Next Article in Journal
Investigating the Correlation between Electrolyte Concentration and Electrochemical Properties of Ionogels
Next Article in Special Issue
Comparative Analysis of Chemical Composition of Zanthoxylum myriacanthum Branches and Leaves by GC-MS and UPLC-Q-Orbitrap HRMS, and Evaluation of Their Antioxidant Activities
Previous Article in Journal
Broad-Spectrum, Potent, and Durable Ceria Nanoparticles Inactivate RNA Virus Infectivity by Targeting Virion Surfaces and Disrupting Virus–Receptor Interactions
Previous Article in Special Issue
Functional Study and Efficient Catalytic Element Mining of CYP76AHs in Salvia Plants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chemistry, Bioactivity, and Prediction of the Quality Marker (Q-Marker) of Ferula Plants in China: A Review

1
International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 Min-Zu Road, Wuhan 430074, China
2
College of Biological and Geographical Sciences, Yili Normal University, Yining 835000, China
3
Traditional Kazakh Medicine Research Institute of Ili Kazakh Autonomous Prefecture, Yining 835000, China
4
Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School, Shihezi University, Shihezi 832003, China
5
Research Division of Medicinal Plants, Research Institute of Forests and Rangelands, Agricultural Research Education and Extension Organization (AREEO), Tehran P.O. Box 13185-116, Iran
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(13), 5191; https://doi.org/10.3390/molecules28135191
Submission received: 24 May 2023 / Revised: 16 June 2023 / Accepted: 19 June 2023 / Published: 4 July 2023

Abstract

:
The genus of Ferula belongs to the family Apiaceae, and many Ferula plants are used as traditional Chinese medicines. Ferula plants were initially identified as early as the “Newly Revised Materia Medica” written in the Tang Dynasty (AD 659), and several of them are also recognized as the traditional medicines of the Uygur, Kazakh, and Mongolian. Ferula plants are distributed in China, Russia, India, Africa, Central Asia, and other places. Currently, the chemical components derived from Ferula plants are mainly coumarins, sesquiterpenes, and volatile oils. Ferula plants can exhibit diverse pharmacological activities such as anti-allergy, analgesia, relieving cough, anticoagulation, and anti-tumor. Therefore, this article summarized the domestic research conducted on the genus Ferula, appropriately combines the research status of the foreign genus Ferula, and describes the chemical composition, biological activity, toxicity issues, and Q-marker prediction. In addition, all the related studies about the genus Ferula are summarized by analyzing the various databases such as CNKI, Wanfang data, PubChem and SciFinder.

1. Introduction

Ferula is a tall perennial herb which belongs to the Apiaceae family, and these plants are commonly characterized by a peculiar pungent garlic odor. There are more than 180 species of Ferula found worldwide [1] and they are mainly located in Central Asia, the Middle East, Siberia, and South Asia [2]. In China, Ferula is mainly distributed in Xinjiang, and a small number of Ferula plants are also found in Gansu, Shanxi, Inner Mongolia, and Tibet [3,4]. The genus Ferula in China primarily comprises more than twenty species, with a few representative plants shown in Figure 1.
The plants belonging to the genus Ferula (Apiaceae) have been implicated in the treatment of various ailments and have a long history of use in many traditional medicines. For instance, Nepali people use Ferula in their daily diets [11,12], and it is used locally as a traditional remedy in Saudi Arabia for the treatment of skin infections [13]. In addition, Ferula plants are widely used in India in food and as a medicine in Indian systems of medicine such as Ayurveda [14]. Interestingly, in China, Ferula plants are also used as an Uyghur medicinal herb, and have a long history of beneficial application [15]. Ferula has already been added to the Uyghur Medicine Criteria [16] and the calendar version of the Chinese Pharmacopoeia [17]. In other ethnic groups and regions of China, Ferula is often used as traditional medicines. The traditional uses of Ferula include beneficial effects against stomach pain, flatulence, intestinal parasites, indigestion, asthma, and flu [18]. For instance, the Kazakhs have used F. soongarica to treat headaches, colds and stomach aches, F. caspica can treat nervous breakdown, and F. lehmannii is used as an anti-parasitic, for anti-malnutrition and to cure cold and pain in the heart and abdomen [12,15]. A number of modern pharmacological and biological studies have indicated that Ferula has anticancer [19,20], anticarcinogenic [21], antimicrobial [22], antidiabetic [23], anti-flu [24], anti-inflammatory [25,26], and cardiovascular system effects [27], as well as use against Alzheimer’s disease [28,29] and in anti-neuropathic pain [30] activities.
Ferula contains a variety of chemical compounds, primarily coumarins [31], particularly sesquiterpenes coumarins [32], volatile oils [33], sulfur-containing compounds [34], and aromatic compounds [35], all of which exhibit diverse biological activities. However, the Q-marker prediction studies of Ferula are relatively scarce. Ferula plants of China are mainly distributed in Xinjiang, and it is less commonly present in other regions. Thus, in addition to the over-exploitation and collection of Ferula resources, the quality of Ferula herbs available on the market is a matter of concern due to the prevalence of counterfeits and inferior species that are accessible to consumers. Furthermore, the genus Ferula seed plant breeding has been continuously shrinking due to the rapid land development, irrigation, highway building, leading to significant decrease in Ferula resources. Therefore, novel strategies have to be developed to protect the resources of Ferula plants.
Traditional Chinese medicines (TCMs) play an important role in China’s modern health care system and have been found to be efficacious in clinical practice. However, due to the lack of criteria for the selection of quality control indicators, both the certification and efficacy of herbal medicines remain unclear [36,37]. Therefore, new approaches are needed to establish Q-markers for determining the quality standards of traditional Chinese medicines, and for identifying the appropriate chemicals present in plants to use them as potential quality indicators [38,39].
There are quite a few Q-marker studies reported for the genus Ferula. Thus, the identification of the various markers for the genus Ferula by the Q-marker method is essential for the identification of its authenticity and the conservation of the plant resources.

2. Distribution of Ferula Plants in China

There are more than 180 species of genus Ferula in the worldwide, and more than 20 species have been recorded in China. The genus Ferula resources are mainly distributed in Xinjiang and a small number of other provinces in China (Table 1). Most Ferula plants are distributed at altitudes between 400 and 3500 m, and the distribution environment is mainly the desert and mountainous areas. However, in recent years, due to the destruction of the plant resources of the genus Ferula, wild Ferula is nearly extinct.

3. Research on the Chemical Composition of Ferula Plants in China

3.1. Coumarins

The full name of 7-hydroxycoumarin is 7-hydroxycoumarin O(7)-glucosiduronic acid. It is a beta-D-glucosiduronic acid. Coumarins are found in the genus Ferula, and most of them are derivatives with 7-hydroxycoumarin as the parent nucleus [56]. Coumarins in the genus Ferula can be further divided into different types of coumarins according to their substituents, mainly including sesquiterpene coumarins and monoterpene coumarins. Among them, sesquiterpene coumarins can be divided into bicyclic sesquiterpene coumarins (A), monocyclic sesquiterpene coumarins (B), and straight-chain sesquiterpene coumarins (C), in addition to some furan coumarins (D) [57] and other coumarins (E) (Table 2, Figure 2, Figure 3 and Figure 4). Researchers obtained a large number of coumarin-like substances from Ferula sinkiangensis, F. lehmannii, F. feruloides and F. fukanensis [58,59,60,61,62,63,64,65,66].

3.2. Sesquiterpenoids and Their Derivatives

In addition to the above-mentioned coumarins, sesquiterpenes are the second most abundant substances in Ferula plants. The sesquiterpenes of the genus Ferula are mainly carotene-type sesquiterpene parent nuclei, but carotene-type sesquiterpenes are rare in domestic Ferula plants and are mostly concentrated in foreign Ferula plants. The researchers extracted the following sesquiterpenoids from F. ferulaeoides, F. caspica, F. fukanensis and F. jaeschkeana [67,68,69,70,71] (Table 3, Figure 5 and Figure 6).

3.3. Volatile Oil

The volatile oils in genus Ferula are mainly terpenoids and polysulfide compounds [72]. Among the genus Ferula of China, the number of terpene components can reach more than 80% in volatile oils, mostly are monoterpenes and sesquiterpenes, while polysulfide compounds are dominated by disulfides, trisulfides, bis-disulfides, and thio-disulfides. A large amount of volatile oil was extracted from F. sinkiangensis, F. fukanensis, F. ferulaeoides, and F. ovina [73,74,75,76] (Table 4). Min Zhi-da et al. [77] identified 26 polysulfides by gas chromatography-mas spectrometry (GC-MS (CI/EI)) in F. sinkiangensis and F. fukanensi (Figure 7).

3.4. Aromatic Compounds

Studies showed that ethyl p-hydroxybenzoate (256), emodin (257), and 1,3,7-trihydroxy-6-methyl-xanthone (258) were isolated from F. fukanensis [78]. Ferulic acid (259) was isolated for the first time from F. sinkiangensis [62]. 4,5-Dimethoxy-2,3-methylenedioxyphenylpropane-7-adamate was isolated from F. licentiana [79] and named as Tong Shan ferulic acid A (260). 2,4-Dihydroxy-α-oxo-phenylacetic acid (261), 3,3′,4,4′-biphenyltetracarboxylic acid (262) and 2,4-dihydroxybenzophenone (263) were isolated from the rhizome of F. songarica [80]. 2,4-Dihydroxy-α-oxo-phenylacetic acid was first isolated from F. songarica and 3,3′,4,4′-biphenyltetracarboxylic acid was a newly discovered compound (Figure 8).

3.5. Other Substances

Veratric acid (264) and β-sitosterol (265) were isolated from F. licentiana [81]. Acetophenone compounds such as (1S,2R,4S)-(−)-borneol acetate (266), (5Z)-2,6,10-trimethyl-1,5,9-undecatriene (267), (Z)-β-farnesene (268), humulen-(v1) (269), ±-trans-nerolidol (270), and carotene (271) were isolated from F. ferulaeoide [82,83] (Figure 9). Seventeen amino acids, including lysine, histidine, arginine, aspartic acid and threonine, were obtained from the roots and leaves of F. lehmannii Boiss [84]. Studies have shown that a series of trace elements including potassium, aluminum, calcium, copper, magnesium, barium, cadmium, and cobalt were isolated from F. sinkiangensis [85]. Radiatinol (272) and scopoletin (273) were isolated from F. dissecta [86].

4. Advances in the Bioactivity of Ferula Plants in China

With the gradual increase in domestic research in the field of biological activity of the genus Ferula, the bioactive compounds were isolated from the Ferula plant by various applied extraction techniques [87], and a number of different compounds with anti-tumor [88], anti-allergy, anticoagulation [89], bacterial inhibition [90], action in the gastrointestinal system [91], nervous system [92], cardiovascular system [93,94], and other pharmacological activities have been found in the genus Ferula [95,96,97].

4.1. Anticancer Activity

The main chemical components of Ferula plants that exert anti-tumor effects are ferulic acids [98], sesquiterpenoids [99], and volatile oils [100]. For instance, Wu Jing [101] found that the inhibition rate of A549 lung cancer xenografts in nude mice gradually increased with increase in dose of ferulic acid administration. The low (25 mg/L), medium (50 mg/L) and high (100 mg/L) dose groups inhibited the expression levels of both mTOR protein and mRNA in the tumor tissues of the nude mice, and decreased the expression levels of Ki-67 protein, which decreased with increasing dose, and also enhanced the expression levels of Caspase-3 protein. The investigators suggested that ferulic acid can attenuate growth of lung cancer probably by downregulating mTOR protein expression, inhibiting Ki-67 expression and promoting Caspase-3 levels. The petroleum ether, n-butanol, ethyl acetate, trichloromethane, and aqueous fractions, as well as volatile oil and 95% ethanol extract of F. ferulaeoides were screened for their pharmacological activities, among which trichloromethane and ethyl acetate fractions exhibited certain inhibitory effects on five distinct gastric cancer cell lines, whereas trichloromethane fractions showed the strongest inhibitory effect on gastric cancer cells SGC-7901 (IC50: 7.98 ± 2.62 mg/L). In addition, volatile oil displayed strong inhibition of cell proliferation on gastric cancer cells AGS (IC50: 8.73 ± 0.55 mg/L), but n-butanol fraction and aqueous fraction showed no significant inhibitory effects on these five gastric cancer cell lines (AGS,MKN-45,BGC-823,MGC-803,SGC-7901) [102]. A sesquiterpene coumarin-like substance,2,3-dihydro-7-hydroxy-2R*,3R*-dim-ethyl-2-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c]coumarin (DAW22), was isolated from F. ferulaeoides root, and was subsequently reported to induce apoptosis in C6 glioma cells through the mitochondria-mediated, endoplasmic reticulum stress and death receptor pathway. The dosing concentration is 20 µM [103]. It also suppressed the proliferation of five different established human Malignant peripheral nerve sheath tumors (MPNST) cancer cell lines (STS-26T, S462, S462-TY, ST8814, T265) by promoting their apoptosis [104]. Moreover, experimental animal studies have revealed that in an immunocompromised nude mouse model with subcutaneous transplantation of STS-26T cells, when DAW22 was administered intraperitoneally at a daily dose of 60 mg/kg/day for 4 weeks, it significantly inhibited tumor xenografts compared to the control group. It was found by Western blotting analysis, that DAW22 inhibited phosphorylation of AKT and ERK and reduced the level of Non-phospho (Active) CTNNB1, there by resulting in an inhibitory effect on cell proliferation and suppression of tumor growth in for MPNST cell lines. These results supported the use of DAW22 as an alternative therapeutic compound to MPNST which can effectively produce anticancer effects by affecting multiple signaling pathways in tumor disease progression. 8-p-Hydroxybenzoyl tovarol (TAW) was isolated from the roots of F.dissecta, and found to inhibit the growth of human cervical cancer HeLa cells by promoting protective autophagy, and in the future, advanced molecular dynamics techniques and free energy calculations and reverse molecular docking can be used to reveal the effects of the major coumarins, sesquiterpenoids, and terpenoids in the genus Ferula on the anticancer mechanism, and there are already documented studies of other plants using similar measurement techniques [105,106].

4.2. Antibacterial Activity

Extraction studies on three distinct kinds of dry roots of F. sinkiangensis, F. lehmannii Boiss. and F. ferulaeoides revealed that the alcoholic and macerated extracts displayed antibacterial effects on Staphylococcus aureus, Bacillus subtilis, and Sporosarcina. The MIC about F. sinkiangensis root’s alcoholic extracts for three bacteria were 31.25 mg/mL, 3.92 mg/mL, and 1000 mg/mL. The MIC about F. sinkiangensis root’s macerated extracts for three bacteria were 1000 mg/mL, 1000 mg/mL, and 1000 mg/mL. The MIC about F. ferulaeoides root’s alcoholic extracts for three bacteria were 3.91 mg/mL, 3.91 mg/mL, and 15.63 mg/mL. The MIC about F. ferulaeoides root’s macerated extracts for three bacteria were 500 mg/mL, 500 mg/mL, and 1000 mg/mL. The MIC about F. ferulaeoides root’s alcoholic extracts for three bacteria were 7.81 mg/mL, 3.91 mg/mL, and 15.63 mg/mL [107]. A total of 27 compounds were extracted and isolated from F. ferulaeoides, and 7 of the 27compounds were found in the plant for the first time. These 27 compounds were screened for their potential antibacterial activity, and only a few coumarins among the 27 compounds were found to affect multi-drug resistant (MDR) and methicillin-resistant Staphylococcusaureus (MRSA) bacteria, and six of them showed inhibition against tetracycline-resistant strain XU212 [108]. In addition, three new sesquiterpene derivatives were obtained from F. ferulaeoides [109] and it was observed that their minimum inhibitory concentrations (MIC) ranged from 0.5–128 μg/mL. It was found that the volatile oil of the seeds of F. olivacea exhibited an inhibitory effect on the Gram-positive bacteria such as Enterococcus faecalis and S. aureus [110,111].

4.3. Anti-Allergic, Anti-Inflammatory, and Immunosuppressive Effects

Using sodium cromoglycate (10 mg/kg) as the control group and volatile oil of F. sinkiangensis (50 mg/kg) as the experimental group, experiments were conducted to evaluate the potential effect of passive skin allergic reaction in mice, the effect of Arthus reaction in rabbits, and the antigen sensitization confrontation in the sensitized guinea pigs [112]. It was found that the F. sinkiangensis volatile oil (200 mg/kg) could significantly inhibit Ig E response to allergic reactions, and the degree of effect was similar to that of sodium cromoglycate. Ferulic acid can also reverse skin sensitization in rats and suppress the delayed hypersensitivity reactions in mice, thus indicating that ferulic acid has inhibitory effect on allergic reactions [113]. In another study, designed to examine the impact of the raw juices of F. sinkiangensis, F. conocaula, and F. feruloides, it was found that all three types of Ferula plants exhibited significant inhibitory effects on foot topography swelling (the dosing concentrations were F. sinkiangensis: 90 mg/kg, F. conocaula: 90 mg/kg, F. feruloides: 80 mg/kg)and capillary permeability (the dosing concentrations were F. sinkiangensis: 34 mg/kg, F. conocaula: 34 mg/kg, F. feruloides: 30 mg/kg) in rabbits induced by p-hornwort, and on the delayed hypersensitivity (the dosing concentrations were F. sinkiangensis: 98 mg/kg, F. conocaula: 97 mg/kg, F. feruloides: 78 mg/kg) and serum hemolysin formation (the dosing concentrations were F. sinkiangensis: 98 mg/kg, F. conocaula: 97 mg/kg, F. feruloides: 78 mg/kg) in mice induced by sheep erythrocytes or dinitrochlorobenzene [114].

4.4. Anticoagulant Effect

The extract of F. lehmannii was obtained by using 95% ethanol, and an anticoagulant assay was then performed. The experimental results revealed that F. lehmannii extract inhibited the endogenous coagulation pathway and showed an inhibitory effect on the activity of thrombinogen. The dosing concentrations were: the high-dose group (100 mg/kg), the medium-dose group (50 mg/kg), and the low-dose group (25 mf/kg) [115]. Radiatinol and scopoletin were isolated from F. dissecta, and both these compounds acted in the endogenous coagulation pathway and prolonged the prothrombin time as well as activated partial thromboplastin time at different concentrations [86].

4.5. Effects on the Cardiovascular System

Interestingly, another study showed that after ferulic acid was administered to C57 mice in the heart failure preclinical model, the mice in the heart failure administration group exhibited different degrees of improvement in cardiac left ventricular ejection fraction, end-systolic and end-diastolic internal diameter changes, collagen area percentage changes, and protein expression levels compared to the control mice. Ferulic acid could improve ventricular remodeling to some extent [116]. In addition, studies conducted with the hydroalcoholic extracted fractions and the aqueous decoction of F. sinkiangensis revealed that they could reduce the heart amplitude and enhance the heart rate in isolated frog hearts [117].

4.6. Effects on the Gastrointestinal Tract

It has been reported in the literature that F. sinkiangensis has been used to treat stomach disorders in Xinjiang [118]. The volatile oil and resin of F. sinkiangensis were found to be effective in three distinct gastric ulcer models, among which the volatile oil was found to be relatively better [119]. The rat model of acetic acid-injected gastric ulcer was established, and the model was tested by administering F. sinkiangensis original herbs and F. sinkiangensis prepared herbs by a folk concoction (0.048 g/mL), frying method (0.048 g/mL), a vinegar moxibustion method (0.048 g/mL), and a boiling method (0.048 g/mL). It was found that the pH value of the rat stomach increased significantly, but the area of gastric ulcer decreased markedly after the administration of F. sinkiangensis original herbs and F. sinkiangensis prepared herbs [120]. In addition, studies on the raw juices of F. feruloides and F. sinkiangensis revealed that these juices can display inhibitory effects on gastric ulcer models in rats with acetylsalicylic acid and can effectively suppress the autonomous activity of smooth muscle in isolated intestinal tubes of rabbits [121]. The compounds were isolated from the F. sinkiangensis resin and they were analyzed in anti-ulcer vivo activity experiments. It was observed that compared with the positive control group famotidine, CHCl3 extract (1.3515 g/kg) showed the best anti-ulcer activity [122].

4.7. Action on the Nervous System

A network pharmacological analysis of Ferula resulted in identification of 12 key active components against Alzheimer’s disease, including Farnesiferol A, Farnesiferol B, conferol, and ferulic acid. It was predicted that Ferula can predominantly act through by modulating 14 key targets in the cholinergic synaptic signaling pathway and AD signaling pathway [123]. A bioactivity-oriented study on F. sinkiangensis showed that F. sinkiangensis significantly inhibited NO production induced by over-activated microglia and exhibited anti-neuroinflammatory effects [58]. Interestingly, a compound, Kellerin, extracted from F. sinkiangensis, was found to produce substantial effects in the rat middle cerebral artery occlusion (MCAO) model, lipopolysaccharide (LPS)-activated microglia model, mouse bilateral common carotid artery occlusion (BCCAO) model and lipopolysaccharide (LPS)-activated microglia model. Kellerin was found to markedly improve the neurological outcomes, reduce the size of cerebral infarcts and decrease brain edema in the rat MCAO model, and the dosing concentrations were: the low-dose group (3.5 mg/kg), the medium-dose group (7.0 mg/kg) and the high-dose group (14.0 mg/kg). Moreover, in the pathological conditions of focal cerebral ischemia, Kellerin could attenuate neuronal damage as well as microglial activation. In addition, in vitro study which LPS-stimulated BV2 cells, revealed that Kellerin protected the neuronal cells from injury by inhibiting microglia activation. Kellerin also reduced levels of pro-inflammatory cytokines, inhibited the NF-κB signaling pathway, and decreased ROS production as well as NADPH oxidase activity, and the [124]. Kellerin was found to alleviate the cognitive impairment, reduce neuronal loss, inhibit microglia activation, and convert microglia from a pro-inflammatory M1 phenotype to an anti-inflammatory M2 phenotype in BCCAO mice. In addition, in another in vitro study, Kellerin was found to modulate the microglia polarization and inhibit NLRP3 and MAPK signaling pathways after LPS treatment [125].

4.8. Other Pharmacological Effects

Furthermore, studies have shown that Ferula resin can exhibit anti-pregnancy, anti-implantation and pregnancy termination effects [126,127]. It can also affect estrogen and progesterone levels in infertile mice and rabbits, thus modulating the smooth muscles of the uterus in mice and rabbits [128]. In another study, d-galactosamine/lipopolysaccharide acute liver injury model was established, and by comparing the d-galactosamine/lipopolysaccharide model group, the biphenyldiglyceride group and the high-, medium- and low-dose ferulic acid groups, it was observed that all the three ferulic acid groups could significantly decrease the activity of enzymes AST and ALT in mouse hepatocytes, increase the activity of antioxidant enzyme SOD as well as peroxidase GSH-Px in mice, and decrease the MDA content, thereby suggesting that ferulic acid exhibited a protective effect on acute liver injury [129]. The petroleum ether, ethyl acetate, n-butanol and methanol fractions of F. sinkiangensis displayed no substantial effect on the lipase activity, whereas the ethyl acetate, n-butanol and methanol fractions lowered total cholesterol in HepG-2 cells and triglycerides in non-alcoholic fatty liver model cells. Thus, it was hypothesized that F. sinkiangensis has certain lipid-regulating effects [130]. Four coumarin-like compounds were extracted from F. moschata showed anti-HIV activity [131].

4.9. Toxicological Studies

Acute toxicity experiments were conducted on the volatile oils of F. feruloides and F. teterrima, and the findings revealed that the LD50 of the volatile oil of F. feruloides was 10.24 g/kg and the LD50 of the volatile oil of F. teterrima was 491.61 mg/kg. After analyzing the volatile oils of both, it was found that the volatile oil of F. teterrima contained more polysulfides, accounting for 60% of the volatile oil, but no polysulfides were detected in the volatile oil of F. feruloides. Polysulfide can cause irritation and possess a garlic-like odor, so it was presumed that polysulfide was more likely to be a toxic component in F. teterrima [132]. However, the acute toxicity studies with polysulfide has not been reported previously in the literature. It was found that goats fed with F. fukanensis (2.5 g/kg) for 15 days showed obvious signs of toxicity, such as loss of appetite, anorexia, diarrhea, vitamin K-dependent reduction in coagulation factors, the toxic component of which may be possibly coumarins, but no obvious symptoms were found in these animals, such as impaired liver and platelet function [133]. Acute toxicity tests were conducted on F. sinkiangensis and F. fukanensis [134], and the LD50 values were calculated by Kou’s method. The LD50 values of F. sinkiangensis volatile oil aqueous suspension and F. fukanensi volatile oil aqueous suspension were found to be 2.82 g/kg and 1.55 g/kg; and the LD50 values of F. sinkiangensis volatile oil emulsion and F. fukanensi volatile oil emulsion were separately 0.39 g/kg and 0.41 g/kg−1. After analyzing ten batches of F. sinkiangensis test samples [135], two of them were observed to contain different ferulic acid contents. No. 1 F. sinkiangensis with (0.775 ± 1.44) mg/g and No. 5 F. sinkiangensis with (0.279 ± 1.63) mg/g, were selected and subjected to the toxicity studies by SRB method. The proliferation rates of No. 1 F. sinkiangensis and No. 5 F. sinkiangensis on the rat renal NRK cells was found to gradually increase with increasing drug concentrations, with IC50 values of No. 1 F. sinkiangensis at 60.60 mg/Land No. 5 F. sinkiangensis at 39.90 mg/L. The comparison of the IC50 values showed that the cytotoxicity of No. 1 F. sinkiangensis was significnatly less than that of No. 5 F. sinkiangensis, but the ferulic acid content of No. 1 F. sinkiangensis was significantly higher than that of No. 5 F. sinkiangensis, thus indicating that the toxicity of F. sinkiangensis may be possibly related to its chemical composition.

5. Quality Marker (Q-Marker) Prediction Analysis

With the depletion of the domestic Ferula plant resources, the phenomenon of substandard and uneven quality of Ferula plants in the market needs to be urgently solved. Thus, the quality markers (Q-marker) [136] need be established for the effective protection of the domestic Ferula plant resources. At present, the Chinese Pharmacopoeia 2020 edition stipulates that the sources of medicinal Ferula plant is the resin of F. sinkiangensis and F. fukanensi and requires that their quality marker is ferulic acid. However, ferulic acid is widely present in different plants such as Angelica sinensis [137], Ligusticum chuanxiong [138], Cimicifugafoetida L. [139], Ligusticum [140] and Fructus Toosendan [141]. It is also an important active component of many traditional Chinese medicines [142]. A large amount of sesquiterpene coumarins are found in Ferula plants, and they may be used as unique chemical components which can effectively help to identify Ferula plants by the Q-marker method.

5.1. Q-Marker Prediction Analysis by Kinship and Chemical Composition Specificity of Ferula Plants

The global distribution of Ferula plants is widespread in Central Asia, Iran and Pakistan. It has been reported that during the course of foreign research on the chemical composition of Ferula plants, it was observed that genus Ferula contained a large number of sesquiterpene coumarins with umbelliferous lactones as the parent nucleus [143], and Ferulenol was the most abundant and the first isolated coumarin from Ferula [144]. Although various sesquiterpenes and coumarins are widely distributed in the Apiaceae family, they constitute the characteristic chemical constituents of genus Ferula, and can be used as important evidence of the affinities of Ferula plants. They could also form a sound basis for the chemical taxonomy of genus Ferula [145], such as Farnesiferol A, Farnesiferol B, Farnesiferol C, and DAW22, because sesquiterpene coumarins are widespread in different plants of the genus Ferula. Additionally, the pharmacopoeia mentions ferulic acid as potentially another characteristic chemical constituent of the genus Ferula.

5.2. Q-Marker Prediction Analysis by Chemical Composition Validity

As an emerging method to evaluate and control the quality of Chinese herbal medicine Q-markers is essential to predict both the effectiveness and safety of Chinese herbal medicines. Therefore, Q-marker analysis of Chinese herbal medicines should be combined with the effectiveness of the target herbal medicines to facilitate better research on the effectiveness of Chinese herbal medicines [146,147]. Because of the presence of the different coumarin sesquiterpenoids in large quantities in the genus Ferula, most studies on the chemical composition effectiveness and safety of Ferula resources have primarily focused on these compounds, which are expected to become quality markers of Ferula plants.

5.2.1. Q-Marker Prediction Analysis by Traditional Drug Properties

The medicinal properties of Ferula plants have been recorded as Ku (bitter), Xing (pungent) and Wen (warm), as well as Guipi and Weijing (benefit for the spleen and stomach) in the Chinese Pharmacopoeia Edition (2020). The chemical compositions of the various bitter medicines in Chinese medicine are mainly alkaloids, glycosides, terpenoids and other bitter substances [148,149]. Terpenes have a bitter taste primarily due to their chemical structure with chelating structures such as lactones, endo-acetals, endo-hydrogen bonds and glycosidic groups [150]. The sesquiterpene coumarins, which are found in relatively large quantities in the genus Ferula, are based on 7-hydroxyumbelliferolactone as the parent nucleus. A number of prior studies have shown that terpenes and volatile oils are found in ample quantities in herbs with the Xing (pungent) taste [151], whereas coumarin sesquiterpenes are similarly abundant in the genus Ferula. In addition, the main chemical components of genus Ferula, such as Farnesiferol A, Farnesiferol B, Farnesiferol C and DAW22, are all coumarin sesquiterpenoids.

5.2.2. Predictive Analysis by the Q-Marker of Traditional Efficacy

The effects of Ferula plants have been recorded in the Chinese Pharmacopoeia 2020 edition and found to be useful as digesting aid, relief of symptoms, dispersal of agglomerates as well as insecticide. They are also used for the treatment of excessive meat consumption, leading to stagnation and masses in the abdomen, and treating bruises, intestinal parasites and the abdominal pain [17]. Their effects are reported in the Xinxiubencao as an insecticide, deodorization, in elimination of intra-abdominal masses, relief of blood stasis, septicemia and pus, and alleviation of symptoms of poisoning [152]. Ferula plants have been also used to treat tuberculosis, eliminate lumps that have accumulated in the abdomen, cure cold, and treat malaria, which are effective against acute gastroenteritis as well as other intestinal diseases. They have a pain-relieving effect on the chest and stomach in addition to pain [153] and similar records have been found in other ethnomedicinal books [154]. Moreover, during the course of modern pharmacological research, the pharmacological effects of Ferula plants are similar to those recorded in ancient books, such as anti-allergy, action on the gastrointestinal tract, anti-bacterial as well as antiseptic effects, anticoagulant effects, and effects related to the cardiovascular system. The CHCl3 extracted fraction of F. sinkiangensis resin can exhibit significant anti-gastric ulcer activity, and the sesquiterpene coumarins contained in this fraction were mainly Farnesiferol B and Farnesiferol C. In addition, prior studies have shown that the raw juices of F. sinkiangensis and F. feruloides have certain effects on gastrointestinal smooth muscle inhibition and anti-experimental gastric ulcer, whereas the chemical compositions of these three types mainly consist of the sesquiterpenes, Farnesiferol A [58], Farnesiferol B, Farnesiferol C. Interestingly, in vivo studies have shown that Farnesiferol B can protect the kidney from I/R-induced damage by reducing oxidative stress and inflammation. In vitro, Farnesiferol B was reported to improve macrophage migration by activating TGR5 [155]. Furthermore, DAW22 was shown in the literature to inhibit five human MPNST cancer cell lines and C6 glioma cells to varying degrees. Overall, ferulic acid possesses numerous pharmacological activities, mainly related to anticancer, anti-bacterial, anti-inflammatory, antioxidant, anti-thrombotic actions, and can also exhibit hypolipidemic effects [156,157].

5.3. Q-Marker Predictive Analysis by Chemical Composition Measurability

The chemical compositions of Chinese medicines are complex, and therefore the chemical measurability of the subject is reported to be an essential element in the Q-marker study [158]. Thus, based on the previous summary study on the specificity and close linkage of the chemical compositions as well as effectiveness of the domestic plants of the genus Ferula, the sesquiterpene coumarins and ferulic acid were further identified as the quality markers of Ferula plants by measuring their chemical compositions. The contents of ferulic acid were determined by HPLC on the different parts of F. sinkiangensis and the roots and leaves of F. fukanensis, and the content of ferulic acid was found to be in the following descending order: gum > root > leaf > stem [159,160], and the content of ferulic acid was also determined by HPLC on the anti-ulcer extract of F. sinkiangensis [161]. In addition, the determination of Farnesiferol A, Farnesiferol C and ferulic acid in the ethyl acetate fraction of F. sinkiangensis was performed by HPLC [162]. The UPLC method was used to estimate the content of DAW22 in F. feruloides. It was observed that the methanolic extract of F. feruloides contained more DAW22, and DAW22 was most abundant in F. feruloides harvested in the first half of May [163].

6. Conclusions

Although the genus Ferula is widespread and diverse throughout the world, its distribution in China is concentrated mainly in Xinjiang and is less diverse. Ferula is native to Central Asia and Middle East, and especially found in eastern Iran and Afghanistan, and is exported to be present worldwide. Nevertheless, Ferula is not native to China, and it has been around for a long time but has become a traditional Chinese herb and ethnic medicine. It plays a vital role in the traditional Chinese medical system.
The document analysis of the China’s Ferula resources revealed that there are several studies describing the chemical compositions and biological activities of Ferula plants. Ferula are mainly composed of coumarins, particularly sesquiterpenes coumarins, volatile oils, sulfur-containing compounds, and aromatic compounds. Interestingly, beneficial pharmacological activity of Ferula is not only traditionally used, but its various components have been reported to possess anticancer, anti-diabetic, antibacterial, anti-flu, anti-inflammatory and other effects.
The documented benefits of Ferula include the elimination of lumps, deworming, antibacterial and other traditional uses. In addition, some traditional uses of Ferula are closely related to the modern research and have clinical relevance. For example, modern phytochemical and pharmacological studies have revealed that DAW22 could target the main components in the MPNST tumorigenic pathways: namely, suppress the phosphorylation of AKT and ERK, and reduce the levels of non-phospho (active) CTNNB1. Moreover, other studies have reported that farnesylate A can facilitate the anticancer drugs to exert substantial cytotoxic effects by inhibiting P-glycoprotein. Another study showed that both Farnesiferol B and Farnesiferol C displayed the best inhibitory effects on P-gp pump efflux and they could be considered as lead scaffolds for further structure modifications. Currently, anticancer activity of F. ferulaeoides is being investigated by the author and his colleagues.
Because of the availability of a wide variety of TCM, their quality standards are not uniform, thereby resulting in the gradual increase in defective medicines, which can adversely the efficacy of TCM. Faced with this challenge, in 2016, Academician Changxiao Liu proposed the concept of the Q-marker. A Q-marker can better identify the various characteristic compounds present in TCM and can be used sequentially as a criterion to evaluate their quality and to better identify their authenticity, thus providing quality assurance for the development of TCM. Additionally, it was found that there were only a few selected studies related to Q-markers of domestic Ferula. Q-marker studies can be of great help to the breeding, conservation and utilization of the domestic Ferula. However, it was observed that the few studies on the Q-markers of domestic Ferula were not conducive to the research development of domestic Ferula spp. in the context of mixed quality.
Moreover, there are only few toxicological studies on the genus Ferula in China. Toxicological studies could be of great help in the clinical analysis of the active components in the domestic Ferula plants, and hence toxicological studies about the chemical composition of domestic Ferula plants should also be strengthened in the future.
Eventually, Ferula plants have a long history of use in China, but Ferula plant resources have suffered great damage due to human activities in recent years, resulting in acute shortage of plant resources and protective measures are urgently needed. However, regarding the rich background of biological activities of Ferula, it appears that there are still a large number of unaccomplished investigations that need to be conducted in the future. Examples include research on the biological characteristics and genetic variation mechanisms of the genus Ferula, for better germplasm conservation and utilization; research on the medicinal value and pharmacological mechanisms of action of the genus Ferula, to explore its application prospects and value in the field of medicine; research on the cultivation and production techniques of Ferula spp. to improve its yield and quality and to meet market demand; analysis and research on the distribution status of the resources and ecological impact of the genus Abies, and the development of conservation strategies to effectively protect and manage these precious plant resources.

Author Contributions

Y.B. drafted the manuscript. X.Y. revised the manuscript. X.Y., H.T., K.P., M.K., F.S. and M.M. contributed to the conception and design of this article and interpreting the relevant literature. All authors have read and agreed to the published version of the manuscript.

Funding

The work was financially supported by the Fundamental Research Funds for the Central Universities, South-Central Minzu University (Grant Number CZZ22002), and the Regional collaborative Innovation Project of Xinjiang Uyghur Autonomous Region—The Science and Technology Partnership Program and International Science and Technology Cooperation Program of Shanghai Cooperation Organization (2022276816).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Salehi, M.; Naghavi, M.R.; Bahmankar, M. A review of Ferula species: Biochemical characteristics, pharmaceutical and industrial applications, and suggestions for biotechnologists. Ind. Crops Prod. 2019, 139, 111511. [Google Scholar] [CrossRef]
  2. Znati, M.; Hichem, B.J.; Cazaux, S.; Bouajila, J. Chemical composition, biological and cytotoxic activities of plant extracts and compounds isolated from Ferula lutea. Molecules 2014, 19, 2733–2747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Dan, R.H.; She, M.L. Flora of China; Science Press: Beijing, China, 1992; Volume 55, pp. 85–117. [Google Scholar]
  4. He, S.; Tan, G.Y. Advances in Studies of Ferula L. J. Xinjiang Agric. Univ. 2002, 25, 1–7. [Google Scholar]
  5. Eigner, D.; Scholz, D. The magic book of Gyani Dolma. Pharm. Unserer. Zeit. 1990, 19, 141–152. [Google Scholar] [CrossRef] [PubMed]
  6. Bandyopadhyay, D.; Basak, B.; Chatterjee, A.; Lai, T.K.; Banerji, A.; Banerji, J.; Neuman, A.; Prange, T. Saradaferin, a new sesquiterpenoid coumarin from Ferula assafoetida. Nat. Prod. Res. 2006, 20, 961–965. [Google Scholar] [CrossRef]
  7. Akaberi, M.; Iranshahy, M.; Iranshahi, M. Review of the traditional uses, phytochemistry, pharmacology and toxicology of giant fennel (Ferula communis L. subsp. communis). Iran. J. Basic Med. Sci. 2015, 18, 1050. [Google Scholar]
  8. Mahendra, P.; Bisht, S. Ferula asafoetida: Traditional uses and pharmacological activity. Pharmacogn. Rev. 2012, 6, 141. [Google Scholar] [CrossRef] [Green Version]
  9. Fan, C.; Li, X.; Zhu, J.; Song, J.; Yao, H. Endangered Uyghur Medicinal Plant Ferula Identification through the Second Internal Transcribed Spacer. Evid. Based Complement. Altern. Med. 2015, 2015, 479879. [Google Scholar] [CrossRef] [Green Version]
  10. Xinjiang Food and Drug Administration. The Xinjiang Uygur Autonomous Region Traditional Chinese Medicine and Uygur Medicine Pieces Concoct Standard; Xinjiang People’s Saitary Press: Urumqi, China, 2010. [Google Scholar]
  11. Chinese Pharmacopoeia Commission. Chinese Pharmacopoeia; Chinese Medicine Science and Technology Press: Beijing, China, 2020; Volume 1, p. 198. [Google Scholar]
  12. Iranshahy, M.; Iranshahi, M. Traditional uses, phytochemistry and pharmacology of asafoetida (Ferula assa-foetida oleo-gum-resin)—A review. J. Ethnopharmacol. 2011, 134, 1–10. [Google Scholar] [CrossRef]
  13. Amalraj, A.; Gopi, S. Biological activities and medicinal properties of Asafoetida: A review. J. Tradit. Complement. Med. 2017, 7, 347–359. [Google Scholar] [CrossRef]
  14. Yatham, P.; Shukla, D.; Srivastava, A.K.; Pragadheesh, V.S.; Kumar, D. Purification and identification of anticancer organosulfides from Ferula assa-foetida gum: Integrative analysis employing GC/GC-MS/RP-HPLC/NMR. Nat. Prod. Res. 2022, 36, 2869–2874. [Google Scholar] [CrossRef] [PubMed]
  15. Saleem, M.; Alam, A.; Sultana, S. Asafoetida inhibits early events of carcinogenesis: A chemopreventive study. Life Sci. 2001, 68, 1913–1921. [Google Scholar] [CrossRef] [PubMed]
  16. Divya, K.; Ramalakshmi, K.; Murthy, P.S.; Jagan Mohan Rao, L. Volatile oils from Ferula asafoetida varieties and their antimicrobial activity. LWT Food Sci. Technol. 2014, 59, 774–779. [Google Scholar] [CrossRef]
  17. Javanshir, S.; Soukhtanloo, M.; Jalili-Nik, M.; Yazdi, A.J.; Amiri, M.S.; Ghorbani, A. Evaluation Potential Antidiabetic Effects of Ferula latisecta in Streptozotocin-Induced Diabetic Rats. J. Pharmacopunct. 2020, 23, 158–164. [Google Scholar] [CrossRef] [PubMed]
  18. Lee, C.L.; Chiang, L.C.; Cheng, L.H.; Chuang, L.C.; Mohamed, H.A.E.-R.; Chang, F.R.; Wu, Y.C. Influenza A (H1N1) Antiviral and Cytotoxic Agents from Ferula assa-foetida. J. Nat. Prod. 2009, 72, 1568–1572. [Google Scholar] [CrossRef]
  19. Ghasemi, Z.; Rezaee, R.; Aslani, M.R.; Boskabady, M.H. Anti-inflammatory, anti-oxidant, and immunomodulatory activities of the genus Ferula and their constituents: A review. Iran J. Basic Med. Sci. 2021, 24, 1613–1623. [Google Scholar]
  20. Mobasheri, L.; Khorashadizadeh, M.; Safarpour, H.; Mohammadi, M.; Anani Sarab, G.; Askari, V.R. Anti-Inflammatory Activity of Ferula assafoetida Oleo-Gum-Resin (Asafoetida) against TNF-alpha-Stimulated Human Umbilical Vein Endothelial Cells (HUVECs). Mediat. Inflamm. 2022, 2022, 5171525. [Google Scholar] [CrossRef]
  21. Safaeian, L.; Ghannadi, A.; Javanmard, S.H.; Vahidian, M.H. The effect of hydroalcoholic extract of Ferula foetida stems on blood pressure and oxidative stress in dexamethasone-induced hypertensive rats. Res. Pharm. Sci. 2014, 10, 326–334. [Google Scholar]
  22. Ellman, G.L.; Courtney, K.D.; Andres, V., Jr.; Featherstone, R.M. A New and Rapid Colorimetric Determination of Acetylcholinesterase Activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  23. Karimi, G.; Iranshahi, M.; Hosseinalizadeh, F.; Riahi, B.; Sahebkar, A. Screening of acetylcholinesterase inhibitory activity of terpenoid and coumarin derivatives from the genus Ferula. Pharmacol. 2010, 1, 566–574. [Google Scholar]
  24. Hashemzaei, M.; SadeghiBonjar, M.A.; Tabrizian, K.; Iranshahi, M.; Iranshahy, M.; Rezaee, R. Evaluation of the analgesic effect of Umbelliprenin and Umbelliprenin-morphine co-administration on the acute, chronic and neuropathic pain. Indian J. Pharm. Educ. Res. 2015, 49, 121–125. [Google Scholar] [CrossRef] [Green Version]
  25. Dastan, D.; Salehi, P.; Aliahmadi, A.; Gohari, A.R.; Maroofi, H.; Ardalan, A. New coumarin derivatives from Ferula pseudalliacea with antibacterial activity. Nat. Prod. Res. 2016, 30, 2747–2753. [Google Scholar] [CrossRef]
  26. Nazari, Z.E.; Iranshahi, M. Biologically active sesquiterpene coumarins from Ferula species. Phytother. Res. 2011, 25, 315–323. [Google Scholar] [CrossRef]
  27. Pavela, R.; Morshedloo, M.R.; Lupidi, G.; Carolla, G.; Barboni, L.; Quassinti, L.; Bramucci, M.; Vitali, L.A.; Petrelli, D.; Kavallieratos, N.G.; et al. The volatile oils from the oleo-gum-resins of Ferula assa-foetida and Ferula gummosa: A comprehensive investigation of their insecticidal activity and eco-toxicological effects. Food Chem. Toxicol. 2020, 140, 111312. [Google Scholar] [CrossRef]
  28. Soltani, S.; Amin, G.R.; Salehi-Sourmaghi, M.H.; Schneider, B.; Lorenz, S.; Iranshahi, M. Sulfur-containing compounds from the roots of Ferula latisecta and their cytotoxic activities. Fitoterapia 2018, 124, 108–112. [Google Scholar] [CrossRef]
  29. Rasulev, B.F.; Saidkhodzhaev, A.I.; Nazrullaev, S.S.; Akhmedkhodzhaeva, K.S.; Khushbaktova, Z.A.; Leszczynski, J. Molecular modelling and QSAR analysis of the estrogenic activity of terpenoids isolated from Ferula plants. SAR QSAR Environ. Res. 2007, 18, 663–673. [Google Scholar] [CrossRef] [PubMed]
  30. Guo, D.A.; Lu, A.; Liu, L. Modernization of traditional Chinese medicine. J. Ethnopharmacol. 2012, 141, 547–548. [Google Scholar] [CrossRef] [Green Version]
  31. Guo, D.A.; Wu, Y.Y.; Ye, M.; Liu, X.; Cordell, G.A. A holistic approach to the quality control of traditional Chinese medicines. Science 2015, 347, 29–31. [Google Scholar]
  32. Liu, C.-X.; Cheng, Y.-Y.; Guo, D.-A.; Zhang, T.-J.; Li, Y.-Z.; Hou, W.-B.; Huang, L.-Q.; Xu, H.-Y. A New Concept on Quality Marker for Quality Assessment and Process Control of Chinese Medicines. Chin. Herb. Med. 2017, 9, 3–13. [Google Scholar] [CrossRef]
  33. Yang, W.; Zhang, Y.; Wu, W.; Huang, L.; Guo, D.; Liu, C. Approaches to establish Q-markers for the quality standards of traditional Chinese medicines. Acta Pharm. Sin. B 2017, 7, 439–446. [Google Scholar] [CrossRef] [PubMed]
  34. State Key Laboratory of Systematic and Evolutionary Botany, I.o.B. The Chinese Academy of Sciences Angiospermae >> Apiaceae >> Ferula >> Ferula sinkiangensis. Available online: http://ppbc.iplant.cn/tu/6312937 (accessed on 15 June 2023).
  35. State Key Laboratory of Systematic and Evolutionary Botany, I.o.B. The Chinese Academy of Sciences Angiospermae >> Apiaceae >> Ferula >> Ferula fukanensis. Available online: http://ppbc.iplant.cn/tu/4351033 (accessed on 15 June 2023).
  36. State Key Laboratory of Systematic and Evolutionary Botany, I.o.B. The Chinese Academy of Sciences Angiospermae >> Apiaceae >> Ferula >> Ferula kingdon-wardii. Available online: http://ppbc.iplant.cn/tu/8596963 (accessed on 15 June 2023).
  37. State Key Laboratory of Systematic and Evolutionary Botany, I.o.B. The Chinese Academy of Sciences Angiospermae >> Apiaceae >> Ferula >> Ferula songarica. Available online: http://ppbc.iplant.cn/tu/6473066 (accessed on 15 June 2023).
  38. State Key Laboratory of Systematic and Evolutionary Botany, I.o.B. The Chinese Academy of Sciences Angiospermae >> Apiaceae >> Ferula >> Ferula syreitschikowii. Available online: http://ppbc.iplant.cn/tu/6509768 (accessed on 15 June 2023).
  39. State Key Laboratory of Systematic and Evolutionary Botany, I.o.B. The Chinese Academy of Sciences Angiospermae >> Apiaceae >> Ferula >> Ferula licentiana. Available online: http://ppbc.iplant.cn/tu/4286927 (accessed on 15 June 2023).
  40. Li, Y.D.; Fu, S.Y.; He, J.; Fan, C.Z.; Li, X.J. Analysis of Specific Medicinal Plants Resources Ferula sinkiangensis in Xinjiang. Mod. Chin. Med. 2016, 18, 714–717. [Google Scholar]
  41. Yan, X.H.; Zhou, B.; Liu, H.B.; Xu, H.Y.; Yang, W.X.; Jiao, X.G.; Tian, S.G. Investigation and protection for endangerd Ferula fukanensis K.M. Shen. J. Xinjiang Med. Univ. 2012, 35, 1155–1158. [Google Scholar]
  42. Xinjiang Institute of Biological Soil and Desert Research. Flora of Xinjiang Medical Plants; Xinjiang People’s Publishing House: Urumqi, China, 1977; Volume 1. [Google Scholar]
  43. Tan, Y.; Gao, T.T.; Ma, Y.; Ding, X.; Chen, Y.H. Pharmacognostic Identification of Ferula syreitschikowii. J. Chin. Med. Mater. 2011, 34, 1694–1696. [Google Scholar]
  44. Editorial Committee of the Chinese Medical Encyclopedia. Uyghur Medicine; Shanghai Scientific & Technical Publishers: Shanghai, China, 2005. [Google Scholar]
  45. Zhou, R.H. Resource of Chinese Medicinal Materials; China Medico-Pharmaceutical Science & Technology Publishing House: Beijing, China, 1993; Volume 05. [Google Scholar]
  46. Editorial Board of “Primary colour mapping of weeds in Chinese farmland”. Primary Colour Mapping of Weeds in Chinese Farmland; China Agriculture Press: Beijing, China, 1990. [Google Scholar]
  47. Ding, X.; Tan, Y.; Liu, J.S.; Zhao, W.B. Pharmacognostical Study on Ferula dissecta (Ledeb.) Ledeb. Lishizhen Med. Mater. Med. 2011, 22, 1555–1556. [Google Scholar]
  48. Science and Technology Commission of Tibet Autonomous Region. Tibetan Plant List; Science and Technology Commission of Tibet Autonomous Region: Lhasa, China, 1980. [Google Scholar]
  49. Jiang, J.W. Quick-Consultative Dicitionary of World Medicinal Plants; China Medico-Pharmaceutical Science & Technology Publishing House: Beijing, China, 2015. [Google Scholar]
  50. Xinjiang Bayi Agricultural College. Search List of Plants of Xinjiang; Xinjiang People’s Publishing House: Urumqi, China, 1983. [Google Scholar]
  51. Shangguan, T.L.; Ma, Z.Q.; Xie, S.L. Rare and Endangered Plants in Shanxi Province; China Science and Technology Press: Beijing, China, 1998. [Google Scholar]
  52. He, J.Q. Flora of Northern Anhui Resources; China Agriculture Press: Beijing, China, 2001. [Google Scholar]
  53. Liu, N.F. Scientific Expedition to Dunhuang Nature Reserve, Gansu; Chinese Forestry Publishing House: Beijing, China, 2001. [Google Scholar]
  54. Southwest Forestry College; Yunnan Forestry Department; Zhongdian County Forestry Bureau. Report on a Comprehensive Scientific Study of the Bitahai Nature Reserve in Yunnan; Southwest Forestry College, Yunnan Forestry Department, Zhongdian County Forestry Bureau: Kunming, China, 2002; Volume 2. [Google Scholar]
  55. Executive Committee of the First Annual Youth Academic Conference of Jiangsu Province. In Proceedings of the First Annual Youth Academic Conference of Jiangsu Province (Science Fascicle), Nanjing, China, 17–19 November 1992; China Science and Technology Press: Beijing, China, 1992.
  56. Lin, J.R.; Jin, M.; Wu, C.M. Advances in studies on chemical constituents and pharmacological effects of Ferula. Strait Pharm. J. 2014, 26, 1–3. [Google Scholar]
  57. El-Razek, M.A.; Ohta, S.; Hirata, T. Terpenoid Coumarins of the Genus Ferula. Heterocycles 2003, 34, 68971. [Google Scholar] [CrossRef]
  58. Xing, Y.; Li, N.; Zhou, D.; Chen, G.; Jiao, K.; Wang, W.; Si, Y.; Hou, Y. Sesquiterpene Coumarins from Ferula sinkiangensis Act as Neuroinflammation Inhibitors. Planta Med. 2017, 83, 135–142. [Google Scholar] [CrossRef]
  59. Wang, J.; Wang, H.; Zhang, M.; Li, X.; Zhao, Y.; Chen, G.; Si, J.; Jiang, L. Sesquiterpene coumarins from Ferula sinkiangensis KM Shen and their cytotoxic activities. Phytochemistry 2020, 180, 112531. [Google Scholar] [CrossRef] [PubMed]
  60. Li, X.Y.; Li, G.Y.; Wang, H.Y.; Wang, Y.; Wang, J.H. Chemical Constituents from Ferula Lehmannii Boiss. Mod. Chin. Med. 2010, 12, 17–20. [Google Scholar]
  61. Yang, J.R.; An, Z.; Li, Z.H.; Jing, S.; Qina, H.L. Sesquiterpene coumarins from the roots of Ferula sinkiangensis and Ferula teterrima. Chem. Pharm. Bull. 2006, 54, 1595–1598. [Google Scholar] [CrossRef] [Green Version]
  62. Wang, Y.E.; Si, J.Y.; Li, X.J.; Jiang, L.; Zhu, J. Chemical Constituents from the Seeds of Ferula sinkiangensis. Mod. Chin. Med. 2011, 13, 26–28. [Google Scholar]
  63. Li, G.; Li, X.; Cao, L.; Zhang, L.; Shen, L.; Zhu, J.; Wang, J.; Si, J. Sesquiterpene coumarins from seeds of Ferula sinkiangensis. Fitoterapia 2015, 103, 222–226. [Google Scholar] [CrossRef]
  64. Meng, H.; Li, G.; Huang, J.; Zhang, K.; Wang, H.; Wang, J. Sesquiterpene coumarin and sesquiterpene chromone derivatives from Ferula ferulaeoides (Steud.) Korov. Fitoterapia 2013, 86, 70–77. [Google Scholar] [CrossRef]
  65. Motai, T.; Kitanaka, S. Sesquiterpene coumarins from Ferula fukanensis and nitric oxide production inhibitory effects. Chem. Pharm. Bull. (Tokyo) 2004, 52, 1215–1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Motai, T.; Daikonya, A.; Kitanaka, S. Sesquiterpene coumarins from Ferula fukanensis and their pro-inflammatory cytokine gene expression inhibitory effects. Chem. Pharm. Bull. 2013, 61, 618–623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Meng, H.; Li, G.; Huang, J.; Zhang, K.; Wei, X.; Ma, Y.; Zhang, C.; Wang, J. Sesquiterpenoid derivatives from Ferula ferulaeoides (Steud.) Korov. Phytochemistry 2013, 86, 151–158. [Google Scholar] [CrossRef] [PubMed]
  68. Kahraman, C.; Topcu, G.; Bedir, E.; Tatli, I.I.; Ekizoglu, M.; Akdemir, Z.S. Phytochemical screening and evaluation of the antimicrobial and antioxidant activities of Ferula caspica M. Bieb. extracts. Saudi Pharm. J. 2019, 27, 525–531. [Google Scholar] [CrossRef]
  69. Motai, T.; Kitanaka, S. Sesquiterpene Chromones from Ferula fukanensis and Their Nitric Oxide Production Inhibitory Effects. J. Nat. Prod. 2005, 68, 1732–1735. [Google Scholar] [CrossRef]
  70. Motai, T.; Kitanaka, S. Sesquiterpene Phenylpropanoids from Ferula fukanensis and Their Nitric Oxide Production Inhibitory Effects. J. Nat. Prod. 2005, 68, 365–368. [Google Scholar] [CrossRef]
  71. Garg, S.N.; Agarwal, S.K. New Sesquiterpenes from Ferula jaeschkeana. Planta Med. 1986, 53, 341–342. [Google Scholar] [CrossRef]
  72. Liu, Q.X.; Hui, H. The chemical constituents of volatile oil from Ferula L. in China and its taxonomical significance. J. Plant Resour. Environ. 1997, 6, 27–32. [Google Scholar]
  73. Lei, L.J.; Teng, L.; Zhao, X.; Yu, F.S.; Wang, C.H. Extraction and assay of volatile oil from Ferula ferlaeoidis (Sted.) Korov. Chin. Tradit. Pat. Med. 2013, 35, 1251–1256. [Google Scholar]
  74. Deng, W.P.; Xie, C.X.; Fu, J.H. Analysis of Volatile Oil from Ferula sinkiangensis by GC/MS. J. Chin. Mass Spectrom. Soc. 2007, 28, 114–116+121. [Google Scholar]
  75. Yang, M.H.; Luo, J.Y.; Qiao, M.L.; Yang, M.H.; Sheng, P. GC-MS Analysis of Volatile Oil from Ferula ferulaeoides and Anti-gastric Cancer Activity of D-Limonene in vitro. Chin. J. Mod. Appl. Pharm. 2020, 37, 806–813. [Google Scholar]
  76. Li, X.; Wang, Y.; Zhu, J.; Xiao, Q. Essential oil composition analysis of three cultivars seeds of Resina Ferulae from Xinjiang, China. Pharm. Mag. 2011, 7, 116–120. [Google Scholar] [CrossRef] [Green Version]
  77. Zhi-da, M.; Qi-fi, M.; Mizuno, M.; Tanaka, T.; Munekazu, I. Polysulfanes in the volatile oils of Ferula species. Planta Med. 1986, 53, 300–302. [Google Scholar] [CrossRef]
  78. Xu, Q.M. Studies of the Chemical Constituents of Varia kweichowensis and Ferula fukanensis KM Shen. Ph.D. Thesis, Peking Union Medical College, Tsinghua University, Beijing, China, 2005. [Google Scholar]
  79. Zhu, G.X.; Zhang, H.Q. A New Compound from the Root of Ferula tunshanica Su. J. China Pharm. Univ. 1998, 29, 19–20. [Google Scholar]
  80. Hu, Y.; Li, X.D.; Li, G.Y.; Li, X.Y.; Zhang, K.; Zuo, W.J.; Zeng, Y.N.; Wang, J.H. Studies on the Chemical Constituents of Ferula songorica. Mod. Chin. Med. 2009, 11, 18–19. [Google Scholar]
  81. Zhu, G.X.; Zhang, H.Q. Studies on the Chemical Constituents in the Root of Ferula tunshanica Su. J. China Pharm. Univ. 1996, 27, 585–588. [Google Scholar]
  82. Liu, T. Chemical Constituents from Two Plants of Apiaceae and Their Biological Avtivities. Ph.D. Thesis, Fudan University, Shanghai, China, 2013. [Google Scholar]
  83. Liu, J. Study on Chemical Constituents and Quality Control Method of Ferula ferulaeoides. Master’s Thesis, Inner Mongolia Medical University, Hohhot, China, 2019. [Google Scholar]
  84. Zhao, W.B.; Tan, Y.; Xiang, Y.; Zhu, Y. Determination of Amino Acids and Mineral Elements Content in Ferulaeoides korov Boiss. Res. Pract. Chin. Med. 2009, 23, 67–68. [Google Scholar]
  85. Deng, W.P. Extract and Study of the Chemical Constituents of Ferula Sinkaingensis. Master’s Thesis, Xinjiang University, Urumqi, China, 2008. [Google Scholar]
  86. Han, H.Y.; Li, G.Y.; Wang, H.Y.; Ma, Q.D.; Gao, J.B.; Wang, J.H. Anticoagulantactivity of radiatinol and scopoletin from Ferula dissecta (Ledeb.) Ledeb. in vitro. J. Nongken Med. 2010, 32, 112–114. [Google Scholar]
  87. Furlan, V.; Bren, U. Helichrysum italicum: From Extraction, Distillation, and Encapsulation Techniques to Beneficial Health Effects. Foods 2023, 12, 802. [Google Scholar] [CrossRef]
  88. Iranshahi, M.; Kalategi, F.; Rezaee, R.; Shahverdi, A.R.; Ito, C.; Furukawa, H.; Tokuda, H.; Itoigawa, M. Cancer chemopreventive activity of terpenoid coumarins from Ferula species. Planta Med. 2008, 74, 147–150. [Google Scholar] [CrossRef] [PubMed]
  89. Monti, M.; Pinotti, M.; Appendino, G.; Dallocchio, F.; Bellini, T.; Antognoni, F.; Poli, F.; Bernardi, F. Characterization of anti-coagulant properties of prenylated coumarin ferulenol. Biochim. Biophys. Acta 2007, 1770, 1437–1440. [Google Scholar] [CrossRef] [PubMed]
  90. Ozkan, H.; Yanmis, D.; Karadayi, M.; Bal, T.; Baris, O.; Gulluce, M. Determination of genotoxic and antigenotoxic properties of essential oil from Ferula orientalis L. using Ames/Salmonella and E. coli WP2 bacterial test systems. Toxicol. Ind. Health 2014, 30, 714–723. [Google Scholar] [CrossRef]
  91. Illuri, R.; Venkataramana, S.H.; Daguet, D.; Kodimule, S. Sub-acute and acute toxicity of Ferula asafoetida and Silybum marianum formulation and effect of the formulation on delaying gastric emptying. BMC Complement. Altern. Med. 2019, 19, 159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Maiuolo, J.; Bava, I.; Carresi, C.; Gliozzi, M.; Musolino, V.; Scicchitano, M.; Macri, R.; Oppedisano, F.; Scarano, F.; Caterina Zito, M.; et al. The Effect of Ferula communis Extract in Escherichia coli Lipopolysaccharide-Induced Neuroinflammation in Cultured Neurons and Oligodendrocytes. Int. J. Mol. Sci. 2021, 22, 7910. [Google Scholar] [CrossRef]
  93. Gholitabar, S.; Roshan, V.D. Effect of treadmill exercise and Ferula gummosa on myocardial HSP72, vascular function, and antioxidant defenses in spontaneously hypertensive rats. Clin. Exp. Hypertens. 2013, 35, 347–354. [Google Scholar] [CrossRef]
  94. Esmaeili, H.; Hafezimoghadam, Z.; Esmailidehaj, M.; Rezvani, M.E.; Hafizibarjin, Z. The effect of asafoetida essential oil on myocardial ischemic-reperfusion injury in isolated rat hearts. Avicenna J. Phytomed. 2017, 8, 338–349. [Google Scholar]
  95. Zhou, Y.; Xin, F.; Zhang, G.; Qu, H.; Yang, D.; Han, X. Recent Advances on Bioactive Constituents in Ferula. Drug Dev. Res. 2017, 78, 321–331. [Google Scholar] [CrossRef]
  96. Xing, Y.C.; Li, N.; Xue, J. Progress on chemical constituents of Ferula genus. J. Shenyang Pharm. Univ. 2012, 29, 730–741. [Google Scholar]
  97. Guo, T.T.; Zhou, Y.P.; Dang, W.; Meng, C.R.; Zhou, D.; Chen, G.; Li, N. “Preexistence” and “present life” of traditional Chinese medicine Ferulae Resina. Chin. Tradit. Herb. Drugs 2021, 52, 5401–5413. [Google Scholar]
  98. Ying, F.H.; Qian, X.Q.; Liu, B.R. Progress in the study of the anti-tumor mechanism of action of ferulic acid. Mod. J. Integr. Tradit. Chin. West. Med. 2010, 19, 4238–4240. [Google Scholar]
  99. Suzuki, K.; Okasaka, M.; Kashiwada, Y.; Takaishi, Y.; Honda, G.; Ito, M.; Takeda, Y.; Kodzhimatov, O.K.; Ashurmetov, O.; Sekiya, M.; et al. Sesquiterpene Lactones from the Roots of Ferula Waria and Their Cytotoxic Activity. J. Nat. Prod. 2007, 70, 1915–1918. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, S.; Sheng, P.; Yao, L.; Du, B.J. GC-MS fingerprint of in vitro anti-gastric cancer active parts from roots of Uygur medicine Ferula ferulaeoides. Chin. Tradit. Herb. Drugs 2015, 46, 2874–2879. [Google Scholar]
  101. Wu, J.; Wang, Y.X.; Wei, N.; Wu, X.S.; Liu, X. Ferulic acid inhibits lung cancer growth and its mechanism. Zhejiang Med. J. 2018, 40, 1303–1306+1311+1414. [Google Scholar]
  102. Yang, M.H.; Luo, J.Y.; Qiao, M.L.; Sheng, P.; Yang, M.H. Mechanism of Uygur Medicine Root of Ferula ferulaeoides in Resisting Gastric Cancer Activity and Inducing Cell Apoptosis and Cell Cycle Arrest in vitro. Chin. J. Exp. Tradit. Med. Formulae 2018, 24, 112–122. [Google Scholar]
  103. Zhang, L.; Tong, X.; Zhang, J.; Huang, J.; Wang, J. DAW22, a natural sesquiterpene coumarin isolated from Ferula ferulaeoides (Steud.) Korov. that induces C6 glioma cell apoptosis and endoplasmic reticulum (ER) stress. Fitoterapia 2015, 103, 46–54. [Google Scholar] [CrossRef]
  104. Li, X.X.; Zhang, S.J.; Chiu, A.P.; Lo, L.H.; Huang, J.; Rowlands, D.K.; Wang, J.; Keng, V.W. Targeting of AKT/ERK/CTNNB1 by DAW22 as a potential therapeutic compound for malignant peripheral nerve sheath tumor. Cancer Med. 2018, 7, 4791–4800. [Google Scholar] [CrossRef] [Green Version]
  105. Zhang, C.; Jiang, Y.; Zhang, J.; Huang, J.; Wang, J. 8-p-Hdroxybenzoyl Tovarol Induces Paraptosis Like Cell Death and Protective Autophagy in Human Cervical Cancer HeLa Cells. Int. J. Mol. Sci. 2015, 16, 14979–14996. [Google Scholar] [CrossRef]
  106. Kores, K.; Kolenc, Z.; Furlan, V.; Bren, U. Inverse Molecular Docking Elucidating the Anticarcinogenic Potential of the Hop Natural Product Xanthohumol and Its Metabolites. Foods 2022, 11, 1253. [Google Scholar] [CrossRef] [PubMed]
  107. Gao, T.T.; Yu, F.H.; Tan, Y.; Liu, W.X. Study on Antimicrobial Activity in vitro of Extracts from Three Species of Ferula Root. North. Hortic. 2013, 24, 156–158. [Google Scholar]
  108. Liu, T.; Wang, S.; Fu, W.; Gibbons, S.; Mu, Q. Sesquiterpenoids with Anti-MDR Staphylococcus aureus Activities from Ferula ferulioides. Chem. Biodivers. 2015, 12, 599–614. [Google Scholar] [CrossRef] [PubMed]
  109. Liu, T.; Osman, K.; Kaatz, G.W.; Gibbons, S.; Mu, Q. Antibacterial Sesquiterpenoid Derivatives from Ferula ferulaeoides. Planta Med. 2013, 79, 701–706. [Google Scholar] [CrossRef]
  110. Nazemisalman, B.; Vahabi, S.; Yazdinejad, A.; Haghghi, F.; Jam, M.S.; Heydari, F. Comparison of antimicrobial effect of Ziziphora tenuior, Dracocephalum moldavica, Ferula gummosa, and Prangos ferulacea essential oil with chlorhexidine on Enterococcus faecalis: An in vitro study. Dent. Res. J. 2018, 15, 111–116. [Google Scholar]
  111. Sonigra, P.; Meena, M. Metabolic Profile, Bioactivities, and Variations in the Chemical Constituents of Essential Oils of the Ferula Genus (Apiaceae). Front. Pharm. 2020, 11, 608649. [Google Scholar] [CrossRef]
  112. Zhang, H.Q.; Hu, J. Anti-allergic pharmacological action of Ferula sinkiangensis. China J. Chin. Mater. Med. 1986, 08, 49–52. [Google Scholar]
  113. Hu, H.J.; Hu, J. Effect of ferulic acid on metabolic reactions. J. China Pharm. Univ. 1991, 12, 49–52. [Google Scholar]
  114. Ye, E.B.; Liu, F.; Xiong, Y.J.; Cheng, G.; Yang, X.Z.; Gzo, Y. Anti-inflammatory and immunosuppressive effects of three types of Ferula from Xinjiang. Northwest Pharm. J. 1993, 8, 72–75. [Google Scholar]
  115. Li, X.Y.; Li, G.Y.; Wang, H.Y.; Lou, M.M.; Han, H.Y.; Zhang, K.; Ma, Q.D.; Wang, J.H. Study on Anticoagulation Effects of Ferula lehmannii Boiss. China Pharm. 2010, 13, 1626–1628. [Google Scholar]
  116. Cai, S.S.; Deng, S.Z.; Liu, L.; Sun, D.M. Effects of Ferulic Acid on Ventricular Collagen Remodeling in Mice with Chronic Heart Failure. Acta Med. Univ. Sci. Technol. Huazhong 2012, 41, 684–687. [Google Scholar]
  117. Jiangsu New Medical College. Dictionary of Chinese Medicines (Upper Volume); Shanghai Scientific & Technical Publishers: Shanghai, China, 1977. [Google Scholar]
  118. Pategule, Y.; Aierxili, T.; Gulishan, M.; Silapu, A. Clinical application and research progress of Uyghur medicine with the Uyghur drug Ferula in Uyghur medicine. J. Med. Pharm. Chin. Minor. 2011, 17, 61–64. [Google Scholar]
  119. Li, X.J.; Jiang, L.; Da, P. Preparation and Investigation of the Phamacodynamics of Effective Antiulcerative Composition in Ferula sinkiangensis KM Shen. Mod. Chin. Med. 2007, 9, 8–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Guo, T.T.; Lu, J.; Jiang, L.; Kang, X.L. The effect of Ferula sinkiangensis K.M. Shen and its processed products on gastric ulcer model in rats. J. Xinjiang Med. Univ. 2013, 36, 1463–1466. [Google Scholar]
  121. Xiong, Y.J.; Liu, F.; Ye, E.B.; Hu, J.; Yng, X.Z. Comparison of the gastrointestinal effects of three types of Xinjiang Ferula. J. Xinjiang Med. Univ. 1993, 16, 300–302. [Google Scholar]
  122. Teng, L.; Ma, G.; Li, L.; Ma, L.; Xu, X. Karatavicinol a, a new anti-ulcer sesquiterpene coumarin from Ferula sinkiangensis. Chem. Nat. Compd. 2013, 49, 606–609. [Google Scholar] [CrossRef]
  123. Xu, M.; Zhang, L.; Li, P.; Wang, C.; Shi, Y. Network pharmacology used to decode potential active ingredients in Ferula assafoetida and mechanisms for the application to Alzheimer’s disease. J. Tradit. Chin. Med. Sci. 2020, 7, 199–209. [Google Scholar] [CrossRef]
  124. Mi, Y.; Jiao, K.; Xu, J.K.; Wei, K.; Liu, J.Y.; Meng, Q.Q.; Guo, T.T.; Zhang, X.N.; Zhou, D.; Qing, D.G.; et al. Kellerin from Ferula sinkiangensis exerts neuroprotective effects after focal cerebral ischemia in rats by inhibiting microglia-mediated inflammatory responses. J. Ethnopharmacol. 2021, 269, 113718. [Google Scholar] [CrossRef]
  125. Zhang, W.; Mi, Y.; Jiao, K.; Xu, J.; Guo, T.; Zhou, D.; Zhang, X.; Ni, H.; Sun, Y.; Wei, K.; et al. Kellerin alleviates cognitive impairment in mice after ischemic stroke by multiple mechanisms. Phytother. Res. 2020, 34, 2258–2274. [Google Scholar] [CrossRef]
  126. Yang, Z.M.; Wang, Y.T.; Wu, C.Q.; Li, C.G.; Zhang, H.Y.; Xing, B. A short summary of animal experimental observations on the anti-fertility of the Chinese medicine Ferula. Jilin J. Chin. Med. 1982, 04, 42–43. [Google Scholar]
  127. Zhang, S.; Zhang, Z.Y.; Zhang, Y.; He, W. Termination of pregnancy by Ferula. Chin. Tradit. Herb. Drugs 1991, 22, 458–459. [Google Scholar]
  128. Wang, D.M.; Wang, D.B. The effect of Ferula on the isolated uterus of mice and rabbits and its relationship with estrogen and progesterone levels in vivo. Chin. Tradit. Herb. Drugs 1986, 17, 28–30. [Google Scholar]
  129. Zhong, Z.L.; Zhang, F.; Jiang, Y.H.; Zhang, W.T.; Tong, J.C.; Li, J.; Chu, J.R.; Xie, H.T. Protective effects of Ferulic acid on acute liver injury induced by D-GalN /LPS in mice. Chin. J. Clin. Pharmacol. Ther. 2018, 23, 1335–1339. [Google Scholar]
  130. Zhang, H.Y.; Shi, H.; Jiang, L.; Zhou, L.L.; Zhou, M.X. Study on Screening of Active Parts of Xinjiang Ferulae Resina for Regulating Blood Lipid. Chin. J. Inf. Tradit. Chin. Med. 2015, 22, 43–46. [Google Scholar]
  131. Ping, Z.; Yoshihisa, T.; Hongquan, D.; Bei, C.; Gisho, H.; Michiho, I.; Yoshio, T.; Olimjon, K.K.; Kuo-Hsing, L. Coumarins and bicoumarin from Ferula sumbul: Anti-HIV activity and inhibition of cytokine release. Phytochemistry 2000, 53, 689–697. [Google Scholar]
  132. Luo, X.; Ma, G.Z.; Dan, M.; Wang, J.; Yu, F.S.; Teng, L.; Wang, C.H. A comparative study of the acute toxicity of two volatile oils of Ferula alba and their chemical composition. Chin. Tradit. Pat. Med. 2014, 37, 1130–1135. [Google Scholar]
  133. Li, J.; Xu, H.Y.; Jia, X.G.; Tian, S.G. Progress in the study of the chemical composition and biological activity of Ferula fukangensis KM Shen. J. Xinjiang Med. Univ. 2012, 35, 1159–1161. [Google Scholar]
  134. Mai, Q.F.; Chen, C.L. Studies on the volatile oils of two types of Ferula from Xinjiang. Chin. Tradit. Herb. Drugs 1983, 14, 10. [Google Scholar]
  135. Ren, Y.; Zhang, H.; Zhou, L. Experimental study on the toxicity of Ferula sinkiangensis. Xinjiang J. Tradit. Chin. Med. 2014, 32, 28–31. [Google Scholar]
  136. Liu, C.X.; Chen, S.L.; Xiao, X.H.; Zhang, T.J.; Hou, W.B.; Liao, M.L. A new concept on quality marker of Chinese materia medica: Quality control for Chinese medicinal products. Chin. Tradit. Herb. Drugs 2016, 47, 1443–1457. [Google Scholar]
  137. Wang, Z.H. Determination of ferulic acid in Angelica and research of quality standards. Hebei J. Tradit. Chin. Med. 2012, 34, 1058–1063. [Google Scholar]
  138. Zhang, X.L. Determination of ferulic acid in rhizoma chuanxiong by HPLC, China. Chin. Pharm. Aff. 2013, 23, 469–471. [Google Scholar]
  139. Zhao, X.D.; Chen, D.H.; Si, J.Y.; Shen, L.G. Studies on the phenolic acid constituents from Chinese medicine “Sheng-Ma”, rhizone of Cimicifuga foetida L. Acta Pharm. Sin. 2002, 37, 535–538. [Google Scholar]
  140. Zhang, J.L.; Zhou, Z.H.; Chen, R.Y.; Xie, F.Z.; Yu, D.Q.; Zhou, T.H. Study on chemistry and pharmcology of genus Ligusticum. Chin. Pharm. J. 2002, 37, 14–17. [Google Scholar]
  141. Li, F.; Zhu, X.; Chen, M.; Hou, Y. Study on the Chemical Constituents from Fructus Toosendan. J. Chin. Med. Mater. 2010, 33, 910–912. [Google Scholar]
  142. Li, D.; Rui, Y.X.; Guo, S.D.; Luan, F.; Liu, R.; Zeng, N. Ferulic acid: A review of its pharmacology, pharmacokinetics and derivatives. Life Sci. 2021, 284, 119921. [Google Scholar] [CrossRef]
  143. Rahman, A.-U. Studies in Natural Products Chemistry; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar]
  144. Appendino, G.; Mercalli, E.; Fuzzati, N.; Arnoldi, L.; Stavri, M.; Gibbons, S.; Ballero, M.; Maxia, A. Antimycobacterial Coumarins from the Sardinian Giant Fennel (Ferula communis). J. Nat. Prod. 2004, 67, 2108–2110. [Google Scholar] [CrossRef]
  145. Yang, X.W. Bioactive Material Basis of Medicinal Plants in Genus Ferula. Mod. Chin. Med. 2018, 20, 123–144. [Google Scholar]
  146. Liu, C.X. Five-year review on development of quality markers of traditional Chinese medicine. Chin. Tradit. Herb. Drugs 2021, 52, 2511–2518. [Google Scholar]
  147. Zhou, K.; Xie, J.L.; Li, Y.J.; Wang, Y.L.; Zhang, X.; Liu, C.H. Advances in the study of the chemical composition and pharmacological effects and quality marker prediction of lysiontus pauciflorus. China Pharm. 2021, 32, 1391–1396. [Google Scholar]
  148. Lian, X.Y.; Wu, Y.L.; Feng, H.S. A brief discussion on the medicinal characteristics of bitter medicines and their combinatory effects. Heilongjiang J. Tradit. Chin. Med. 1999, 1, 52–53. [Google Scholar]
  149. Hao, X.X. Overview of bitter substance research. J. Huanggang Norm. Univ. 2008, 28, 90–92. [Google Scholar]
  150. Meng, Z.H.; Sun, W.N.; Fu, X.Y.; Zhou, X.; Wan, J. Progress on the mechanism of bitterness and evaluation methods of traditional Chinese medicine. J. Guangdong Pharm. Univ. 2016, 32, 537–540. [Google Scholar]
  151. Zhou, F.H.; Yi, Z.X.; Luo, X.F. Analysis of the chemical composition of xing(sin) taste herbs. J. Anhui Agric. Sci. 2006, 12, 2760–2782. [Google Scholar]
  152. China Herb Company. Commonly Used Chinese Herbs in China; Science Press: Beijing, China, 1995. [Google Scholar]
  153. Liang, Y.H.; Liu, W.X.; Wang, X.F.; Zhu, Y. Materia medica evidence of the efficacy of Uyghur medicine Ferula. J. Chin. Med. Mater. 2017, 40, 1478–1481. [Google Scholar]
  154. Liu, Y.M.; Shawuti, Y. Uyghur Pharmacopoeia (First Book); Xinjiang People’s Publishing House: Xinjiang, China, 1999; pp. 250–255. [Google Scholar]
  155. Zhang, L.; Fu, X.; Gui, T.; Wang, T.; Wang, Z.; Kullak-Ublick, G.A.; Gai, Z. Effects of Farnesiferol B on Ischemia-Reperfusion-Induced Renal Damage, Inflammation, and NF-kappaB Signaling. Int. J. Mol. Sci. 2019, 20, 6280. [Google Scholar] [CrossRef] [Green Version]
  156. Liang, N.; Sun, S.P.; Luo, Y.E.; Qiu, L.Z.; Liu, B. Advances in ferulic acid research. Heilongjiang J. Tradit. Chin. Med. 2009, 38, 39–40. [Google Scholar]
  157. Hu, Y.Y.; Xu, X.Y. Advances in the chemical and pharmacological study of ferulic acid. Chin. Tradit. Pat. Med. 2006, 28, 253–255. [Google Scholar]
  158. Xu, J.X.; Xu, J.; Cao, Y.; Zhu, Y.J.; Li, X.Y.; Ge, D.Z.; Ma, L.; Zhang, T.J.; Liu, C.X. Modern research progress on Chinese medicine Cynanchum otophyllum Schneid and its predictive analysis of quality markers. China J. Chin. Mater. Med. 2021, 46, 5486–5495. [Google Scholar]
  159. Gao, T.T.; Ding, X.; Tan, Y.; Wang, H.; Wang, X.F. Determination of Ferulic Acid in Different Parts of Ferula sinkiangensis Using HPLC. Nat. Prod. Res. Dev. 2013, 25, 1237–1239. [Google Scholar]
  160. Wang, D.D.; Liu, H.B.; Song, H.L.; Yang, W.X.; Jia, X.G.; Tian, S.G. Determination of Ferulic acid content in roots and leaves of Ferula fukanensis KM Shen. by HPLC. J. Xinjiang Med. Univ. 2012, 35, 1139–1142. [Google Scholar]
  161. Ainiwaer, A.; Ma, L.Y.; Ma, G.Z. HPLC determination on ferulic acid content of Xinjiang asafetida antiulcer extract. J. Xinjiang Med. Univ. 2009, 32, 1436–1438. [Google Scholar]
  162. Zhang, H.Y.; Li, W.; Zhou, L.L.; Jiang, L.; Zhou, M.X. Study on Quality Control of Ethyl-acetate Parts of Ferula sinkiangensis. Chin. J. Inf. Tradit. Chin. Med. 2016, 23, 83–86. [Google Scholar]
  163. Zhu, Y.; Zhang, K.; Liang, X.; Tan, Y.; Huang, J.; Wang, J.H. Content Determination of Sesquiterpene Coumarin (DAW22) in Ferula ferulaeoides by UPLC. Chin. J. Exp. Tradit. Med. Formulae 2016, 22, 63–65. [Google Scholar]
Figure 1. Field morphological pictures of several Ferula plants. (A). Ferula sinkiangensis [5]; (B). F. fukanensis [6]; (C). F. kingdon-wardii [7]; (D). F. songarica [8]; (E) F. syreitschikowii [9]; (F). F. licentiana [10]. ((AF) are from the Plant Photo Bank of China, http://www.iplant.cn/, 16 June 2023).
Figure 1. Field morphological pictures of several Ferula plants. (A). Ferula sinkiangensis [5]; (B). F. fukanensis [6]; (C). F. kingdon-wardii [7]; (D). F. songarica [8]; (E) F. syreitschikowii [9]; (F). F. licentiana [10]. ((AF) are from the Plant Photo Bank of China, http://www.iplant.cn/, 16 June 2023).
Molecules 28 05191 g001
Figure 2. Chemical structures of compounds 124 from Ferula plants.
Figure 2. Chemical structures of compounds 124 from Ferula plants.
Molecules 28 05191 g002
Figure 3. Chemical structures of compounds 2547 from Ferula plants.
Figure 3. Chemical structures of compounds 2547 from Ferula plants.
Molecules 28 05191 g003
Figure 4. Chemical structures of compounds 4870 from Ferula plants.
Figure 4. Chemical structures of compounds 4870 from Ferula plants.
Molecules 28 05191 g004
Figure 5. Chemical structures of compounds 7186 from Ferula plants.
Figure 5. Chemical structures of compounds 7186 from Ferula plants.
Molecules 28 05191 g005
Figure 6. Chemical structures of compounds 87106 from Ferula plants.
Figure 6. Chemical structures of compounds 87106 from Ferula plants.
Molecules 28 05191 g006
Figure 7. Chemical structures of compounds 230255 from Ferula plants.
Figure 7. Chemical structures of compounds 230255 from Ferula plants.
Molecules 28 05191 g007
Figure 8. Chemical structures of compounds 256263 from Ferula plants.
Figure 8. Chemical structures of compounds 256263 from Ferula plants.
Molecules 28 05191 g008
Figure 9. Chemical structures of compounds 264273.
Figure 9. Chemical structures of compounds 264273.
Molecules 28 05191 g009
Table 1. Distribution table of domestic Ferula plants.
Table 1. Distribution table of domestic Ferula plants.
NoPlant NameMajor OriginsGrowing EnvironmentLiterature
1F. sinkiangensis K.M. ShenYining, Xinjiang750~1000 m in alpine meadows, stony slope areas[40]
2F. fukanensis K.M. ShenFukang, Xinjiang, Southern Gurbantunggut Desert, XinjiangThe arid inland Gobi Desert[41]
3F. ferulaeoides (Steud.) KorovinJunggar Basin and Tacheng Basin, XinjiangGrowing in 430~1000 m sand dunes, sandy land, the environment is mostly desert land[42]
4F. conocaula Korov.Ucha County, XinjiangElevation 2700~3000 m in mountain valleys, environment is mostly mountainous yellow-brown desert soil[42]
5F.syreitschikowii K.-Pol.Yili, Tacheng, Bole, XinjiangThe environment is mostly low mountain brown calcareous soils, wastelands and gravelly slopes [43]
6F. songarica Pallas ex SprengelTacheng and Altay, XinjiangThe environment is mostly in mountainous grassy slopes and mountainous bushes[42]
7F. krylovii Korov.Tori County, XinjiangThe environment is mostly clayey saline grassland[44]
8F. lehmannii Boiss.Manas County, XinjiangOn low mountain slopes at 600~700 m elevation, the environment is mostly clayey gravelly sandy calcareous soil[45]
9F. bungeana KitagawaHeilongjiang, Jilin, Liaoning, Inner Mongolia, Hebei, Henan, etc.The environment is mostly sandy and gravelly desert soil or near sandy areas[46]
10F. dissecta (Ledeb.) LedebTacheng Region, Altay Region, Junggar Basin, XinjiangThe environment is mostly sandy and gravelly desert slopes, mostly in the mountains dominated by Artemisia spp.[47]
11F. jaeschkeana VatkaAli, Zada, TibetThe environment is mostly at 3600 m above sea level on mountain slopes[48]
12F. akitschkensis B. Fedtsch. ex K.-Pol.Xinjiang Alatau Mountains, Altai Mountains and western Junggar MountainsThe environment is mostly mountain shrublands and gravelly slopes at 900~2100 m above sea level.[3]
13F. ovina (Boiss.) Boiss.Altai, Tacheng, XinjiangThe environment is mostly gravelly hillsides[3]
14F. hexiensis K. M. ShenSouthern GansuThe environment is mostly hillside with low humidity[3]
15F. olivacea (Diels) Wolff ex Hand.-Mazz.Lijiang, YunnanThe environment is mostly in canyons and mountain gaps, woods and grasses[49]
16F. caspica M. Bieb.Burqin and Tacheng, XinjiangThe environment is mostly low mountain slopes and mountain gaps and desert areas[50]
17F. sumbul (Kauffm.) Hook. f.Zhaosu County, XinjiangThe environment is mostly mountainous scrub and gravel slopes[49]
18F. licentiana Hand.-Mazz.Taihang MountainsThe environment is mostly valley grassland at 400~600 m above sea level[51]
19F. licentiana Hand.-Mazz.Jiangsu, Shandong, Anhui, etc.The environment is mostly sunny slopes, mountain rock crevices and hillsides[52]
20F. gracilis (Ledeb.) Ledeb.Xinjiang Altai RegionThe environment is mostly meadows, riverside forest edges and gravelly mountain slopes[3]
21F. karataviensis (Regel et Schmaih.) Korov.Xinyuan County, XinjiangThe environment is mostly gravelly hillsides[3]
22F. dubjanskyi Korov. ex PavlovGansu Province, Anxi and Su Bei, Mahaoshan areaThe environment is mostly desert and Gobi desert in the sand and dunes[53]
23F. canescens (Ledeb.) Ledeb.Fuyun County, XinjiangGravelly hillsides in a mostly desert environment[3]
24F. kingdon-wardii WolffNorthwest YunnanThe environment is mostly grassy slopes and rock crevices at 2700~3200 m above sea level[54]
25F. kirialovii PimenovTianshan Mountains and Junggar Basin, XinjiangThe environment is mostly gravelly grassy slopes and shrubby places at an altitude of about 1500 m[55]
26F. lapidosa Korov.Chabchal County, XinjiangThe environment is mostly mountainous gravelly slopes and grassy areas[3]
Table 2. Coumarin compounds in domestic Ferula plants.
Table 2. Coumarin compounds in domestic Ferula plants.
NoCompoundTypeLiterature
1(3′S, 8′R, 9′S, 10′R)-sinkianol AA[52]
2(5′S, 8′R, 9′S, 10′R)-ferukrinoneA[52]
3FerukrinA[52]
4(3′S, 5′S, 8′R, 9′S, 10′R)-kellerinA[52]
5(3′S, 5′S, 8′R, 9′S, 10′R)-deacetylkellerinA[52]
6Farnesiferol AA[52]
7Farnesiferone AA[52]
8GummosinA[52]
9PolyanthininA[52]
10(3′R, 5′R, 10′R)-sinkianol BB[52]
11Farnesiferol BB[52]
12Farnesiferol CB[52]
13Galbanic acidB[52]
14Methyl galbanateB[52]
15Sinkiangenol AA[52]
16KaratavicinolC[52]
17UmbellipreninC[52]
18Sinkiangenol BA[53]
19Sinkiangenol CB[53]
20Sinkiangenol DA[53]
21Sinkiangenol EA[53]
22Sinkiangenol FE[53]
232,3-Dihydro-7-hydroxy-2R*,3S*-dimethyl-2-
[4-methyl-5-(4-methyl-2-furanyl)-3(E)-pentenyl]-furano[3,2-c] coumarin
D[53]
242,3-Dihydro-7-hydroxy-2R*,3R*-dimethyl-2-
[4-methyl-5-(4-methyl-2-furanyl)-3(E)-pentenyl]-furano[3,2-c]coumarin
D[53]
2512′-hydroxy-karatavicinolC[53]
26FekrynolB[53]
27ActylfekrynolB[53]
28FerocaulidinA[53]
29FekrolB[53]
30FerucrinoneA[53]
31DeacetylkellerinA[53]
32KellerinA[53]
33ColladocinA[53]
34LehmannololA[53]
35KamoloneA[53]
36Assafoetidnol BA[53]
37Assafoetidnol AA[53]
38Lehmannolone AA[54]
39AssafoetidinB[54]
40Lehmannolone B[55]
41SinkianoneB[55]
42ColladoninE[56]
43EpisamarcandinA[57]
44Sinkiangenorin DA[57]
45FekoloneB[57]
46FeselolA[57]
47Ferulin AD[58]
48Ferulin BD[58]
49Ferulin CD[58]
502,3-Dihydro-7-hydroxy-2S*,3R*-dimethyl-2-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c] coumarinD[58]
512,3-Dihydro-7-hydroxy-2R*,3R*-dimethyl-2-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c] coumarin (DAW22)D[58]
522,3-Dihydro-7-hydroxy-2S*,3R*-dimethyl-2-[4-methyl-5-(4-methyl-2-furanyl)-3(E)-pentenyl]-furo-[3,2-c] coumarinD[58]
532,3-Dihydro-7-hydroxy-2R*,3R*-dimethyl-2-[4-methyl-5-(4-methyl-2-furanyl)-3(E)-pentenyl]-furo[3,2-c] coumarinD[58]
542,3-Dihydro-7-methoxy-2S*,3R*-dimethyl-2-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c] coumarinD[58]
552,3-Dihydro-7-methoxy-2R*,3R*-dimethyl-2-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c] coumarinD[58]
562,3-Dihydro-7-hydroxy-2S*,3R*-dimethyl-3-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c] coumarinD[58]
572,3-Dihydro-7-hydroxy-2R*,3R*-dimethyl-3-[4,8-dimethyl-3(E),7-nonadienyl]-furo[3,2-c] coumarinD[58]
584,7-Dihydroxy-3-[3,7,11-trimethyl-2(E),6(E),10-dodecatrienyl] coumarinE[58]
592,3-Dihydro-7-hydroxy-2R*,3R*-dimethyl-2-[4,8-dimethyl-3(E),7-nonadiene-6-onyl] furo[3,2-c] coumarinD[59]
60Fukanefuromarin AD[59]
61Fukanefuromarin BD[59]
62Fukanefuromarin CD[59]
63Fukanefuromarin DD[59]
64Fukanemarin AE[59]
65Fukanefuromarin HD[60]
66Fukanefuromarin ID[60]
67Fukanefuromarin JD[60]
68Fukanefuromarin KD[60]
69Fukanefuromarin LD[60]
70Fukanefuromarin ME[60]
Table 3. Sesquiterpenoids in Domestic Ferula Plants.
Table 3. Sesquiterpenoids in Domestic Ferula Plants.
NoCompoundLiterature
71Ferulaeone A[61]
72Ferulaeone B[61]
73Ferulaeone C[61]
74Ferulaeone D[61]
75Ferulaeone E[61]
76Ferulaeone F[61]
77Ferulaeone G[61]
78Ferulaeone H[61]
79Dshamirone[61]
80(4E,8E)-1-(2,4-dihydroxyphenyl)-2-hydroxy-5,9,13-trimethylt-etra-deca-4,8,12-trien-1-one[61]
81(6E)-1-(2,4-dihydroxyphenyl)-3,7,11-trimethyl-3-vinyl-6,10-dodecadien-1-one[61]
82(6E)-1-(2,4-dihydroxyphenyl)-3,7-dimethyl-3-vi-nyl-8-(4-methyl-2-furyl)-6-octen-1-one[61]
833-(2,4-dihydroxybenzoyl)-4S*,5R*-dimethyl-5-
[4,8-dimethyl-3(E),7(E)-nonadien-1-yl] tetrahydro-2-furanone
[61]
843-(2-hydroxyl-4-methoxybenzoyl)-4S*,5R*- dimethyl-5-
[4,8-dimethyl-3(E),7(E)-nonadien-1-yl] tetrahydro-2-furanone
[61]
853-(2,4-dihydroxybenzoyl)-4R*,5R*-dimethy-5-
[4,8-dimethyl-3(E),7(E)-nonadien-1-yl] tetrahydro-2-furanone
[61]
861-(2′,4′-dihydroxyphenyl)-3,7,11-trimethyl-3-vinyl-6(E),10-dodecadien-1-one[62]
872,3-dihydro-7-hydroxy-2,3-dimethyl-2-[4′,8′-dimethyl-3′,7′-nonadienyl]
-furo [3,2,c] coumarin
[62]
882,3-dihydro-7-hydroxy-2,3-dimethyl-3-[4′,8′-dimethyl-3′,7′-nonadienyl]
-furo [3,2,c] coumarin
[62]
89Fukanefurochromones A[63]
90Fukanefurochromones B[63]
91Fukanefurochromones C[63]
92Fukanefurochromones D[63]
93Fukanefurochromones E[63]
942,3-dihydro-7-hydroxy-2S*,3R*-dimethyl-2-[4,8-dimethyl-3(E),7-nonadienyl]-furo[2,3-b] chromone[63]
95Fukanedone A[64]
96Fukanedone B[64]
97Fukanedone C[64]
98Fukanedone D[64]
99Fukanedone E[64]
100Fukaneketoester A[64]
101Feruone[5]
1025α-(p-hydroxybenzyl) ester of ferutriol[65]
103Ferutriol[65]
1045ɑ-isovalerate of ferutriol[65]
105Jaeschkeanadiol[65]
106Lapidin[65]
Table 4. The composition of volatile oils of F. sinkiangensis, F. fukanensis, F. ferulaeoides, F. ovina.
Table 4. The composition of volatile oils of F. sinkiangensis, F. fukanensis, F. ferulaeoides, F. ovina.
NoCompoundMolecular FormulaLiterature
107(1S)-β-PineneC10H16[73]
1081,7,7-Trimethyl-tricyclo[2.2.1.0 (2,6)] heptaneC10H16[73]
109(1R)-α-PineneC10H16[73]
110CampheneC10H16[73]
1116-Methyl-5-hepten-2-oneC8H14O[73]
112β-PineneC10H16[73]
113α-PhellandreneC10H16[73]
1143-CareneC10H16[73]
115TerpinoleneC10H16[73]
1161-Isopropyl-2-methylbenzeneC10H16[73]
117d-LimoneneC10H16[73]
118(E)-3,7-Dimethyl-1,3,6-octatrieneC10H16[73]
119(Z)-3,7-Dimethy-1,3, 6-octatrieneC10H16[73]
120γ-TerpineneC10H16[73]
121l-CamphorC10H16O[73]
122BorneolC10H16O[73]
123(−)-α-CopaeneC15H24[73]
1242-Methoxy-4-methyl-1-(1-methylethy)-benzeneC11H16O2[73]
1251-Methoxy-4-methyl-2-(1-methylethyl)-benzeneC11H16O[73]
126L-Bornyl acetateC12H20O2[73]
127Lavandulol acetateC12H20O2[73]
128(+)-α-LongipineneC15H24[73]
129(Z)-2,6,10-Trimethy-1,5,9-undecatriene C14H24[73]
130CaryophylleneC15H24[73]
131(1S,2S,4R)-2-Acetate-1,3,3-trimethyl
-bicyclo[2.2.1] hepten-2-ol
C12H20O2[73]
1321-Ethenyl-1-methyl-2-(1-methylethenyl)-
4-(1-methylethylidene)-cyclohexane
C15H24[73]
1331S-(1α,4α,7α)]-1,2,3,4,5,6,7,8-Octahydro
-1,4-dimethyl-7-(1-methylethylenyl)-azule
C15H24[73]
134(4αS,9αR)-2,4α,5,6,7,8,9,9α-Octahydro-
3,5,5-trimethyl-9-methyllene-benzocycloheptene
C15H24[73]
135(3Z,6E) -3,7,11-Trimethyl-1,3,6,10-dodecanetetraeneC15H24[73]
136α-FarneseneC15H24[73]
137Isolongjflene-8-olC15H26O[73]
138Tran-nerolidolC15H26O[73]
139GuaiolC15H26O[73]
140α-EudesmolC15H26O[73]
141(2R,4αR)-1,2,3,4,4α,5,6,7-Octanhydro-α, α
-3,8-tetramethyl-naphthalenemethanol
C15H26O[73]
142[3S-(3α,3,5α)]-1,2,3,3α,4,5,6,7-Octahydro-α, α
-3,8-tetramethyl-5-azulenemethanol
C15H26O[73]
143(1S)-1-[(1S)1,5-Dimethyl-4-hexen-l-
4-methyl-3-cyclohexen-1-ol
C15H26O[73]
144IsoledeneC15H24[73]
145EthanolC2H6O[74]
146Ethyl esterC4H8O2[74]
1473-Methyl-butanalC5H10O[74]
1482-ButanethiolC4H10S[74]
1492,5-Dimethyl-furanC6H8O[74]
150FurfuralC5H4O2[74]
151Methyl 1-propenyl disulfdeC4H8S2[74]
152Dimethyl trisulfideC2H6S3[74]
153n-Propyl sec-butyl disulfideC7H16S2[74]
154Propyl n-butyl disulfideC7H16S2[74]
1551,2-DithiacyclopentaneC3H6O2[74]
1562-Ethyl-hexanethiolC8H18S[74]
157bis (1-Methylpropyl)-disulfideC8H18S2[74]
1582,2′-BioxiraneC4H6O2[74]
1593-Methyl-4-heptanolC8H18O[74]
1601,1-bis (Methylthio)-ethaneC4H10O2[74]
1611,1-Dimethoxy-propaneC5H12O2[74]
162HeptadecaneC17H36[74]
1632-Ethylthio-butaneC6H14S[74]
164α-HumuleneC15H24[74]
165β-SelineneC15H/[74]
166HexadecaneC16H32[74]
1672-Methylthio butyrat methyl esterC6H12OS[74]
1682,3-Dimeth yl-3-hexanolC8H18O[74]
169HedycaryolC15H26O[74]
1702,2-bis (Methylthio)-propaneC5H12S2[74]
171OctadecatrieneC18H32[74]
172Palmitic acidC16H32O2[74]
173Oleic acidC18H34O2[74]
174α-MyrceneC10H16[75]
175o-CymeneC10H14[75]
176EnolC10H16O[75]
177Limonene oxideC10H16O[75]
178CamphorC10H16O[75]
179ThymolC10H14O[75]
180α-TerpineolC10H18O[75]
181Anisyl acetateC12H2O2[75]
182Carvacrol acetateC10H16O[75]
1831-Methoxy-4-methyl-2-(1-ethyl)-benzeneC11H16O[75]
184CarvoneC10H14O[75]
185Bornyl acetateC12H2O2[75]
186(−)-VerbenoneC10H16O[75]
187α-(E)-BETA-FARNESENEC15H24[75]
1881-Cycloethyl-1-pentyneC11H18[75]
189α-BergamoteneC15H24[75]
1901,11-HexadecadiyneC16H26[75]
191α-Bisabolene C15H24[75]
192AsaroneC12H16O3[75]
1933,7,11-trimethyl(E)1,6,10-dodecatrien-3-olC15H26O[75]
194FarnesolC15H26O[75]
195Graphene oxideC15H24O[75]
196Disulfide bis(1-methylpropyl)C8H18S2[75]
197n-Propyl sec-butyl disulfideC7H16S2[76]
198OcimeneC10H16[76]
199Ocimene (Mixture of isomers)C10H16[76]
200Disulfide, bis[1-(methylthio)ethyl]C6H14S4[76]
2012-Methylbutyl benzeneC11H16[76]
202Ethyl 1-methylpropyl disulfideC6H14S2[76]
203(Z)-1,6,10-Dodecatriene-7,11-dimethyl-3-methyleneC15H24[76]
204cis-α-BisaboleneC15H24[76]
205Dipropyl disulfideC6H14S2[76]
206HinesolC15H26O[76]
207NeoisolongifoleneC15H22[76]
208Bicyclo[4.4.0]dec-l-ene,2-isopropyl-5-methyl-9-methyleneC15H24[76]
2092H-Pyran, tetrahydro-4-methyl-2-(2-methyl-1-propenyl)C10H18O[76]
2102,4,6-Octatriene, 2,6-dimethyl-, (E,Z)-C10H16[76]
211EstragoleC10H12O[76]
2121-Methoxy-4-methyl-2-(1-methylethyl) BenzeneC11H16O[76]
2132,6-Dimethyl-2,6-octadieneC10H18[76]
214Methyl 2-(methylthio)butyrateC6H12O2S[76]
2151-Methyl-4-(1-methylethyl)-1,3-cyclohexadiene,C10H16[76]
216Methyl sec-butyl disulphideC6H12O2S[76]
2173-(Methylthio)-2-butanoneC5H10OS[76]
218Naphthalene,1,2,3,4.4a,5,6,8a-octahydro-7
-methyl-4-methylene-1-(1-methyl-ethyl)-, (1α.,4aβ.,8aα)-
C15H22[76]
2191,4-Methanoazulene, decahydro-4,8,8-trimethyl-9-methylene-,
[IS-(l.a.,3a.β.,4a.,8a.β)]-
C15H26O[76]
220Tetrahydro thiazoleC5H12S2[76]
221(4aS-cis)-2,4a,5,6,7,8,9,9a-octahydro-
3,5,5-trimethyl-9-methylene -1H-Benzocycloheptene
C5H10OS[76]
222ThiopropionamideC3H7NS[76]
223Longifolene-(V4)C15H24[76]
224Naphthalene, 1,2,3,4,4a,5,6,8a-octahydro
-4a,8-dimethyl-2-(1-methyl ethenyl)-, [2R-(2α.,4aα.,8aβ)]
C15H24[76]
225SeychelleneC3H7NS[76]
226(R)-2,4a,5,6,7,8-hexahydro-3,5,5,9-tetramethyl-1H-benzocyclohepteneC15H24[76]
227[S-(Z)]-3,7,11-trimethyl-1,6,10-Dodecatrien-3-olC15H24[76]
228E-Famesene epoxideC3H7NS[76]
229Di-epi-α-cedreneC15H24[76]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bahetjan, Y.; Muhaxi, M.; Pang, K.; Kizaibek, M.; Tang, H.; Sefidkon, F.; Yang, X. Chemistry, Bioactivity, and Prediction of the Quality Marker (Q-Marker) of Ferula Plants in China: A Review. Molecules 2023, 28, 5191. https://doi.org/10.3390/molecules28135191

AMA Style

Bahetjan Y, Muhaxi M, Pang K, Kizaibek M, Tang H, Sefidkon F, Yang X. Chemistry, Bioactivity, and Prediction of the Quality Marker (Q-Marker) of Ferula Plants in China: A Review. Molecules. 2023; 28(13):5191. https://doi.org/10.3390/molecules28135191

Chicago/Turabian Style

Bahetjan, Yerlan, Muguli Muhaxi, Kejian Pang, Murat Kizaibek, Hui Tang, Fatemeh Sefidkon, and Xinzhou Yang. 2023. "Chemistry, Bioactivity, and Prediction of the Quality Marker (Q-Marker) of Ferula Plants in China: A Review" Molecules 28, no. 13: 5191. https://doi.org/10.3390/molecules28135191

Article Metrics

Back to TopTop