetes 59<br />

Actinomyces 73, 89<br />

Actinomycetes 127, 203<br />

Actinorhiza 2, 80<br />

Adhatoda vasica 77, 243, 247<br />

Adhesion pad 219<br />

Aequorea victoria 438<br />

Aerenchyma 36<br />

Aerobacter 73<br />

Aeromaonas 73, 89<br />

AFLPs 10, 551, 556<br />

Agaricus 73, 402, 412, 415<br />

A. bisporus 90, 597<br />

Agglutination 24<br />

Agrobacterium 82, 88, 420, 421<br />

A. tumefasciens 3, 121, 124<br />

Agrostis hiemalis 163<br />

Alcaligenes eutrophus 63<br />

Aldehyde dehydrogenase 580<br />

Alternaria 73, 89<br />

Alkaline phosphatase 335, 337<br />

Allelochemicals 82<br />

Alnus 81<br />

AM colonization 78<br />

AM fungal symbiosis 591<br />

Amanita gemmata 260<br />

A. muscaria 5, 7, 203, 260, 597<br />

A. rubescens 260<br />

A. spissa 260<br />

A. strobiliformis 260<br />

AMF 262<br />

AMF-like 597<br />

1-Aminocyclopropane-1-carboxylic acid<br />

(ACC) 4<br />

Aminotransferase 397, 417<br />

Ammonifier 56<br />

Ammonium transport 399<br />

AMOVA 562<br />

Amplifier rDNA restriction analysis 75<br />

a-Amylase 124<br />

Amylolytic 128<br />

Amyloplasts 301<br />

Anabaena 73<br />

Anaerobic stress 74<br />

Anastomoses 593<br />

Aneura pinguis 242, 243<br />

Annoxic sites 2<br />

Antagonists 361<br />

Anthyllis cytisoides 359<br />

Antibiotic resistance marker cassette<br />

460<br />

Antibiotics 201


616<br />

Subject Index<br />

Antifungal activity test 434<br />

Antiport 413<br />

Apoplast 165, 380<br />

Apoplastic space 164<br />

Apoptosis 582<br />

Arabidopsis thaliana 3, 243, 256, 399,<br />

Arabinose 173, 602<br />

Arbuscular mycorrhiza 60, 185, 567<br />

Arbuscule 308, 310, 567<br />

Archaea 41<br />

Artemisia annua 243, 247<br />

Arthrobacter globiformis 63, 82, 89<br />

Arum-type mycorrhizas 334<br />

Ascomycetes 76<br />

Asparagine 604<br />

Aspergillus eutrphus 64<br />

A. globiformis 64<br />

A. flavus 240<br />

A. muscaria 203<br />

A. nidulans 580, 590<br />

A. niger 85, 240<br />

A. sydowii 240<br />

A. terreus 580<br />

A. tubingensis 85<br />

Asymbiotic phase 579<br />

Atkinsonella hypoxylon 160, 164, 166<br />

ATP 575<br />

dATP 582<br />

32p-dATP 585, 588<br />

Aureofungin 88<br />

Autofluorescent proteins 8, 18, 431, 438<br />

Automated sequencer 579<br />

Autotrophic organisms 65<br />

Auxin 4, 88, 315<br />

Auxin-type phytohormones 355<br />

Axenically 237<br />

Azadirachta indica 243, 247<br />

Azoarcus 83<br />

Azorhizobium 82, 531<br />

Azospirillum 73, 83, 200, 239, 355, 360<br />

A. brasilense 89, 455<br />

Azotobacter 73, 82, 83, 198<br />

A. choroococcum 84<br />

B<br />

Bacillus 2, 63, 73, 83, 198<br />

B. cereus 127, 128<br />

B. geophilum 203<br />

B. megaterium 127<br />

B. subtilis 3,127, 239<br />

B. thuringiensis 4, 121, 535, 540<br />

B. thuringiensis subsp. Galleriae 127<br />

B. thuringiensis var kurstaki 126<br />

Bacteria fungi interaction 197<br />

Bacopa monniera 243, 247,248<br />

Bacterial extraction method 457, 460<br />

Bacterial morphotype 531, 532<br />

Balansia sp. 158<br />

Basidiomycetes 76, 260, 267<br />

Basidiomycota 595<br />

Beauvaria sp. 597<br />

Beijerinckia sp. 73, 83<br />

b-galactosidase 19, 576<br />

b-1,3-glucanase 82<br />

b-glucosidase 87<br />

b-glucuronidase 19<br />

b-importin 590<br />

Bi Dye cycle sequencing kit 579<br />

Biocontrol 432<br />

Biodegradation 74<br />

Biofertilizer 613<br />

Biofilm 154<br />

Biogeochemical 74<br />

Biogeochemical cycles 51, 64<br />

Biogeography 541-542<br />

Bioindicators 54<br />

Bio-insecticide protein 4<br />

Bio-insecticides 121, 122<br />

Biological control agents 361<br />

Biological hardening 613<br />

Biomass production 245<br />

Bioprotector 613<br />

Bioregulator 613<br />

Bioremediation 206<br />

Biosurfactant 153<br />

Biotic factors 26<br />

Biotic signals 2<br />

Biotin 106<br />

Biotrophic 159<br />

Biotrophs 157<br />

BiP protein 590<br />

BLAST 579<br />

BLAST analysis 581<br />

BLASTX 580, 581<br />

BLASTX alignment tool 579<br />

Blue fluorescent protein (BFP) 441<br />

BM25.8 cells 577<br />

Boletinus cavipes 260<br />

B. edulis 260<br />

B. erythropus 260<br />

Boletus luridus 260<br />

B. piperatus 260<br />

Borrelia burgdorferi 4<br />

B. burgdorferi B31 3


Botanophila 171<br />

Bradyrhizobium 73, 82, 238<br />

B. japonicum 2, 3<br />

Brassica juncea 255<br />

B. oleracea 255<br />

Brassicaceae 76<br />

Brevibacterium 89<br />

Brightfield microscopy 510, 519, 524<br />

Bryophyte 242<br />

BSA 569<br />

Bt toxin 181, 184<br />

BT transgenic <strong>plant</strong>s 4<br />

Bt-maize 122<br />

B-transducin 590<br />

Bt-transgenic <strong>plant</strong>s 121<br />

Bulk soil 197, 450, 459, 464<br />

Burkholderia 82, 198, 200, 201<br />

B. cepacia 201<br />

Burkholderia-like bacteria 358<br />

C<br />

Caenorhabd iris elegans 580<br />

Calcium oscillations 112<br />

cAMP 376<br />

Candida sp. 89<br />

C. albicans 597<br />

Capsule 506, 508, 512, 513<br />

Carboxytates (complexone) 90<br />

Cassette vector 440<br />

Cassia angustifolia Vahl 243<br />

Casuarina sp. 81<br />

Casuarinaceae 81<br />

Catabolic diversity 73<br />

Catabolic response profile (CRP) 73<br />

Catecholate siderophores 90<br />

Cauliflower mosaic virus 124<br />

Caullinite 125<br />

Cdc2a kinase 316<br />

Cdc42 306<br />

Ceanothus 81<br />

Cell attachment 505, 508<br />

Cell cycle switch protein 580<br />

Cell division 314<br />

Cell motility 505, 527-528<br />

Cell wall 303<br />

Cellobiohydrolase 87, 88<br />

Cellular interaction 267<br />

Cellulase 54, 80,522<br />

Cellulolytic fungi 86<br />

Cellulomonas sp. 82<br />

Cenococcum sp. 393, 410, 416<br />

Cephalozia biscuspidata 242<br />

Subject Index 617<br />

Cercospora 89<br />

Chalamydospores 237, 594<br />

Charge couple device (CCD) 439<br />

Chemo-heterotrophic 122<br />

Chenopodiaceae 76<br />

Chitinase 82<br />

Chlamydia tracchomatis 3, 4<br />

Chlamydomonas reinhardtii 239<br />

Chlorobium sp. 73<br />

Chlorophytum borivillianum 243, 247<br />

C. tuberosum Baker 243<br />

Cholesterol 124<br />

Chromaspin-400 Columns 573<br />

Chum synthase I 590<br />

Cicer arietinum 245<br />

C. borivillianum 247<br />

C. purpurea 174<br />

C. sinensis 158<br />

Citrobacter sp. 89<br />

Cladosporium sp. 89, 597<br />

Clathrin adapter protein 590<br />

Clavicipitaceae 4, 158<br />

Clavicipitaleans sp. 157<br />

Clostridium sp. 73<br />

CMC-ase 261<br />

CMEIAS 9, 531, 544<br />

Co-cultivation 252<br />

Co-culture 252<br />

Coffea arabica 243<br />

Coils 567<br />

Colacogloea peniophorae 269<br />

Colacosomes 6, 268<br />

Collectotrichum 287<br />

Colonization 149, 242<br />

Community level physiological profile<br />

(CLPP) 463<br />

Competitive colonisation 17<br />

Computer-assisted microscopy 526,<br />

528, 530, 544<br />

Confocal laser scanning microscopy<br />

(CSLM) 8, 355, 451, 509, 540, 543<br />

Contact angle 471, 472<br />

Coprogen 90<br />

Coralloid 595<br />

Cordyceps militaris 158<br />

Cordycipitoideae 158, 159<br />

Cortical microtubules 298<br />

Cortinarius varius 260<br />

Crack entry 510, 531<br />

cry genes 124<br />

Cry protein 123, 124<br />

Cry1Ab toxin 125


618<br />

Subject Index<br />

Cryosection 317<br />

Cultivation 595<br />

Culturability 449<br />

Cunninghamella 240<br />

C. echinulata 597<br />

Cuticle 211, 221, 471<br />

Cuticular penetration 481, 482<br />

Cuticular permeability 149, 153, 479<br />

Cuticular transport 479<br />

Cuticular wax 147, 473, 474<br />

Cu-Zn Superoxide dismutase 580<br />

Cyan fluorescent protein (CFP) 441<br />

Cyathus 85<br />

Cyclic glucans 109<br />

Cyclic trihydroxamate 90<br />

Cyclin 315<br />

Cycloheximide 433, 440<br />

Cylindrocarpon sp. 201<br />

Cymbopogon martinii 243<br />

32p-dCTP 585, 588<br />

Cynodon dactylon 595<br />

Cyperaceae 76<br />

Cysteine-rich proteins 219<br />

Cytidindeaminase 580<br />

Cytokinin 88, 89, 170<br />

Cytoplasmic streaming 300<br />

Cytoskeletal organization 308<br />

Cytoskeleton 6, 505, 516, 527, 529<br />

Czapek-Dox medium 435<br />

D<br />

Dactylorhiza majalis 243<br />

D. incarnata 243<br />

D. maculata 243<br />

D. majalis 246<br />

D. purpurella 243<br />

D. fuchi 243<br />

D. purpurella 246<br />

Dalbergia sissoo 252<br />

Damping off seedling 157<br />

DAPI 458, 594<br />

Darkfield microscopy 525, 527<br />

Daucus carota 243, 259<br />

dCTP 582<br />

Deciduous trees 596<br />

Decomposer 74<br />

Decomposition 74<br />

Dehydrogenisation 54<br />

Deionized H20 574<br />

Deleterious rhizosphere organisms 352<br />

De-mineral 65<br />

Denaturant gradient gel electrophoresis<br />

(DGGE) 75<br />

Dendrobium crumenatum 580<br />

Denhardt’s solution 586<br />

Denitrifiers 72<br />

Denitrifying bateria 72<br />

Dephosphorylated l TriplEx2 vector<br />

574<br />

Depolymerisation 54<br />

Derxia sp. 83<br />

Desferriform 89<br />

Desmostachya sp. 77<br />

Desulfovibrio sp. 73, 83<br />

DGGE-finger printing 461, 462<br />

dGTP 582<br />

Diazotrophic baterial 157<br />

Diglycosyl diacylglycerol glycolipid 508<br />

Dikaryotic hyphae 296<br />

Di-potassium hydrogen phosphate 606<br />

Discosoma sp. 441<br />

Disease index 27<br />

Diterpenoid acids 88<br />

Differential expression 590<br />

cDNA 569, 573<br />

– clones 568<br />

– DNA library 569, 577<br />

– DNA polymerase mix 569<br />

– DNA probes 584, 585, 588<br />

– DNA synthesis 569<br />

DNA concentration 553<br />

DNA polymerase 572<br />

DNA sequencher 579<br />

16s rDNA sequense analysis 8<br />

DNA-hybridization 75<br />

DNAse 569<br />

dNTPs 569, 578, 584<br />

Dolipores 595<br />

Double-Stranded adapters 552<br />

Double-stranded cDNA 569<br />

Douglas fir 414, 415<br />

Drosophila melanogaster 580<br />

Dsc DNA 585<br />

DsRed 441, 442, 456<br />

DTT 584<br />

Dual colour imaging 440<br />

Dynactin complex 312<br />

DynamicET cycle sequencing kit 579<br />

Dynein 310, 312<br />

E<br />

Ecological significance 393<br />

Ecological specificity 332


Ectendomycorrhizas 76<br />

Ectomycorrhizal fungi 597<br />

Ectomycorrhiza 5, 185, 211, 295<br />

Ectomycorrhizal ascomycetes 261<br />

Ectomycorrhizas 567<br />

Ectorhizosphere 8, 450, 458, 464<br />

Elaeagnceae 81<br />

Eleagnus sp. 81<br />

Electrophoresis 579<br />

Electroporation 124<br />

E-MAP-115 590<br />

Endo-b-1,6-glucanase 164, 165<br />

Endocellulase 87<br />

Endochitinase 164<br />

Endomycorrhiza 296, 613<br />

Endomycorrhizal fungi 261, 597<br />

Endopeptidase 580<br />

Endophytes 6, 355<br />

Endophytic hyphae 162<br />

Endophytic mycelium 160<br />

Endorrhizosphere 8, 450, 458, 464<br />

Enterobater sp. 73<br />

E. agglomerans 84<br />

Entomopathogenic 164<br />

Environmental fitness 460<br />

Enzymatic isolation 476<br />

Epacridaceae 79<br />

Ephelidial conidia 169<br />

Epichloe festucae 158<br />

E. typhina 157<br />

E. clarkii 157<br />

Epicuticular wax 148<br />

Epidermal eroded pits 522, 524<br />

Epifluorescence 8, 431, 594<br />

Epiphyllic microflora 150<br />

Epiphyllic microorganisms 9, 473, 477<br />

Epiphyllous mycelium 160<br />

Epochloe sp.158<br />

Epolionts 157<br />

Equimolar concentrations 606<br />

Ergot alkaloids 168<br />

Ergovaline 168<br />

Ericaceous host <strong>plant</strong> 80<br />

Ericaceous mycorrhizas 76<br />

Ericaeae 79<br />

Ericoid fungi 80<br />

Ericoid mycorrhizal fungi 73, 80<br />

Erwinia sp. 73, 89<br />

Escherichia coli 0157:H7,EDI.933 3<br />

E. coli 0157:H7, Sakai 3<br />

E. coli XL-I blue 575<br />

EST 579, 590<br />

Subject Index 619<br />

EST clones 581<br />

Estrogenic activity 106<br />

Ethylene 4, 88, 134, 489, 492<br />

Eurhynchium praelongum 242<br />

Exoenzymes 58<br />

Exo-poly-saccharides 110, 355<br />

Expressed Sequence Tags (ESTs) 10, 568<br />

Extracellular microfibrils 508, 526, 527<br />

Extraradical 360<br />

Extragenic palindromic- PCR 10<br />

Extramatrical 246<br />

Extraradical hyphae 78, 358<br />

Extraradical mycelia 199<br />

Exudate 197<br />

F<br />

Fahraeus slide culture 504, 506<br />

FASTA format 579<br />

Fatty acid methyl ester profiling (FAME)<br />

75<br />

Fatty acid methyl esters (FAME) 463<br />

Fatty acid-derived signals 105<br />

Ferrated siderophore 89<br />

Ferribacterium 73<br />

Ferrichrome 90<br />

Ferricrocin 90<br />

Festuca arizonica 157<br />

F. versuta 158<br />

Fimbriae 355<br />

Fingerprinting techniques 75<br />

First strand buffer 569<br />

Flavanoids 102, 105<br />

Flavobacterium 73, 82, 89<br />

Flourescent in situ hybridization (FISH)<br />

8<br />

Fluorescent pseudomonads 72<br />

Fluorescence in situ hybridization (FISH)<br />

75, 449, 453, 460<br />

Fluorescence marker-tagged bacteria<br />

449, 456<br />

Fluorescence microscopy 510, 520, 525,<br />

530, 553<br />

Fluorescent-activated cell sorters (FACS)<br />

439<br />

Fluorometers 569<br />

Frankia sp. 73, 80<br />

Fructose 374<br />

Fructose 2,6-bisphophate 376<br />

Functional Genomic Approaches 567<br />

Fungal sheath 379<br />

Fungicide cycloheximide 433<br />

Fusarinines (fusigens) 90


620<br />

Subject Index<br />

Fusarium sp. 73, 89<br />

F. culmorum 87<br />

F. moniliforme 201<br />

F. solani 597<br />

F. oxysporum 78, 201, 435<br />

F. oxysporum f.sp radicis-lycopersici<br />

431<br />

Fusion mycoparasites 275<br />

fusion-interaction 275<br />

G<br />

Gaeumannomyces sp. 5<br />

G. graminis 240<br />

Gametophyte 242<br />

Gas vascular transport 38<br />

Gel electrophoresis 569<br />

Gelatin 578<br />

Geldanamycin 82<br />

Gene expression profiling 568<br />

Gene pool 72<br />

Gene regulation 385<br />

Genetically modified <strong>plant</strong>s (GMP) 4,<br />

179,196<br />

Genomenet 579<br />

Genomes 567<br />

Genomic DNA 10<br />

Geostatistics 532, 540, 544<br />

Germination 246<br />

Gfp half-life 441<br />

Gibberella 73, 89<br />

Gibberellins (GA) 88<br />

Gigaspora decipiens 332<br />

Gi. margarita 333, 356, 580<br />

Gi. gigantia 597<br />

Gliocladium 90<br />

Glomales 332<br />

Glomeromycota 353<br />

Glomus sp. 396, 409<br />

G. caledonium 597<br />

G. clarum mycelia 62<br />

G. clarum 78<br />

G. coronatum 597-<br />

G. deserticola 79<br />

G. etunicatum 26, 261<br />

G. fasciculatum 78, 333<br />

G. geosporum 597<br />

G. intraradices 63, 78, 597<br />

G. invermaium 334<br />

G. lamellosum 597<br />

G. mosseae 63, 248, 376, 597<br />

G. proliferum 597<br />

Glucose 602, 604<br />

Glucosinolates 76<br />

Glutamate dehydrogenase (GDH) 397,<br />

409, 410, 414, 416, 417<br />

Glutamate synthase (GOGAT) 397, 410,<br />

413, 415, 418<br />

Glutamine 604<br />

Glutamine synthetase 397, 408, 410,<br />

412, 420<br />

Glutathione S transferase 580<br />

Glycine max 243, 255<br />

Glycogen 377, 572, 574<br />

Glycolysis 376<br />

Gnotobiotic bioassay 435<br />

Gnotobiotic system 14<br />

Gnotobiotic test 8<br />

G-protein coupled receptor 305<br />

Gram-positive bacteria 459<br />

Green fluorescence protein (GFP) 456<br />

Green fluorescent protein (GFP) 355,<br />

438<br />

Griseofulvin 88<br />

Growth factors 106<br />

Gymnosporangium 88<br />

H<br />

Haemocytometer 435<br />

Hansenula 396, 407, 409<br />

Hartig net 221, 379<br />

Hartig net formation 5<br />

Haustoria 6, 165<br />

Heat shock protein HSS1 580<br />

Hebeloma 395, 397, 398, 415, 421<br />

H. crustuliniforme 260<br />

H. edurum 260<br />

H. hiemale 260<br />

H. sunapizans 260<br />

Hedera (ivy) 151, 475<br />

Helper bacteria 198, 200<br />

Hemicellulases 80<br />

Herbaspirillum 83, 198, 200<br />

Herbicide-resistance 179, 180<br />

Heterothallic 595<br />

Heterotrophic 57, 599<br />

Hexose transporter 375<br />

Hexose gradient 379<br />

Hierarchical clustering 579<br />

High-throughput sequencing 567<br />

Histidine 604<br />

Histone H4 580<br />

Homeobox genes Hox-2.6 590<br />

Homo sapiens 580, 590<br />

Homogenous 255


Horizontal gene transfer (HGT) 4, 191<br />

Horizontal growth station 505<br />

Host lectin 505, 513<br />

Hosts 268<br />

Humus 53<br />

Hyaline 593<br />

Hybridization 569<br />

Hybridization analysis 584<br />

Hybridization hypothesis 172<br />

Hydanthocidin 82<br />

Hydrolytic Enzymes 164, 165<br />

Hydrophobin 220<br />

Hydroponics 432<br />

Hydroxamate 90<br />

Hydroxamate siderophores 90<br />

Hydroxyapetite 84<br />

Hymenomycetes 595<br />

Hymenoscyphus ericae 79<br />

Hyperdermium sp. 159<br />

Hyperdermium bertonii 159<br />

Hypertrophied 166<br />

Hypha 238, 269, 593<br />

Hyphal attachment 218<br />

Hyphal tip 311<br />

Hyphosphere 61, 197, 358<br />

Hypocrella sp. 159<br />

H. africana 159<br />

H. gaertneriana 159<br />

H. schizostachyi 159<br />

I<br />

Image analysis 526, 544<br />

Immunoelectron microscopy 509, 517<br />

Immunofluorescence labelling 449, 453<br />

Immunofluorescence microscopy 6<br />

In situ gene expression 525, 526<br />

In situ microbial ecology 504, 529, 544<br />

Incubation 251<br />

Indigenous microflora 503, 530, 535<br />

Indirect immunofluorescence<br />

microscopy 298<br />

Infection process 504, 529<br />

Infection-related biological activity<br />

505, 516, 520<br />

Inflorescence primordium 166<br />

Inflorescens 248<br />

Inhibitory zone 434<br />

Inoculation 251<br />

Interface 270<br />

Intergenic spacer (IGS) region 462<br />

Intergrin-adhesion-receptor 2<br />

Internal transcribed spacer (ITS) 75<br />

Interhyphal spaces 200<br />

Interwoven 593<br />

Intracellular 238<br />

Intracellular acid phosphatase 606<br />

Intraradical hyphae 335, 337<br />

Introns 567<br />

Ion transport 568<br />

IPTG 576<br />

IRS 1-like protein 590<br />

Isocitrate lyase 580<br />

Isoflavonoids 102<br />

Isolated cuticle 475, 476<br />

Isolation of bacillus 434<br />

Isolation of pseudomonads 433<br />

ITS 551<br />

ITS-RFLP 559<br />

J<br />

Juglans sp. 146<br />

Juncaceae 76<br />

Subject Index 621<br />

K<br />

Kanamycin resistance 187, 188<br />

Kinases 124<br />

Kinesin 309, 311<br />

Kinetin 170<br />

Klebsiella 83, 89<br />

L<br />

Laccaria 395, 410, 412, 414, 416, 421<br />

L. amethystea 212<br />

L. amethystina 260<br />

L. bicolor 201, 581, 590<br />

L. proxima 201<br />

Lactarius delicious 260<br />

L. deterrimus 260<br />

L. necator 260<br />

L. rufus 260<br />

L. subdulcis 200<br />

L. torminosus 260, 597<br />

L. vellereus 200<br />

Lac tonohydrolase 590<br />

Lactose mannose 602<br />

Lambda zap 569<br />

Laminar flow hood 576<br />

Larix decidua 200<br />

LB agar 576<br />

LB/Carbenicllin plates 577<br />

LB/MgSO4 agar 576<br />

LbAut7 590<br />

LCO receptor 112<br />

Leaf <strong>surface</strong> 145, 471


622<br />

Subject Index<br />

Leaf <strong>surface</strong> colonisation 483, 485, 486<br />

Leaf <strong>surface</strong> roughness 148<br />

Leaf <strong>surface</strong> wetting 150<br />

Lecaythidaceae 76<br />

Leccinum scabrum 260<br />

L. versipelle 260<br />

Lectin 124, 505, 508<br />

Lentinula edodes 580, 597<br />

Leptothrix sp. 73<br />

Ligation 572, 574<br />

Ligninases 80<br />

Lignin-rich organic 79<br />

Lignins 287<br />

Lignocellulolytic enzyme activity 86<br />

Lignocellulolytic Microorganisms 85<br />

Lipase 82<br />

Lipochitooligosaccharides 2<br />

Lipooligosaccharide Nod factor 505,<br />

526<br />

Lipopolysaccharide 110, 505, 509<br />

Long distance PCR 569<br />

Long root 212<br />

Lotus japonicus 337, 339<br />

Luteolin 104<br />

Lycopersicon escuslentum 248<br />

LZK protein kinase 590<br />

M<br />

Macroarray mycorrhizal symbiosis 590<br />

Macroarray techniques 10<br />

Macrofauna 129<br />

Malate synthase 590<br />

MALDI-TOF 203<br />

Maleylacetale isomerase 2 580<br />

Maltase 54<br />

Mangrove <strong>plant</strong>s 76<br />

Mannitol 199<br />

MAP 309<br />

Maturation time 442<br />

Medicago arborea 361<br />

M. sativa 361, 580<br />

M. truncatula 336, 339<br />

Mesorhizobium sp. 82<br />

M. loti 2, 3<br />

M. mediterraneum 854<br />

Metabolization of flavonoids 104<br />

Methane cycle 35<br />

Methane oxidation 37<br />

Methane production 38<br />

Methannobacterium thermoautotrophicum<br />

3<br />

Methanogens 2, 35, 72<br />

Methanosarcina mazei 3<br />

Methanotroph 2, 44<br />

Methylcellulose 435<br />

Microaggregates 73<br />

Microarrays 568, 584, 568<br />

Microbial communities 202, 503, 541<br />

Microbial community analysis 449<br />

Microbial diversity 71<br />

Microbiota 123, 351<br />

Micro-centrifuge 568<br />

Micrococcus sp. 82, 200<br />

Microcolony 23<br />

Microcosm 51, 351, 362<br />

Microfilaments 293<br />

Microhabitats 2, 72<br />

Micro-propagated 252<br />

Microscopic in situ approach 450, 464<br />

Microsymbiont 2, 504, 526<br />

Microtubules 293<br />

Mineralisation 56<br />

MMN 239<br />

Mobilisation 56<br />

Model of nitrogen uptake and release<br />

384<br />

Molecular microscopy 511, 514<br />

Monilia sp. 89<br />

Monoclonal antibody 509, 531<br />

Monocots 25<br />

Monosaccharides 600<br />

Montmorillonite 125<br />

Moraxella sp. 200<br />

Morchella conica 261<br />

M. elata 261<br />

M. escuslenta 261<br />

Morphotypes 339<br />

Mucilage 23, 255<br />

Mucor sp. 89<br />

Multalin 579<br />

Multi print replication device 584<br />

Multiblot replicator 568<br />

Multiscreen filter plate 583<br />

Mummifying 166<br />

Mus musculus 590<br />

Mussoorie rock phosphate 84<br />

Mutagenesis 440<br />

Mycelia 255, 593<br />

Mycelium 599<br />

Myc - mutants 342<br />

Mycobacterium sp. 82<br />

Mycobionts 262<br />

Mycoparasites 267<br />

Mycoparasitic 357


Mycoparasitic activity 165<br />

Mycoparasitism 165, 274<br />

Mycorrhiza 2, 197, 247, 255, 613<br />

Mycorrhiza formation 252<br />

Mycorrhiza-helper-bateria 357, 360<br />

Mycorrhizal complex 60<br />

Mycorrhizal symbiosis 567, 591<br />

Mycorrhizosphere 61, 197, 199, 358,<br />

Mycorrhizosphere bacteria 199<br />

Myosin 307<br />

MYP 256<br />

Myrica 81<br />

Myricaceae 81<br />

Myriogenospora 166<br />

M. atramentosa 166<br />

N<br />

N-acetyl-D-glucosamine 199<br />

N-acetylglutamic acid 508<br />

Naegleria fowleri 580<br />

NCBI 579<br />

Necrotic lesions 431<br />

Necrotrophic 259<br />

Necrotrophied 158<br />

Neighbor-Joining 562<br />

Nematophagous 164<br />

Neotyphodial conidia 169, 170<br />

Neotyphodium sp. 157, 158, 164<br />

N. lolii 174<br />

N. coenophialum 157, 174<br />

Neurospora sp. 73, 89, 395, 402, 407<br />

N. crassa 3, 580<br />

Nicotiana attenuata 243<br />

N. tabaccum 243, 248<br />

Nigericin 82<br />

Nitrate reduction 405–409<br />

Nitrate transport 394, 407, 409<br />

Nitrifying bacteria 72<br />

Nitrogen cycling 381<br />

Nitrogen metabolism 568<br />

Nitrogen status 383<br />

Nitrogen uptake and translocation 394<br />

Nitrogenase 86<br />

Nitrosomonas europeae 3<br />

Nocardia sp. 73, 89<br />

Nod factors 2, 107, 108<br />

nodABC genes 109<br />

Nomarski interference contrast<br />

Nonmycorrhizal 77<br />

Nonmycorrhizal fungi 597<br />

Nonrecombinant plaques 577<br />

Nonrhizosphere 82, 197<br />

Nostoc sp. 73<br />

Nostoc sp. PCC 7120 3<br />

Notch trafficking 100<br />

Nuclear movement 300, 313<br />

Nylon membrane 584<br />

Subject Index 623<br />

O<br />

Oidiodendron sp. 79<br />

Oligo dT primer 584<br />

Oligo nuc1eotide 584<br />

Oligonucleotide probes 8<br />

Oligosaccharide 505, 514, 529<br />

Oligotrophic 124, 129<br />

Oospores 27,436<br />

Orchidaceous mycorrhizas 76<br />

Orchids 246<br />

Organotrophs 74<br />

Ornithine carbamoyl transferase 580<br />

Oryza sativa 243<br />

O. sativa L. ssp. indica 3<br />

O. sativa L. ssp. japonica 3<br />

Ovis aries 580<br />

Oxidases 124<br />

P<br />

Paenibacillus sp. 89<br />

Parafilm 576<br />

Parasexual recombination 172<br />

Parasites 1<br />

Parenthosomes 595<br />

Particle bombardment 124<br />

Pathogen attraction 74<br />

Paxillus involutus 199, 200, 260, 597<br />

P. involutus 201<br />

PCR 569, 583<br />

PCR anchor primer 585, 588<br />

PCR buffer 585, 588<br />

PCR products 584<br />

PCR reaction mix 583<br />

PCR reactions 582<br />

PCR-base approaches 354<br />

PCR-based techniques 10<br />

PCR-Fingerprinting 551<br />

PCR-RFLP 551<br />

PCR-single-strand conformation polymorphism<br />

(SSCP) 354<br />

PCR-temperature gradient gel electrophoresis<br />

(TGGE) 354<br />

Pectin 222<br />

Pectinases 80<br />

Pelotons 247<br />

Penetration 259, 273


624<br />

Subject Index<br />

Penicillium sp. 72,73, 89<br />

P. bilalii 85<br />

P. griseofulvum 88<br />

PEP carboxykinase 590<br />

Peptidases 54<br />

Peptide mass fingerprint 205<br />

Perithecium 168<br />

Perithiquious flagella 122<br />

Peroxidase 303, 524, 525, 529<br />

Pestatoria 89<br />

Pesticides 113, 122<br />

Petroselinum crispum 243<br />

PGPR 82, 355<br />

Phage buffer 575<br />

l-phage packaging mix 575<br />

Phanerochate chrysosporium 590<br />

Phase-contrast light microscopy 504,<br />

510, 516, 521, 524, 528<br />

Phaseolus aureus 245<br />

pH-dependent regulation 382<br />

Phenotypes 51,75<br />

Phenylacetic derivatives 88<br />

Phenylpropanoids 102<br />

Phomopsis sp. 287<br />

Phosphatases 59, 80<br />

Phosphate 80<br />

Phosphate-solubilizing microorganisms<br />

84<br />

Phosphate-solubilizing rhizobateria<br />

360<br />

Phosphate metabolism 580<br />

Phosphatidylinositol 101<br />

Phospholipid fatty acid (PLFA) 75<br />

Phosphor screen 585, 588<br />

Phosphorimager 569, 585, 588<br />

Phosphorus-rich soils 613<br />

Phyllosphere 4, 122, 147, 532, 535, 540<br />

Phyllosticta 287<br />

Phylogenetic probes 452<br />

Phylogenetic relationships 332<br />

Physiological heterogeneity 380<br />

Phytoestrogens 105<br />

Phytohormones 4, 88, 202<br />

Phytopromotional 245<br />

Phytotoxins 82<br />

Picea abies 200, 212<br />

Piloderma croceum 252<br />

Pinus pinea 200, 201<br />

P. resinosa 589<br />

P. sylvestris 199, 200<br />

384-Pin dot blot tool 584<br />

Piriformospora indica 237, 352, 597<br />

Pisolithus alba 201<br />

P. tinctorius 201, 220, 261, 597<br />

Pisum sativum 245, 255, 333<br />

Plant cell wall architecture 505, 522<br />

Plant growth promotion 133,137, 489<br />

Plant litter 373<br />

Plant survival 613<br />

Plaque forming units (pfu) 576<br />

Plasmid miniprep kit 577<br />

Plasmid vectors 439<br />

PLFA profiling 75<br />

Pligotrophic 4<br />

Poa ampla 164, 174<br />

Polarized growth 304<br />

Polarized light microscopy 524, 528<br />

Poly-A RNA 569<br />

Polyamies 251<br />

Polyethylene/CaCl 2 -mediated transformation<br />

437, 442<br />

Polygalacturonase 174, 261, 522<br />

Polymerase chain reaction (PCR) 75<br />

Polymerises 65<br />

Polyphenol oxidases 80<br />

Polyphosphate 335<br />

Polyubiquitine 580<br />

Populus tremula 243, 252<br />

P. tremuloides Michx. (clone Esch5) 243<br />

Powerscript reverse transcriptase 584<br />

Prehybridization solution 585, 588<br />

Pre-mRNA cleavage factor 580<br />

Primer for RAPD 554<br />

Primordia 160, 260<br />

Principal component analysis 562<br />

Proliferation 248<br />

Propagules 6<br />

Prophylactic 361<br />

Prosopis chilnensis 243<br />

P. juliflora (Sw.) DC. 243<br />

Protease 80, 82, 122, 382, 417<br />

Protease inhibitors 124<br />

14-3-3 Protein 580<br />

Proteinase K 572<br />

Proteobacteria 200<br />

Proteolytic 67, 128<br />

Proteome 203<br />

Protocorm 247, 299<br />

Protoplasts 79, 437<br />

Protozoans 56<br />

Protrusions 604<br />

Prunus 152<br />

Pseudomonas 2, 63, 82, 152, 198, 477<br />

Ps. putida 9, 90, 238, 456


Ps. aeruginosa 3<br />

Ps. chlororaphis 201<br />

Ps. fluorescence 2, 198, 238<br />

Ps. synringae 3<br />

Ps. chlororaphis 439<br />

Pseudotsuga menziesii 200<br />

P-solubilizing bacteria 84<br />

Puccinia graminis 580<br />

pVSl 21<br />

Pyoverdines 90<br />

Pyoverdine siderophores 90<br />

Pythium sp. 89<br />

P. ultimum 435, 436<br />

Q<br />

Qiaquick columns 585, 588<br />

Quantitative microscopy 503, 504<br />

Quantity one Software 585, 588<br />

Quercus robur 243, 252<br />

Quorum sensing 543, 544<br />

R<br />

Raffinose 602<br />

Random primer labeling 585, 588<br />

RAPD 551, 553<br />

Ras related protein 590<br />

Receptor site 514, 522, 529<br />

Receptor-like kinase 99<br />

Recombinant plaques 577<br />

Red fluorescent protein (drFP 583 or<br />

DsRed) 441<br />

Red pine 589<br />

RedTaq DNA polymerase 578, 582<br />

Regulatory pathways 101<br />

rep 21<br />

Reporter constructs 449, 456<br />

Reporter gene 19<br />

Restionaceae 76<br />

Rhamnose 602<br />

Rhicadhesin 508<br />

Rhizobacteria sp. 4, 355<br />

Rhizobium sp. 73, 82, 184, 503, 532,544<br />

Rhizobium etli 2<br />

R. meliloti 85, 90<br />

R. tropici 2<br />

Rhizobium-legume symbiosis 81, 503,<br />

529, 533, 534<br />

Rhizobium-rice association 531,541,542<br />

Rhizoctonia sp. 73, 85<br />

R. bataticol 597<br />

R. solani 157, 256, 597<br />

Rhizodeposition 67, 126<br />

Subject Index 625<br />

Rhizodermal 259<br />

Rhizodermis 4, 256<br />

Rhizoids 247<br />

Rhizoplane 8, 127, 450, 458<br />

Rhizopogon roseolus 89, 597<br />

R. vulgaris 597<br />

Rhizopus sp. 88, 89<br />

R. microsporus 90<br />

R. oryzae 240<br />

R. stolonifer 240<br />

Rhizosphere 2, 38, 197<br />

Rhizosphere colonization 352<br />

Rhizosphere compartments 450, 464<br />

Rhizosphere interactions 442<br />

Rhizosphere of a mycorrhizal <strong>plant</strong> 358<br />

Rhizosphere/rhizoplane 529, 530, 540<br />

Rhizosphere-stable plasmid 21<br />

Rho GTPase590<br />

Rhythmic 600<br />

Ribosomal Database 559<br />

Ribosomal genes (rRNA) 354<br />

Ribosomal intergenic space analysis<br />

(RISA) 75<br />

Ribosomal RNA/DNA 461, 462<br />

16S ribosomal RNA-directed 8<br />

16S rRNA gene amplification 355<br />

Ribosomal sequences 579<br />

Rice 35<br />

Rickettsia prowazekii 3<br />

RNA Extraction buffer 569<br />

RNAse-fTee DNAse 569<br />

Robustum 158<br />

Root 38<br />

Root colonization 13, 78,450, 533, 540,<br />

544<br />

Root exudates 101<br />

Root exudation 38<br />

Root hair attachment 505,508, 511, 516<br />

Root hair deformation 505, 508<br />

Root hair infection 505, 509, 515, 529<br />

Root hair infection thread 509, 524, 529<br />

Root hair tips 111<br />

Root proliferation 314<br />

RT reactions 585, 588<br />

Russula aeruginea 261<br />

R. foetens 261<br />

R. violeipes 261<br />

S<br />

S238 N 201<br />

Saccharomyces cerevisiae 3, 580, 597<br />

S. pombe 590


626<br />

Subject Index<br />

Salmon sperm DNA 585, 588<br />

Salmonella typhimurium 439<br />

Sapotaceae 76<br />

Saprobes 69, 287<br />

Saprophytic fungi 78<br />

Saprotrophic 79<br />

Scanning electron microscopy 507, 527<br />

Scatter plot analysis 589<br />

Schizophyllum commune 203, 590, 597<br />

Schizosaccharomayces pombe 3,580, 590<br />

Scleroderma citrinum 261<br />

Sclerotinia homeocarpa 158<br />

S. sclerotiorum 597<br />

Sc. solani 597<br />

Scutellospora gilmorei 248, 597<br />

S. calospora 334<br />

SDS 586<br />

Sebacina vermifera 239<br />

S. vermifera var senu 597<br />

Secondary metabolites 288<br />

Seed disinfection 15<br />

Seed inoculation 17<br />

Septin Spn3 590<br />

Serratia 89<br />

S. liquefaciens 456<br />

Setaria italica 244, 245<br />

Sfi I enzyme 573<br />

Sheered hyphae 162<br />

Shepherd’s crook 508, 509, 524<br />

Short root 212, 297<br />

Short root branching 315<br />

Siderophores 88, 90, 202, 200<br />

Signal molecules 106<br />

Signal perception 111<br />

Signal transduction 568<br />

Sinorhizobium sp. 82<br />

S. meliloti 3, 555<br />

Small GTPases 305<br />

SMART cDNA library construction kit<br />

569<br />

SMART cDNA synthesis kit 584<br />

SMART III Oligonucleotide 569<br />

SMART IV 584<br />

Sodium alginate 79<br />

Sodium hypochlorite 15<br />

Solanum melongena 243, 248<br />

S. xanthocarpum 77<br />

Solidification 253<br />

Sorghum vulgare 243<br />

Spatial distribution of microbes<br />

532–544<br />

Spatial isolation 73<br />

Specific efflux mechanisms 80<br />

Spectrophotometer 569<br />

Spermatia 165<br />

Spilanthes calva 243, 248<br />

Spinacia oleracea 255<br />

Spitzenkörper 311<br />

Spliceosome-associated protein 580<br />

Sporocarps 200<br />

Sporobolus sp. 77<br />

Sporodochia 169, 170<br />

Sporulation 255, 598<br />

sscDNA 585, 588<br />

sss (site-specific recombinase) 25<br />

sta 21<br />

Staphylococcus 200<br />

S. hycius 90<br />

Stomates 145<br />

Straw 40<br />

Streptomyces sp. 73, 82, 89<br />

Streptoverticillium cinnamoneum 88<br />

Styela plicata S 580<br />

Suberin layer 214<br />

Substrate utilization profile 463<br />

Subunit G of vacuolar ATP synthase<br />

580<br />

Sucrose 602, 604<br />

SUG1 subunit 8 590<br />

Sugar regulation 377, 378<br />

Suillus bovinus 199, 203<br />

S. granulatus 201, 261<br />

S. grevillei 200, 261<br />

S. luteus 261<br />

S. variegatus 597<br />

Sulfate-reducers 72<br />

Superscript II 569<br />

Survival 245<br />

Suspension 256<br />

Symbionts 1<br />

Symbiosis 60, 295, 567<br />

Symbiosis-specific manner 79<br />

Symbiosome membrane 99<br />

Symbiotic communication 114<br />

Symbiotic fungi 60<br />

Symbiotic hyphal growth 306<br />

Symptoms 242<br />

SYMRK 99<br />

Synchytrium sp. 88<br />

Synergistic microbial interactions 360<br />

T<br />

TAE agarose gel 583<br />

TAE buffer 583


T4 DNA ligase 575<br />

Tagetes erecta 243, 248<br />

Tagging bacteria 439<br />

TAMRA 558<br />

Taq DNA polymerase 585<br />

Tectona grandis 243<br />

TEF 590<br />

Tephrosia purpurea 243<br />

Terminal restriction fragment length<br />

polymorphism (T-RFLP) 75<br />

Termnalia arjuna 243, 247<br />

Thermal cycler 568, 569<br />

Thiobacillus sp. 73<br />

Tissue permeabilization 317<br />

Tissue-culture 613<br />

Titration 577<br />

Tn7 440<br />

TnSlacZ 17<br />

Tomato foot and root rot 431<br />

Tomycocol 274<br />

Transformation 124<br />

Transcriptional factor StuA 580<br />

Transcriptional regulation 582<br />

Transformation of fungi 437<br />

Transgenic manipulation 79<br />

Transgenic <strong>plant</strong>s 121, 124<br />

Transition zone 301<br />

Trans-Kingdom 158<br />

Transmission electron microscopy 509,<br />

512, 522<br />

Transpiration 149<br />

Trans<strong>plant</strong>s 253<br />

Transporter 80, 375<br />

Transposon vectors 439<br />

Trehalase 199, 374<br />

Tremelloid haustoria1 cells 275<br />

Tricalcium phosphate 84<br />

Tricarboxylic acid cycle 54<br />

Trichoderma sp. 73, 83<br />

T. harzianum 85, 86, 164<br />

T. viride 87<br />

Tricholoma imbricatum 261<br />

T. lascivum 261<br />

T. scaplpturatum 261<br />

T. subannulatum 261<br />

T. ustaloides 261<br />

Trifolium alexandrium 78<br />

T. repens 85, 333<br />

Truncated genes 124<br />

Tryptic soy agar (TSA) 433<br />

Tryptic soy broth (TSB) 435<br />

dTTP 582<br />

Tuber sp. 261<br />

Tubulin expression 294<br />

a-Tubulin 590<br />

Tubulin genes 294<br />

Tubulin-GFP 318<br />

Type III secretion systems 113<br />

U<br />

Ubiquitinine-1 124<br />

Ultrastructure 211<br />

Unstable gfp 441<br />

UPGMA 562<br />

Ustilago sp. 88<br />

U. maydis 590, 597<br />

Utilization of proteins 417, 418<br />

UV-crosslinker 584<br />

V<br />

Vacuolar motility 313<br />

Vacuum manifold 583<br />

Verticillium sp. 89<br />

Vicia faba 342<br />

Video microscopy 505, 527, 528<br />

Vigna radiata 245<br />

Vip proteins 124<br />

Viridochromogenes 82<br />

Virulence factor 123<br />

Virulent root 240<br />

Vitamins 106<br />

W<br />

Wall-degrading enzymes 262<br />

Water permeability 480<br />

Waters AccQ. Tag Method 9<br />

Well 96-PCR plates 582<br />

Wetting 472<br />

Wilcoxon-Mann-Whitney V-test 18<br />

Withania somnifera 243, 247<br />

WPM 253<br />

X<br />

Xanthomonas campestris 3<br />

Xenopus laevis 590<br />

Xerocomus chrysenteron 261<br />

X. subtomentosus 261<br />

X-gal 576<br />

Xylanase 261<br />

Xylene cyanol 573<br />

Xylose 173, 602<br />

Subject Index 627


628<br />

Subject Index<br />

Y<br />

Yellow fluorescent protein (YFP) 441<br />

Yersinia pseudotuberculosis 439<br />

Z<br />

Zea mays 78, 244, 245<br />

Zizyphus nummularia Burm. fil. 243<br />

Zoosphere 361<br />

Zygomycota 596<br />

Zygomycotina 76<br />

Zygophylaceae 76

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!