Next Article in Journal
New Bi-Nuclear Nickel(II) Complex-Based Salen Schiff Base: Synthesis, Crystal Structure, Spectroscopic, Thermal, and Electrical Investigations
Next Article in Special Issue
Promising Anticancer Activity of β-Carboline Derivatives: Design, Synthesis, and Pharmacological Evaluation
Previous Article in Journal
Sweet, Sugar-Coated Hierarchical Platinum Nanostructures for Easy Support, Heterogenization and Separation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Traditional Uses, Phytochemical Composition, Pharmacological Properties, and the Biodiscovery Potential of the Genus Cirsium

1
Council of Scientific & Industrial Research—Institute of Himalayan Bioresource Technology, Palampur 176061, India
2
Department of Zoology, Mata Gujri College, Fatehgarh Sahib 140407, India
3
Department of Chemistry, Sant Longowal Institute of Engineering and Technology, Sangrur 148106, India
4
Department of Chemistry, Chandigarh University, Mohali 140413, India
5
Department of Food Science & Technology, Govt. Degree College Shopian, Jammu & Kashmir 192303, India
6
University Centre for Research & Development, Department of Chemistry, Chandigarh University, Mohali 140413, India
*
Author to whom correspondence should be addressed.
Chemistry 2022, 4(4), 1161-1192; https://doi.org/10.3390/chemistry4040079
Submission received: 27 August 2022 / Revised: 25 September 2022 / Accepted: 27 September 2022 / Published: 30 September 2022

Abstract

:
Medicinal plants are rich in phytochemicals, which have been used as a source of raw material in medicine since ancient times. Presently they are mostly used to treat Henoch–Schonlein purpura, hemoptysis, and bleeding. The manuscript covers the classification, traditional applications, phytochemistry, pharmacology, herbal formulations, and patents of Cirsium. The main goal of this review is to impart recent information to facilitate future comprehensive research and use of Cirsium for the development of therapeutics. We investigated numerous databases PubMed, Google Scholar, Springer, Elsevier, Taylor and Francis imprints, and books on ethnopharmacology. The plants of the genus Cirsium of the family Asteraceae contain 350 species across the world. Phytochemical investigations showed that it contains flavonoids, phenols, polyacetylenes, and triterpenoids. The biological potential of this plant is contributed by these secondary metabolites. Cirsium plants are an excellent and harmless agent for the cure of liver diseases; therefore, they might be a good clinical option for the development of therapeutics for hepatic infections. The phytochemical studies of different Cirsium species and their renowned pharmacological activities could be exploited for pharmaceutic product development. Furthermore, studies are required on less known Cirsium species, particularly on the elucidation of the mode of action of their activities.

Graphical Abstract

1. Introduction

In recent years, there has been a surge in interest in natural products for the prevention and cure of many diseases such as cancer, arthritis, cardiovascular disorders, and diabetes. Plants and their natural products are reservoirs of phytoconstituents that have antimicrobial, antioxidant, anticancer, and antidiabetic properties and are used in traditional medicine [1]. Plant extracts provide limitless opportunities for novel medicinal discoveries due to their unrivaled chemical variety and biocompatible nature. Many studies are being conducted to identify an alternative therapy using medicinal plants. In reality, roughly 25% of the medications on the market are derived directly or indirectly from plants [2]. Many of these plants have recently been suggested for their capacity to act as a preservative and food additive, serving a dual function of culinary taste and bioactive constituent [3].
Cirsium is a genus of perennial and rarely annual prickly Asteraceae plants. It gets its name from the Greek word “khirsos,” which means “swollen vein.” According to the Plants of the World online collections at the Royal Botanic Gardens of Kew, there are around 450–480 recognized species in this genus [4,5]. These plants are found in the northern hemisphere, including Eurasia, Asia, North America, and North Africa. There are around 120 and 50 Cirsium species in Japan and China, respectively [6,7]. The Cirsium has been used traditionally as a folk medicine for the treatment of various ailments in India and in many neighboring countries such as China for centuries. Many species of Cirsium are exploited as an herbal remedy for the cure of cardiovascular diseases and used as anti-inflammatory and diuretic agents traditionally. Cirsium is still employed clinically as cold blood hemostatic medicine. Pharmacological research studies have revealed that Cirsium is valued for its wide range of therapeutic benefits, such as hepatoprotection, antioxidant, anticancer, antimicrobial, and anti-inflammation, which may untie the multiple medicinal applications of Cirsium [8]. Moreover, some Cirsium species have been used for the cure of diabetes, nervousness, and overweightness. Cirsium is a multifunctional herb that is utilized for the cure of hemoptysis, hematuria, distressing bleeding, and Henoch–Schonlein purpura. Phytochemical investigations showed that the Cirsium species contained flavonoids, polyacetylenes, acetylenes, phenolic acids, phenylpropanoids, sterols, and terpenoids [9]. Among them, flavonoids, phenylpropanoids, and terpenoids are considered to be the main phytocompounds and are accountable for a large number of biological properties found in the different species of this genus. Variation in pharmacological activity is attributed to the different chemical compositions in different Cirsium species from different regions [10,11]. This review summarizes and evaluates the existing traditional uses, botanical description, phytochemical composition, pharmacokinetics, and biological properties of the Cirsium genus. Furthermore, the progress of research on herbal formulations, patents, and the safe profile of Cirsium is also discussed. This review article represents updated information on Cirsium to benefit future scientific investigations for the production of novel drug formulations and clinical trials.

2. Methodology

This review article was designed by collecting relevant information about the genus Cirsium from various databases, i.e., PubMed, Google Scholar, Chem Spider, Springer, World Scientific, Science Direct Elsevier, Taylor and Francis imprints, peer-reviewed journals, and some of the unpublished data. In addition, several ‘grey literature’ sources, such as Wikipedia, online sites, ethnobotanical books, and chapters, are included in the data sources.

3. Cirsium

There are nearly 450 Cirsium species found across the world [8]. Around 200 of them are found in various regions of Asia, Central America, Europe, North Africa, and North America [12]. Out of these species, about 16 known species are found in Indian evergreen forests and some regions bordering Nepal and China also, viz. C. argyracanthum, C. arvense, C. lineare, C. eriophoroides, C. falconeri, C. flavisquamatum, C. interpositum, C. verutum, C. nishiokae, C. phulchokiense, C. shansiense, C. souliei, C. tibeticum, C. verutum, C. wallichii and C. glabrifolium [13]. Cirsium originated in Eurasia and Northern Africa, and roughly 60 species have been identified from North America. It can be found in every continent except Antarctica, albeit its range is largely limited to the northern and southern temperate regions [14,15,16] (Table 1). Cirsium genus comes under the Asteraceae family (Figure 1), and this family consists of about 1600 genera and 23,000 species [15]. Most of the species of this family are perennial thistles and have spines on leaves, flowers, stems, and roots or some time on the whole plant. Species of genus Cirsium are entitled ‘common thistle’, and ‘field thistle’ (Table 1). More precisely, they are termed ‘Plume thistles’ since some genera, i.e., Carduus, Silybum, and Onopordum, are frequently labeled by the term ‘thistle’ [16]. Cirsium is considered a weed that grows anywhere, including near farmed areas. Despite their status as weeds, they have the potential for allelopathic impact and biological pest control [17]. Some species are even planted in gardens owing to their aesthetic appeal [18,19].

4. Botanical Description of Genus Cirsium

Thistles are recognized by their demonstrative flower heads (purple, rose, pink, yellow, or white) with many tiny flowers. Flowers are radially symmetrical disc-like structures blooming at the end of the branches. Their flowers bloom from April to August [9]. Plant constitutes straight stems and prickly leaves having a distinctive enlarged base of flower which are usually spiny; leaves are alternate, some species can be less or more hairy; leaf extension leading to the stems called wings, can be conspicuous (Cirsium vulgare), lacking, or inconspicuous (Figure 2). They can reproduce by pollination and also through rhizomes (Cirsium arvense) and are annual, biennial, and some are perennial in growth form [10,26].

5. Traditional Uses

Currently, traditional plants have emerged as a viable avenue for the discovery of novel therapeutic molecules to treat a variety of severe ailments, such as diabetes and cancer [27]. More than 10 species of Cirsium are well known in Chinese medicines to cure jaundice, hemorrhaging, gastrointestinal issues, and vascular diseases [28]. Traditionally, Cirsium is also exploited as an herbal remedy for the healing of leukemia and peptic ulcers [29]. In Central America and Mexico, C. mexicanum is utilized in traditional medicine for the cure of respiratory problems, hepatic infections, diarrhea, dysentery, and stomach pain [23]. Moreover, Cirsium rivulare is utilized traditionally in Poland for anti-anxiety effects [30]. The fruits and roots are also used to cure constipation, dyspepsia, skin problems, chest discomfort, and as a tonic by the people of the Himachal region [31]. The indigenous culture of Meghalaya uses leaf extract to treat gastrointestinal ailments, primarily diarrhea, and dysentery. The Bhotiya tribal community of central Himalaya, India, uses the Cirsium the cure rheumatism [32]. Cirsium arvense roots are diuretic, astringent, antiphlogistic, and hepatoprotective. Root decoction of Cirsium has been traditionally employed for treating worm infection in children. Moreover, root paste of Cirsium arvense in combination with Amaranthus spinosus is used for the treatment of indigestion. Rheumatic joint pains have long been treated using a hot brew made from Cirsium arvense. Decoction of the whole plant has been used as a curative medicine internally and externally for bleeding piles [33]. It has been surveyed in Central Italy that Cirsium arvense leaf soup is used to alleviate digestive problems and stomach discomfort. Moreover, Cirsium arvense is also utilized to halt the flow of blood from wounds in an emergency [34]. Extract and infusions of C. arvense leaves are a good repository of fibers, vitamins, and important minerals, and these were used by North American Indians for toothaches, tuberculosis, throat sores, and cancer sores due to their potent anti-inflammatory properties [35]. Other species, such as Cirsium rivulare, have also been used traditionally for anxiety-related issues [36] and have been documented to exhibit antimitotic action [37]. Moreover, in Polish medicine, C. arvense and C. oleraceum have been reported to be used as a diuretic, antiphlogistic, and astringent [38]. C. rivulare, C. oleraceum, and C. vulgare have long been recognized for their anxiolytic properties in Poland’s Northeastern regions [39]. Aerial portions of C. chanroenicum have been reported to be utilized in Chinese medicine to cure pyrexia, detoxify, and improve blood circulation [40]. In addition to it, C. japonicum has been utilized in Chinese medicine as a hypertensive, anti-hemorrhagic, and anti-hepatitis agent [25], as well as in folk medicine for the cure of malignancies such as uterine and liver cancer [41]. The water extract of C. arisanense displayed a hepatoprotective effect and has been applied in Taiwanese traditional medication for hundreds of years [42]. In another work, Cirsium species seeds and root decoction have been employed in Turkish traditional medicine to cure hemorrhoids. Flowers of Cirsium species ameliorated peptic ulcers, and stems are used for the cure of cough and bronchitis in Anatolia [43]. Root paste of C. falconeri and C. verutum is useful for the cure of arthritis [22]. Moreover, the dried powder of flowers and leaves of C. falconeri has been found to mediate protective effects against Cerebral edema in the Himalayan province of India [44]. The stems and young leaves of C. setidens are edible in nature and high in calcium, proteins, and vitamin A. This species of Cirsium has been used to treat emesis, hypertension, and hematuria in Korean medicines [45]. Moreover, Cirsium vulgare is persistently grown in gardens nowadays to attract birds and butterflies. Hummingbirds are attracted to the flowers of C. vulgare, and many immigrants keep hummingbirds in their botanical gardens [16].

6. Culinary Uses of Cirsium

The leaves, roots, and stem of Cirsium are edible and can be consumed raw or cooked. This plant can be utilized in salads or in combination with other vegetables traditionally. The stem of Cirsium can be peeled and roasted like Asparagus. In several European countries, the leaves of a few Cirsium species are used to make tea, which displays tremendous medicinal properties [36,46,47,48]. The leaves of Cirsium have a fairly mild flavor and can be consumed either raw or cooked. The intake of boiled leaves serves as an effective diuretic and liver tonic. Moreover, a mixture of the soaked leaves and roots of the Cirsium is used as a remedy for the healing of neural problems. The flowers and roots of Cirsium are employed for the preparation of an infusion for drinking or applying vaginal douches. The seed oil of Cirsium is utilized for cooking and also as a lamp oil [49].

7. Phytochemical Composition

7.1. Flavonoids

Flavonoids are a kind of polyphenol present in medicinal plants that have been shown to have antioxidant and anticancer effects. The various species of the Cirsium contain all the categories of flavonoids, i.e., flavones, flavanones, and flavanols [17]. Most of the flavonoids are identified in C. rivulare, C. japonicum, and C. arvense. The chemical investigation recognized major flavones (Figure 3), which are common in many Cirsium species were Linarin, Luteolin, Luteolin 7-O-β-D-glucoside, Pectolinarin, Apigenin, Apigenin 7-O-glucoside, and Hispidulin [16,23,35,46,47,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64].
Besides these compounds, the infrequently distributed flavones were also found in Cirsium (Figure 4), namely Pectolinarigenin, Acacetin, 5,7-Dihydroxy-6,4′-dimethoxyflavone, Cirsimaritin, Cirsimarin, Rutoside and Tricin [51,53,54,57,59,60,65]. In addition to it, flavones glucoside was also detected in species of Cirsium (Figure 5), specifically Pectolinarigenin-7-O-glucopyranoside, Hispidulin-7-neohesperidoside, Hispidulin-7-glucoside, Luteolin 7-O-β-D-glucuronide, Isokaempferide-7-glucuronide, Isokaempferide-7-O-β-D-(6″-methylglucuronide), Apigenin 7-(6″-methylglucuronide), and 6-Hydroxyluteolin 7-O-glucoside [50,51,56,64]. In another work, Eriodictyol 7-O-glucoside [56], Kaempferol, Kaempferol 3-galactoside, Kaempferol 3-glucoside, and Isorhamnetin were detected in Cirsium species [50,61,63,66]. Moreover, some polyphenolic phytoconstituents 4-Vinyl guaiacol, 4-Ethyl guaiacol, Scopoletin, and 6,7-Dimethoxycoumarin were also reported in different species (Table 1 and Table 2) of Cirsium (Figure 6) [17,35,51]. These compounds are responsible for the natural aroma in plants and possess fungicidal properties. They can halt conidium germination and germ tube elongation in several plant pathogenic fungi [32,67].

7.2. Terpenoids and Sterols

The terpenes or terpenoids constitute the largest class of secondary products. They are synthesized by the Mevalonic acid pathway in the chloroplast of the plants. There are various types of terpenes utilized for the treatment of several infectious diseases. According to many studies, terpenes possess very good immunomodulatory properties [74]. The main triterpenes (Figure 7) detected in Cirsium species were Lupeol, Lupeol acetate, Taraxasterol acetate, 25-Hydroperoxycycloart-23-en-3β-ol, 24-Hydroperoxycycloart-25-en-3β-ol, α-Amyrin acetate, β-Amyrin acetate, β-Amyrin, and Faradiol [23,72]. In addition to it, other terpenes found in Cirsium species were α-Tocopherol, α-Tocospiro, 4 (15),10 (14)-Guaiadien-12, 6-olide (mokko lactone), t-Phytol, and Dihydroactinidiolide [17,23,35,70,71]. Among the sesquiterpenes, Caryophyllene oxide and β-Caryophyllene alcohol [71] were also observed in the Cirsium. Moreover, major sterols identified in Cirsium species were Acylglycosyl β-Sitosterol, β-Sitosterol glucoside, Taraxasterol, Stigmasterol, and Daucosterol (Figure 8) [23,46,61,72].

7.3. Phenolic Acids

The phenolic acid (Figure 9) in the different species of Cirsium were found to be Protocatechuic acid, Caffeic acid, Vanillic acid, Chlorogenic acid, p-Coumaric acid, p-Hydroxybenzoic acid, trans-Cinnamic acid, Syringic acid, and Caffeic acid [52,56,63,75].

7.4. Polyacetylenes, Acetylenes, and Hydrocarbons

The Acetylenes and Polyacetylenes have been proven in studies to be distinctive of the Asteraceae family [75,76,77,78,79,80,81,82,83]. However, Polyacetylenes and Acetylenes were found in the Cirsium, namely Aplotaxene, Dihydroaplotaxene, Pentacosane, Tetrahydroaplotaxene, and 1-Pentadecene [41,66,71,73]. Ciryneol and 1,2,15,16-Diepoxyhexadecane are two hydrocarbons detected in the Cirsium (Figure 10). Ciryneol is a cyclic ether that was found in C. arvense [51,71]. All these Acetylenes and Polyacetylenes were isolated from the non-aerial part of the Cirsium species [16].

7.5. Fatty acids, Aldehydes, and Ketones

The fatty acids identified in the Cirsium were 9,12,15-Octadecatrienoic acid, 9,12-Octadecadienoic acid, Hexadecanoic acid, and Palmitic acid [17,23,35,41,71,72]. Other carbonyl compounds (Figure 11) recognized in the Cirsium were Sinapaldehyde and 6,10,14-Trimethyl-2-pentadecanone [66,71].

8. Essential Oil Composition

The essential oil was extracted from C. acaule, C. arvense, C. creticum, C. decussatum, C. dissectum, C. eriophorum, C. heterophyllum, C. japonicum, C. ligulare, C. oleraceum, C. palustre, C. pannonicum, C. rivulare, and C. setidens [65,68,71,73,84,85,86]. It was observed that essential oil from Cirsium was found to be a rich source of Aplotaxene and Hexadecanoic acid. The maximum concentration of Aplotaxene was observed in the C. japonicum, C. palustre, and C. rivulare. However, Hexadecanoic acid was found to be more in C. japonicum, C. creticum, and C. setidens [68,71,73,86]. Other components such as (Z)-8,9-Epoxyheptadeca-1,11,14-triene in C. palustre and C. rivulare [73]; Pentadecanoic acid, Heptacosane, Heptadecanoic acid, Tetradecanoic acid, Palmitic acid, Caryophyllene oxide, and Myristic acid in rhizomes of C. japonicum [65]; Phytol in C. setidens and C. arvense [71,84]; 4-Ethyl guaiacol, (E)-β-Damascenone and Dihydroactinidiolide in C. creticum [17]; α-Bisabolol, δ-Cadinene, Hexacosane, β-Selinene, α-Humulene, Docosane, Octadecane, Eicosane, Germacrene-D, Nonacosane in C. arvense [85]; Thymol in inflorescences of C. pannonicum, and C. decussatum were detected [65].

9. Pharmacological Studies

Cirsium has abundant pharmacological activities due to the presence of a wide range of phytochemicals. Till date, a broad spectrum of biological properties such as antimicrobial, antioxidant, analgesic, anticancer, hepatoprotective, and anti-inflammatory have been reported from the different species of Cirsium (Table 1).

9.1. Antimicrobial Activity

A huge number of studies have been carried out on various species of Cirsium, which proves that it possesses a great number of antimicrobial properties (Table 3). Nazaruk and Jakoniuk [50] used the flowers and leaves of Cirsium rivulare to test the microbicidal potential against bacterial strains namely Klebsiella pneumoniae, Bacillus subtilis, Pseudomonas aeruginosa, Escherichia coli, Micrococcus and Staphylococcus aureus and fungus Candida albicans. It was found that all Cirsium rivulare extracts exerted antiproliferative and bactericidal activity. However, water extract from leaves of Cirsium was observed to be more active against Gram-positive bacteria [50]. The antibacterial activity of five compounds, namely Tracin, 9,12,15-Octadecatrienoic acid, Luteolin, Hispidulin, and α-Tocopherol isolated from the Cirsium arvense, was tested against different bacterial strains. It was observed that Tracin, Luteolin, and Hispidulin exhibited marked protective efficacy against bacterial strains, whereas α-tocopherol showed moderate antibacterial activity. However, 9,12,15-Octadecatrienoic acid showed low bactericidal activity. In addition to it, the antifungal activity of these compounds was also monitored against six pathogenic fungi, Trichophyton longifusus, Microsporum canis, Fusarium solani, Candida glabrata, Candida albicans, and Aspergillus flavus. It was shown that all the phytocompounds had a moderate to low antifungal efficacy, except 9,12,15-Octadecatrienoic acid, which has no antifungal impact [35]. In another study, flowers and leaf extracts from C. vulgare, C. rivulare, C. palustre, C. oleraceum, or C. arvense have been shown to exhibit a significant antimicrobial effect against P. aeruginosa, S. aureus, and B. subtilis. Flower extract of Cirsium species showed a more potent antimicrobial effect as compared to the leaf extract [87]. Moreover, flavonoids present in C. oleraceum also displayed substantial antibacterial and antifungal effects [88]. Nazaruk et al. [89] investigated the antibacterial effects of Cirsium species against bacterial strains such as S. aureus, P. aeruginosa, B. subtilis, and the fungal strain C. albicans. Among the Cirsium species studied, C. palustre had the strongest antibacterial activity [39]. However, the protective potential of bioactive components extracted from C. canum was determined against the Gram-negative and positive bacteria. It was noticed that the extract and fractions of C. canum showed no effect on the growth of Gram negative bacteria. However, it showed potent inhibitory activity against the microbes S. pneumonia, S. aureus, S. epidermidis, B. subtilis, and B. cereus [63]. Moreover, Strawa et al. [90] found that hexane and defatted methanol extracts of Cirsium roots have a strong bactericidal effect against Gram-positive and Gram-negative bacteria with MIC and MBC values ranging from 25 to 200 μg/mL. In another investigation, the microbicidal effect of crude extract and fractions of C. scabrum was determined against twenty-two Gram (+) and thirteen Gram (−) microbial strains. Ethyl acetate and Butanol fraction of C. scabrum showed profound effect against the S. aureus and D. hominis strains [47]. Flavonoids isolated from C. japonicum demonstrated mild antibacterial activity against two strains of human skin bacilli and six strains of S. aureus [10]. Ciryneol D, isolated from the C. setosum, inhibited the development of mycelium in a variety of fungi [91].

9.2. Antioxidant Activity

Various in vitro investigations indicated that the roots, leaves, and flowers of C. arvense had a robust antioxidant effect (Table 3) [46]. Hossain et al. (2016) examined ethanolic extract of C. arvense for antioxidant activity. It was observed that ethanolic extract of C. arvense showed marked antioxidant activity [52]. The antioxidant potential of various extracts from C. vulgare, C. rivulare, C. palustre, C. oleraceum, and C. arvense was studied by Nazaruk [39], and the total antioxidant activity was found to be in the range of 0.98 to 2.71 m/mL [39]. In another investigation, crude aqueous extracts of the Cirsium species were monitored for total antioxidant potential through the ABTS technique. It was observed that C. vulgare (2.31 m/mL), C. rivulare (2.78 m/mL), C. palustre (2.78 m/mL), C. oleraceum (2.76 m/mL), and C. arvense (2.74 m/mL) showed the remarkable antioxidant activity [89]. In another study, Lee et al. [89] used the DPPH free radical test to determine the antioxidant capacity of leaves and root extracts of C. setidens. The IC50 values for the butanolic fraction of leaves and roots were observed to be 33.53 g/mL and 9.75 g/mL, respectively. The free radical scavenging potential of C. setidens was found to be more than the tocopherol and ascorbic acid [89]. Moreover, it was observed that the methanolic extract of C. rivulare roots exhibited more DPPH scavenging activity in contrast to the hexane extract [90]. Balanophonin, Vanillic acid, and Kaempferol-3-O-L-rhamnopyranoside purified from the C. sipyleum, C. eriophorum, and C. leucopsis showed huge antioxidant activity [95]. In another study, it was found that hexane extracts of Cirsium species contain the active component Linoleic acid, which displayed a remarkable antioxidant activity [90]. Chlorogenic acid was identified as the potent antioxidant compound among the different species of Cirsium in Poland [90]. In another investigation, the major antioxidant components in Cirsium japonicum were found to be Luteolin and Silibinin [97]. Moreover, Luteolin, apigenin, and their glucosides present in different species of Cirsium also showed antioxidant and hepatoprotective effects [98]. In addition to it, major flavonoids Pectolinarin and Pectolinarigenin present in Cirsium exhibited strong antioxidant activity [58]. In another study, Apigenin, Diosmetin, and Silicristin were found to be present in significant amounts in different species of Cirsium and also displayed potent DPPH radical-scavenging properties [99].

9.3. Antiproliferative Activity

The antiproliferative potential of methanolic and chloroform extracts from different parts of C. arvense was tested against C6 cells (Rat brain tumor cells), Hela cells (Mammalian uterine carcinoma), and Vero cells (African green monkey renal cells). The root extracts of C. arvense revealed maximum inhibitory activity against the proliferation of C6 cells. However, the stem extracts showed huge inhibition against the Vero and HeLa cell lines. Moreover, the phytoconstituents isolated from the C. arvense showed antiproliferative activity in the order: Arvense 1 < Stigmasterol < Linarin < Daucosterin < 5-FU [46]. In addition to it, Tocospiro C, Tocospiro A, and Tocospiro B isolated from the leaves and stem of C. setosum showed inhibitory action against mammalian stomach cancer, ovarian cancer, lung adenocarcinoma (A549), hepatoma (Bel7402), and colon cancer (HCT-8) cells. Tocospiro C and B exerted maximum selective inhibition against the mammalian colon cancer cells, and IC50 was observed to be 0.03 µM and 0.12 µM, respectively (Table 4). However, Tocospiro A revealed very low inhibitory activity against HCT-8 cells, and IC50 was found to be 12.8 µM. All the compounds showed similar inhibition against the other cancer cell lines and displayed IC50 > 20 µM [70]. In another study, it was observed that aerial parts of C. setidens contain major phytoconstituents Tocopherol, 24-Hydroperoxycycloart-25-en-3β-ol, trans-Phytol, 9, 12, 15-Octadecatrienoic acid, Hexadecanoic acid, and Sitosterol. It was observed that maximum cytotoxicity against the mammalian cancer cell lines was exhibited by the 24-Hydroperoxycycloart-25-en-3β-ol, and ED50 was observed to be in the range from 2.66 to 11.25 µM. The remaining phytoconstituents showed negligible cytotoxic action on the cancer cell lines [23]. Similarly, the cytotoxic potential of leaf extracts of C. scabrum was monitored against the mammalian macrophage cell line. It has been noticed that methanolic, and petroleum ether fractions of C. scabrum displayed selective cytotoxic activity with the IC50 value of 11.53 and 12.12 μg/mL, respectively [47].
In another investigation, the antiproliferative activity of essential oil purified from C. palustre and C. rivulare was determined against the adenocarcinoma cell line. It revealed moderate inhibitory action against the adenocarcinoma with an IC50 value of 110–140 g/mL [73]. Likewise, extracts of C. palustre and C. arvense showed a little cytotoxic effect on the normal skin fibroblasts in a dose-dependent manner [87].

9.4. Analgesic and Anti-Inflammatory Activity

The water extracts from the aerial portion of C. subcoriaceum and its active component pectolinarin were monitored for analgesic and anti-inflammatory activity (Table 4). It was observed that crude extract of C. subcoriaceum and pectolinarin hampered the acid-induced writhing in mice in a concentration dependent manner. The pectolinarin (ED50 28.44 mg/kg) was observed to be more effective as an analgesic as compared to the crude extract of C. subcoriaceum (ED50 83.18 mg/kg). It has been observed that Pectolinarin showed similar protective efficacy as a standard analgesic compound, Acetylsalicylic, at a similar concentration. The water extract of C. subcoriaceum and pectolinarin hindered the edema induced by carrageenan. The ED30 of C. subcoriaceum and pectolinarin was detected to be 25 mg/kg and 3.7 mg/kg, respectively [58]. Similarly, Pectolinarigenin and Pectolinarin extracted from the aerial parts of C. chanroenicum hindered cyclooxygenase-2 mediated prostaglandin E2 synthesis and leukotrienes in LPS-treated macrophages, which resulted in lesser production of eicosanoid. Moreover, oral administration of Pectolinarigenin and Pectolinarin declined inflammation and allergy in the animal control group. Thus, these components might play some part in the anti-inflammatory properties of crude extract of C. chanroenicum. COX inhibitors are utilized as anti-inflammatory drugs, while 5-lipoxygenase inhibitors have anti-allergic action [59]. In another research, the crude extract and its flavonoid Cirsimaritin caused the marked inhibition of nitric oxide and nitric oxide synthase expression in macrophage cells. It repressed the production of tumor necrosis factor-α, interleukin-6, and NO in macrophage cells induced by lipopolysaccharide. Moreover, pretreatment with Cirsimaritin caused the reduction of phosphorylation of IκBα and Akt in LPS-induced macrophage cells. Cirsimaritin decreased the induction of FOS and STAT3 (signal transducer and activator of transcription 3) signaling in macrophages which depicts its anti-inflammatory nature [102]. Likewise, Silibinin purified from C. japonicum decreased the growth of human mast cells and caused a reduction in the expression of pro-inflammatory cytokines. Furthermore, Silibinin declined the phosphorylation of IκBα and NF-κB transcriptional activity in stimulated mast cells. Therefore, it can be employed for the cure of mast cells mediated allergic inflammatory ailments [103].

9.5. Hepatoprotective Activity

The hepatoprotective activity of water extract of roots and leaves of C. arisanense was monitored in mammalian hepatocellular carcinoma cell lines and mice (Table 5). Roots and leaves of C. arisanense shielded the hepatocellular carcinoma cells against tacrine-stimulated hepatotoxicity and decreased the expression of hepatitis B surface antigen. Furthermore, roots and leaves of C. arisanense ameliorated the hepatic damage in mice induced due to tacrine as they lowered the concentration of serum glutamate-pyruvate transaminase (SGPT) and serum glutamic oxaloacetic transaminase (SGOT). These effects of the roots of C. arisanense might be due to an enhancement in the concentration of liver glutathione [42]. Likewise, the administration of butanol extract of C. setidens at 500 mg/kg caused a reduction in the hepatic damage in rats induced due to CCl4. Treatment of C. setidens elevated the concentration of antioxidant markers glutathione peroxidase, glutathione peroxidase, and superoxide dismutase (SOD) in the liver of rats. Histological studies substantiated the biochemical analysis, indicating that the extract of C. setidens caused a remarkable decrease in hepatic ballooning degeneration [21]. Moreover, the active components Pectolinarin and Pectolinarigenin present in methanolic extracts of C. setidens leaves decreased the hepatic derangement in rats induced due to galactosamine toxicity. The concentration of SGOT, SGPT, alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) were observed to be decreased after the treatment of Pectolinarin and Pectolinarigenin, indicating the hepatoprotective potential of both of the components. Treatment with Pectolinarin and Pectolinarigenin boosted the antioxidant enzymes glutathione, glutathione transferase, glutamylcysteine synthetase, glutathione reductase, and SOD. It revealed that the hepatoprotective activity of Pectolinarin and Pectolinarigenin is attributed to the induction of the antioxidant system [60]. In another investigation, the crude extracts of Cirsium normalized the levels of SGOT and SGPT in mice that had been raised by CCl4 injection. Cirsium japonicum and Cirsii herba decreased the CCl4 stimulated liver necrosis and restored the levels of hepatic antioxidant enzymes and malondialdehyde [104].

9.6. Immunomodulatory Activity

C. japonicum and its phytoconstituent Pectolinarin hindered the growth of tumors in mice and enhanced the humoral and cellular immune responses (Table 5). It boosted the complement pathway in the tumor-bearing mice and also caused the improvement in the transformation of spleen cells as well as natural killer cell activity [57].

9.7. Anticancer Activity

There are numerous studies indicating the anticancerous nature of Cirsium (Table 4) and its promising therapeutic ability to prevent malignancies. The Pectolinarin and 5,7-Dihydroxy-6,4-dimethoxyflavone obtained from C. japonicum were monitored for anticancer activity in the mouse. It was observed that both of the compounds exhibited a remarkable reduction in the multiplication of tumor cells in a concentration-dependent manner [53]. Similarly, the extract of Cirsium japonicum and Cirsimaritin caused a reduction in the breast cancer cells indicated by the suppression of expression of Akt, VEGF, and ERK in MDA-MB-231 cells. Furthermore, C. japonicum extract exhibited an antiproliferative effect in MCF-7 cancer cells by restricting the cell cycle in the G1 phase and also triggered cell death by influencing mitochondrial apoptotic pathways [100]. Triterpenes present in Cirsium setosum revealed moderate cytotoxicity against human colon and ovarian cancer cells. [101], while 3β-Hydroxy-22-oxo-20-dandelion-30-oleic acid displayed a potent selective inhibition on the ovarian tumor cell line A2780 [110]. Silybin, derived from Cirsium japonicum, hindered gastric cancer cells by reducing the production of cell cycle proteins and blocking gastric tumor cells in the G2/M phase, which ultimately resulted in apoptosis (Figure 12) [111].

9.8. Oviposition Stimulatory Activity

Treatment of roots essential oil of C. japonicum induced the oviposition in Ostrinia zealis (Table 5). It was observed that root essential oil of plants contains majorly Aplotaxene, which might serve as a stimulator of oviposition [41]. In another investigation, treatment of C. japonicum extract to ovariectomized rats caused a marked decline in cholesterol, body weight, and triglyceride, as well as remarkable enhancement in estradiol and bone mineral density. Moreover, Molecular docking studies revealed that the active phytoconstituents of C. japonicum have a binding affinity with the ligand-binding sites of the estrogen receptor. It indicated the potential of C. japonicum extract in the relief of symptoms of pre-menopause and post-menopause [100].

9.9. Anti-Anxiety Effect

The ethanolic extract of Cirsium japonicum was screened for the anti-anxiety effect in mice. The extract of C. japonicum showed no effect on the movement of mice in the open-field test. However, it increased the exploration in the undefended center zone. Administration of plant extract also enhanced the duration of mice in the elevated plus-maze test, which pointed out that the Cirsium japonicum exhibited anti-anxiety properties. It was observed that C. japonicum’s anti-anxiety effects were equivalent to those of the benzodiazepine. Further, the anti-anxiety effect of C. japonicum was confirmed by examining its effect on human neuroblastoma cells. Treatment of C. japonicum upregulated the influx of chloride ions in neuroblastoma cells in a concentration-dependent manner, which was reduced by coadministration of bicuculline [112]. In another investigation, C. japonicum had an antidepressant-like impact on mice in the forced-swimming test by dramatically lowering immobility. It was observed that administration of C. japonicum did not boost the locomotor activity in the open field test. Moreover, it enhanced the influx of Cl- ions without affecting the uptake of monoamine in mammalian neuroblastoma cells. Only Luteolin, one of the active components of the C. japonicum extract, revealed similar antidepressant-like effects. Therefore, the antidepressant-like action of C. japonicum extract was believed to be due to the presence of luteolin in it [113].

9.10. Nephroprotective

Cirsium is nephroprotective in nature, as it shields the kidney from various toxins (Table 5). Pectolinarin and flavones purified from Cirsium japonicum were studied for their nephroprotective activity in diabetic rats induced by streptozotocin followed by a high-carbohydrate/high-fat diet. Both flavones exhibited antidiabetic activity in rats. However, a combination of Pectolinarin and 5,7-Dihydroxy-6,4’-dimethoxy flavone in diabetic rats was found to be more potent in the reduction of Cholesterol, Glucose, and Triglycerides. Treatment of flavones in diabetic rats caused the reversal of abnormal levels of glucose metabolism-related enzymes. It increased the concentration of adiponectin in diabetic rats, but there was no discernible impact of the flavones on the abnormal concentration of insulin or leptin and glucose transporter 4 [108]. Moreover, Cirsium japonicum flavones improved adipocyte development by enhancing the levels of PPARγ. It enhanced the insulin-induced glucose intake in adipocytes, which is probably due to the more levels of adiponectin and GLUT4 [109]. In another study, the glucosidase enzyme is inhibited by taraxastane-type triterpenoids derived from Cirsium setosum [110]. Additionally, the leaves of Cirsium maackii and its flavonoids showed an antidiabetic effect by hampering the production of glycation end products [114].

9.11. Other Therapeutic Effects

Treatment with C. japonicum (50–100 mg/kg/day) enhanced cognitive skills by diminishing the oxidative stress in amyloid β-peptide-induced mice, and it can be utilized as a potent agent for the cure of Alzheimer’s disease (Figure 13). In addition to it, many investigators have examined the hemostatic action and mechanism of different Cirsium species. Administration of Cirsium setosum extract exhibited a substantial activity on hemostasis, blood coagulation, and hemorrhage in rats [115,116]. Wang (2018) also reported the hemostatic action of nano-scale constituents in Cirsium charcoal [117]. Moreover, Shikokiol A obtained from C. nipponicum roots was tested in vitro for enzyme inhibitory activities. It was observed to be a potent inhibitor of the non-heme iron-containing enzyme, i.e., lipoxygenase, in guinea pig tracheal contraction [118,119].
The C. sipyleum, C. leucopsis, and C. eriophorum exhibited 16–57% inhibition against acetyl- and butyryl-cholinesterase activity [21,105]. Similarly, Lee et al. [54] showed that ethanolic fraction and flavonoids of C. japonicum displayed aldose reductase inhibition activity with IC50 values of 0.21 μg/mL and 0.77 μM, respectively [54]. In another study, Cirsium japonicum and its constituent apigenin caused a marked reduction in the expression of Hif-2α and decreased the levels of metalloproteinases and cyclooxygenases in chondrocytes. This study depicted the potential of Cirsium japonicum and its constituents for the development of therapeutics for hindering osteoarthritis [104]. The extracts of C. japonicum hindered the adipogenesis in adipocytes by diminishing the concentration of triglycerol. The chloroform fraction of C. japonicum was observed to reveal the maximum inhibition of adipocyte differentiation. The extract downregulated the expression of lipoprotein lipase, PPARγ, adiponectin, and fatty acid synthetase intricated in adipogenesis. Therefore, C. japonicum extract can be used as an ideal candidate for the treatment of obesity [120].

10. Herbal Formulations and Clinical Trials

MS-10, a formulation of Cirsium and Thyme extract, was examined on 71 premenopausal women for 28 and 84 days in a randomized, double-blind clinical trial. Treatment of MS-10 significantly reduced the onset of menopause by 48%. Moreover, MS-10 enhanced the insulin-like growth factor-1 (IGF-1) and estrogen, which might show protective inhibitory effects on menopause and aging in women. MS-10 also promoted bone health in women by increasing bone formation and absorption indicators such as osteocalcin, alkaline phosphatase, collagen, and N-telopeptides of type I collagen. It was observed that MS-10 therapy improved the levels of cortisol and upgraded the psychological well-being index in women [121]. Another herbal formulation, Fufang Zhenzhu Tiaozhi (FTZ), contains Ligustrum lucidum, Citrus medica, Coptis chinensis, Atractylodes macrocephala, Panax, Cirsium japonicum, Salvia miltiorrhiza, and Eucommia ulmoides [122]. FTZ has been employed clinically for the cure of hyperlipidemia, diabetes, osteoporosis, and atherosclerosis [123,124]. Zhang et al. [125] reported that the FTZ treatment lessened cardiac hypertrophy in mice through the downregulation of expression of miR-214 and upregulation of SIRT3 expression. Several studies indicated the therapeutic potential of FTZ is linked to a variety of pharmacotherapeutic activities [126,127]. Moreover, various human and animal studies demonstrated that FTZ has an excellent ability to reduce total cholesterol, triglyceride in the blood. This formulation also exhibited protective action against metabolic diseases such as atherosclerosis, hyperlipidemia, and hepatic infections by modifying the concentration of glucose and lipids in the blood [128,129]. Diao et al. (129) demonstrated that FTZ decreased atherosclerosis by hindering endothelial–mesenchymal transition through the β-catenin pathway. Treatment of FTZ bettered dyslipidemia and dysfunctioning of endothelial cells in the atherosclerotic mice. Moreover, FTZ administration caused the reduction of total bad cholesterol and boosted HDL. It also enhanced the levels of endothelial markers such as CD31 and cadherin and diminished the mesenchymal markers, signifying that it impeded the endothelial–mesenchymal transition.
In another study, the protective efficacy of FTZ was determined in a mouse model of Polycystic ovary syndrome (PCOS). It was found that FTZ remarkably increased the levels of adiponectin, thus modifying adipose-ovary crosstalk to decline PCOS. Moreover, the administration of FTZ decreased the disruption of the estrous cycle, cystic follicles, and insulin resistance [96]. In another investigation, Yang et al. [130] described that FTZ repressed renal inflammation and fibrosis by the suppression of NF-κB and IL-17. Administration of FTZ also reduced the levels of urea, glucose, triglycerides, cholesterol, fibronectin, and collagen. Similarly, a multicenter, randomized, double-blind trial also reported the protective potential of FTZ on diabetic coronary heart disease [131]. In another clinical study, flower extract of Cirsium japonicum improved the wrinkles and elasticity of the skin, and it can be preferred as an active component of antiaging cosmetics [132].

11. Patents

A formulation comprising Cirsium japonicum extract as a potent agent for the induction of melanogenesis: The melanogenesis-stimulating formulation includes Cirsium japonicum extract as an active component, and this composition can be safely utilized for the prevention and treatment of vitiligo, white hair, or hypopigmentation [133].
A process for the cure of fatty liver: A method was designed for the preparation of hepatoprotective formulation from the extract of Cirsium (Table 6) [134].
A method for the enhancement of Lipolysis: The extracts of Cirsium showed huge lipolysis when orally administrated or dermatologically apply through local administration. It pointed out the potential of Cirsium in the control and treatment of obesity [135].
Anti-acne formulation: A composition was prepared from Quercus robur, Sesamum indicum, Houttuynia cordata, Cirsium japonicum, and Thuja orientalis as potent agents for the cure of acne. This composition revealed maximum antibacterial activity against C. acnes [136].
Enhancer and moisturizer of Ceramide production: Composition containing Cirsium japonicum singly or in combination with Chenopodium hybridum, Melia toosendan, Indigofera tinctoria, Catalpa ovata, and Tagetes erecta showed an excellent capacity for the enhancement of ceramide [137].

12. Pharmacokinetics

Pharmacokinetics serves a critical role in preclinical drug development, screening of toxicity of the drug, and optimization of the concentration of the drug. It is measured as an effective way of detecting the potential active constituents and explaining the mode of action of plants or drug formulations. So far, there are very few scientific investigations in the literature on the pharmacokinetic behavior of Cirsium. A liquid chromatography–mass spectrometry (LC-MS) technique was employed to monitor the different flavonoids of C. setosum in rat plasma. It was observed that Rutin, Acacetin, Naringin, Wogonin, and Quercetin were the long-acting constituents of the C. setosum, with more elimination time and bioavailability [138].
Similarly, Zhang et al. [139] detected twenty-seven flavonoids in the blood, bile, and urine of rats after the administration of Cirsium japonicum through the UPLC-MS. In another study, after treatment with the extract of Cirsium japonicum in rats, the maximum concentration of Linarin, Pectolinarigenin, Hispidulin, Pectolinarin, Diosmetin, Acacetin, and Apigenin in plasma was observed to be 86 ng/mL, 6 ng/mL, 32 ng/mL, 876 ng/mL, 37 ng/mL, 19 ng/mL and 148 ng/mL, respectively. Pectolinarin, Linarin, Pectolinarigenin, Hispidulin, Diosmetin, and Acacetin were absorbed rapidly and reached their maximum concentration in plasma in five minutes (Table 7). However, Apigenin was absorbed slowly and reached its maximum concentration after 360 min of its administration [140].

13. Toxicology

A systemic safety study of Cirsium extract is required for the expansion of novel pharmaceuticals or drugs. Oral administration of C. setidens extract in rats for 28 days at the dose of 1.25, 2.5, and 5 g/kg body weight did not exhibit significant toxicological alterations such as mortality, hematology, and biochemical parameters. Treatment of C. setidens extract revealed normal histological architecture of the liver, heart, spleen, and kidney, which indicated the wide safety index of the plant extract [141]. Similarly, treatment with extract of C. japonicum at the concentration of 2 g/kg body weight for 15 days revealed no marked toxic effects and mutagenicity [142].

14. Nanoformulations of Cirsium

Shin et al. [143] examined the C. setidens-derived selenium nanoformulations for their protective potential against oxidative stress. The nanoparticles of C. setidens extract unveiled zeta potential of −27.4 mV with a particle size of 117.8 nm. It was noticed that nanoformulation of C. setidens extract displayed more antioxidant and antibacterial activities as compared to the alone C. setidens extract. In addition to it, nanoformulation of C. setidens was observed to be harmless to normal fibroblast cell lines. However, they showed marked cytotoxic effects against A549 mammalian lung cancer cells through rupturing the mitochondrial membrane and nucleus [143]. Moreover, C. arvense-derived silver nanoparticles showed robust protective activity against Escherichia coli [144,145]. In another investigation, Cirsium vulgare-derived cobalt oxide nanoparticles augmented the electrocatalytic action for the monitoring of cysteine [146]. Moreover, C. japonicum extract served as a reducing and stabilizing agent for the synthesis of nontoxic silver nanoparticles. These silver nanoparticles showed 98% degradation of bromo phenyl blue in twelve minutes, which indicated the robust reductive potential of silver nanoparticles in the water cleansing and altering some organic harmful compounds to harmless components. In another investigation, Cirsium arvense derived copper nanoparticles (CA-CuNP) were examined for antibacterial and photocatalytic properties. CA-CuNP showed the photocatalytic potential and caused the complete degradation of Rhodamine B in thirty minutes. CA-CuNP exhibited microbicidal potential by hindering the growth rate of S. aureus and E. coli and displayed zone of inhibition 18 mm and 21 mm respectively [147,148]. In addition to it, water extract of Cirsium setosum carbonisata (CSC) was used for the synthesis of carbon dots (CD), which were spherical and even in size and unveiled a little noxiousness against macrophage cells. Moreover, tail and liver bleeding experiments showed a lower bleeding time in CSC-CD-treated mice in contrast to normal saline-treated mice. It was noticed that CSC-CD can enhance the extrinsic blood coagulation pathway and stimulate the fibrinogen proteins, which are projected towards the hemostatic effect of CSC-CD [149].

15. Conclusions and Future Perspectives

The Cirsium genus has been given a lot of attention due to its widespread usage in traditional medicine. The chemical compounds extracted from the Cirsium genus include mostly flavonoids, phenylpropanoids, and triterpenoids, which contribute to the multiple medicinal properties. From a phytochemical standpoint, a wide range of chemical structures have been identified from Cirsium species of varying distribution, which can result in a variety of biological effects. The current review suggests a wide range of potential applications of Cirsium in the field of pharmaceuticals, cosmetics, and health foods. Pharmacological studies revealed that Cirsium species have several biological activities, such as hepatoprotective, antibacterial, antioxidant, antitumor, and anti-inflammatory, which tends to untangle the various traditional applications of the Cirsium genus. The phytochemical studies of different Cirsium species and their renowned pharmacological activities could be exploited for pharmaceutic product development in the future. Furthermore, studies are required on less known Cirsium species, particularly on the elucidation of the mode of action of their biological activities. Further investigations should be carried out on pharmacodynamics, pharmacokinetics, and quality control system for Cirsium, which is critical for broadening its medicinal potential in the future. Moreover, the toxic and safe profile of Cirsium has not been well investigated; thus, further research is needed in this domain.

Author Contributions

G.A.: Formal analysis, writing—original draft, and data curation. G.K.: Writing—original draft. A.B.: Writing—original draft. H.S.S.: Chemical structures and software. G.B.: Methodology, software, and editing. G.A.N.: Writing—review and editing. V.M.: Writing—original draft—nanotechnology. A.S.: Conceptualization, methodology, supervision, validation, review, and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABTS2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)
ALTAlanine aminotransferase
ARIAldose reductase inhibition
ASTAspartate aminotransferase
Bel7402Human hepatocellular carcinoma
CmaxMaximum plasma concentration
CCl4Carbon tetrachloride
COX-2Cyclooxygenase-2
DPPH2,2-diphenyl-1-picryl-hydrazyl-hydrate
ED50Median effective dose
5-FUFluorouracil
GABAGamma-aminobutyric acid
G1Growth 1 phase
G2/MGrowth 2 phase
HCT-8Human colon cancer cell line
HeLa cellHenrietta Lacks cell
Hif-2αHypoxia-Inducible Factor-2α
HPLC-MSHigh-performance liquid chromatography–mass spectrometry
IC50Half-maximal inhibitory concentration
IL-6Interleukin-6
MBCMinimum bacterial concentration
MCF-7Human breast cancer cell line
MDA-MB-231Human mammary carcinoma
MICMinimum inhibitory concentration
MMP3Matrix metalloproteinase-3
MMP13Matrix metalloproteinase-13
NF-κBNuclear factor kappa B
NONitric oxide
PPARγPeroxisome proliferator-activated receptor gamma
p-AktProtein kinase B
p-ERKExtracellular signal-regulated kinase
SODSuperoxide dismutase
VEGFVascular endothelial growth factor
CA-CuNPCirsium arvense-derived copper nanoparticles
CSCCirsium setosum Carbonisata
CDCarbon dots

References

  1. Asghar, M.; Younas, M.; Arshad, B.; Zaman, W.; Ayaz, A.; Rasheed, S.; Shah, A.H.; Ullah, F.; Saqib, S. Bioactive potential of cultivated Mentha arvensis l for preservation and production of health-oriented food. J. Anim. Plant. Sci. 2022, 32, 835–844. [Google Scholar]
  2. Cláudio, A.F.M.; Cognigni, A.; de Faria, E.L.; Silvestre, A.J.; Zirbs, R.; Freire, M.G.; Bica, K. Valorization of olive tree leaves: Extraction of oleanolic acid using aqueous solutions of surface-active ionic liquids. Sep. Purif. Technol. 2018, 204, 30–37. [Google Scholar] [CrossRef]
  3. Salehi, B.; Kumar, N.V.A.; Şener, B.; Sharifi-Rad, M.; Kılıç, M.; Mahady, G.B.; Vlaisavljevic, S.; Iriti, M.; Kobarfard, F.; Setzer, W.N.; et al. Medicinal Plants Used in the Treatment of Human Immunodeficiency Virus. Int. J. Mol. Sci. 2018, 19, 1459. [Google Scholar] [CrossRef]
  4. Marmitt, D.J.; Shahrajabian, M.H. Plant species used in Brazil and Asia regions with toxic properties. Phytotherapy Res. 2021, 35, 4703–4726. [Google Scholar] [CrossRef]
  5. Plants of the World Online: Cirsium Mill. Available online: http://www.plantsoftheworldonline.org/taxon/urn:lsid:ipni.org:names:30001899-2 (accessed on 8 August 2022).
  6. Bureš, P.; Wang, Y.-F.; Horová, L.; Suda, J. Genome Size Variation in Central European Species of Cirsium (Compositae) and their Natural Hybrids. Ann. Bot. 2004, 94, 353–363. [Google Scholar] [CrossRef]
  7. Kadota, Y. Species Diversification of Genus Cirsium (Asteraceae) in Japan. Korean J. Plant Taxon. 2007, 37, 335–349. [Google Scholar] [CrossRef]
  8. Ashmita, P.; Singh, L.; Kumar, D.; Antil, R.; Dahiya, P. Cirsium arvense: A Multi-potent Weed. Ann. Biol. 2020, 36, 442–447. [Google Scholar]
  9. Shahrajabian, M.H. Spear Thistle (Cirsium Vulgare L.) And Ramsons (Allium Ursinum L.), Impressive Health Benefits and High-Nutrient Medicinal Plants. Pharmacogn. Commun. 2021, 11, 168–171. [Google Scholar] [CrossRef]
  10. Luo, W.; Wu, B.; Tang, L.; Li, G.; Chen, H.; Yin, X. Recent research progress of Cirsium medicinal plants in China. J. Ethnopharmacol. 2021, 280, 114475. [Google Scholar] [CrossRef]
  11. Moore, R.J. The Thistles of Canada. Ottawa, Canada. 1974. Available online: https://archive.org/details/thistlesofcanada00moor/page/19/mode/1up?ref=ol&view=theater&q=cirsium (accessed on 24 August 2021).
  12. Mabberley, D.J. Mabberley’s Plant-Book: A Portable Dictionary of Plants, Their Classifications and Uses, 3rd ed.; Cambridge University Press: Cambridge, UK, 2008. [Google Scholar]
  13. Karthikeyan, S.M.; Sanjappa, S.; Moorthy, S. Flowering Plants of India. In Dicotyledons; BSI: Kolkata, India, 2009. [Google Scholar]
  14. Bohm, B.A.; Stuessy, T.F. Flavonoids of the Sunflower Family (Asteraceae); Springer: Wien, Austria, 2001. [Google Scholar]
  15. Venner, L. Common Name: Bull Thistle, (Common Thistle, Spear Thistle, Scotch Thistle, Black Thistle, Plume Thistle). Available online: https://azkurs.org/common-name-bull-thistle-common-thistle-spear-thistle-scotch-t.html (accessed on 22 August 2021).
  16. Jordon-Thaden, I.E.; Louda, S.M. Chemistry of Cirsium and Carduus: A role in ecological risk assessment for biological control of weeds? Biochem. Syst. Ecol. 2003, 31, 1353–1396. [Google Scholar]
  17. Formisano, C.; Rigano, D.; Senatore, P.F.; De Feo, V.; Bruno, M.; Rosselli, S. Composition and allelopathic effect of essential oils of two thistles: Cirsium creticum (Lam.) D.’Urv. ssp. triumfetti (Lacaita) Werner and Carduus nutans L. J. Plant. Interact. 2007, 2, 115–120. [Google Scholar] [CrossRef]
  18. Chon, S.-U.; Kim, Y.-M.; Lee, J.-C. Herbicidal potential and quantification of causative allelochemicals from several Compositae weeds. Weed Res. 2003, 43, 444–450. [Google Scholar] [CrossRef]
  19. van der Kooi, C.J.; Pen, I.; Staal, M.; Stavenga, D.G.; Elzenga, J.T.M. Competition for pollinators and intra-communal spectral dissimilarity of flowers. Plant Biol. 2015, 18, 56–62. [Google Scholar] [CrossRef]
  20. Parus, A.; Grys, A. Cirsium oleraceum (L.) Scop-active substances and possible usage. Postapy. Fitoter. 2011, 2011, 100–105. [Google Scholar]
  21. Boğa, M.; Yılmaz, P.K.; Cebe, D.B.; Fatima, M.; Siddiqui, B.S.; Kolak, U. Chemical Constituents and Biological Activities of Cirsium leucopsis, C. sipyleum, and C. eriophorum. Z. Für Nat. C 2014, 69, 381–390. [Google Scholar] [CrossRef]
  22. Chakraborty, T.; Saha, S.; Bisht, N.S. First Report on the Ethnopharmacological Uses of Medicinal Plants by Monpa Tribe from the Zemithang Region of Arunachal Pradesh, Eastern Himalayas, India. Plants 2017, 6, 13. [Google Scholar] [CrossRef]
  23. Bin Lee, W.; Kwon, H.C.; Cho, O.R.; Lee, K.C.; Choi, S.U.; Baek, N.I.; Lee, K.R. Phytochemical constituents of Cirsium setidens Nakai and their cytotoxicity against human cancer cell lines. Arch. Pharm. Res. 2002, 25, 628–635. [Google Scholar]
  24. Loizzo, M.R.; Statti, G.A.; Tundis, R.; Conforti, F.; Ando’, S.; Menichini, F. Antimicrobial activity and cytotoxicity of Cirsium tenoreanum. Fitoterapia 2004, 75, 577–580. [Google Scholar] [CrossRef]
  25. Ishida, H.; Umino, T.; Tsuji, K.; Kosuge, T. Studies on antihemorrhagic substances in herbs classified as hemostatics in Chinese medicine. VII On the antihemorrhagic principle in Cirsium japonicum DC. Chem. Pharm. Bull. 1987, 35, 861–864. [Google Scholar] [CrossRef]
  26. Cheriet, T.; Ben-Bachir, B.; Thamri, O.; Seghiri, R.; Mancini, I. Isolation and Biological Properties of the Natural Flavonoids Pectolinarin and Pectolinarigenin—A Review. Antibiotics 2020, 9, 417. [Google Scholar] [CrossRef]
  27. He, X.-F.; He, Z.-W.; Jin, X.-J.; Pang, X.-Y.; Gao, J.-G.; Yao, X.-J.; Zhu, Y. Caryolane-type sesquiterpenes from Cirsium souliei. Phytochem. Lett. 2014, 10, 80–85. [Google Scholar] [CrossRef]
  28. Walesiuk, A.; Nazaruk, J.; Braszko, J.J. Pro-cognitive effects of Cirsium rivulare extracts in rats. J. Ethnopharmacol. 2010, 129, 261–266. [Google Scholar] [CrossRef] [PubMed]
  29. Dang, S. Plants Profile for Cirsium vulgare, USDA Plant Database 1984. Available online: https://plants.usda.gov/home/plantProfile?symbol=CIVU (accessed on 14 August 2021).
  30. Nazaruk, J.; Gudej, J. Flavoniod compounds from the flowers of Cirsium rivulare (JACQ) All. Acta. Pol. Pharm. 2003, 60, 87–89. [Google Scholar] [PubMed]
  31. Singh, A.; Lal, M.; Samant, S.S. Diversity, indigenous uses and conservation prioritization of medicinal plants in Lahaul valley, proposed Cold Desert Biosphere Reserve, India. Int. J. Biodivers. Sci. Manag. 2009, 5, 132–154. [Google Scholar] [CrossRef]
  32. Singh, B.; Borthakur, S.K. Wild medicinal plants used by tribal communities of Meghalaya. JETBD 2011, 35, 331–339. [Google Scholar]
  33. Creeping Thistle Facts and Health Benefits, Heal Benefits Times (n.d.). Available online: https://www.healthbenefitstimes.com/creeping-thistle (accessed on 2 March 2022).
  34. Guarrera, P.M. Traditional phytotherapy in Central Italy (Marche, Abruzzo, and Latium). Fitoterapia 2005, 76, 1–25. [Google Scholar] [CrossRef]
  35. Khan, H.U.Z.; Khan, S.; Chen, Y.; Wan, P. In vitro antimicrobial activity of the chemical constituents of Cirsium arvense (L). Scop. J. Med. Plants Res. 2013, 7, 1894–1898. [Google Scholar]
  36. Paluch, A. Folk herbal medicine in Poland in the nineteenth and early twentieth centuries. Pol. Tow. Ludozn. Wrocław 1989, 10, 98–99. [Google Scholar]
  37. Grzycka, K.; Krzaczek, T.; Miłkowska, J. Research on the biological activity of selected species of flower plants. Ann. Univ. Mariae Curie Sklodowska Lublin Pol. Sect. D 1978, 33, 275–283. [Google Scholar]
  38. Ożarowski, A.; Jaroniewski, W. Rośliny Lecznicze i ich Praktyczne Zastosowanie (Medicinal Plants and Their Practical Applying), Wydawniczy Związków Zawodowych, Warsaw, n.d. Available online: https://scholar.google.co.in/scholar?hl=en&as_sdt=0%2C5&q=Ożarowski+A.%2C+Jaroniewski+W.+1989.+Rośliny+lecznicze+i+ich+praktyczne+zastosowanie+%28Medicinal+plants+and+their+practical+applying%29.+Warszawa%3A+Instytut+Wydawniczy+Związków+Zawodowych&btnG= (accessed on 19 August 2021).
  39. Nazaruk, J. Antioxidant activity and total phenolic content in Cirsium five species from north–east region of Poland. Fitoterapia 2008, 79, 194–196. [Google Scholar] [CrossRef]
  40. Cho, S.; Lee, J.; Lee, Y.K.; Chung, M.J.; Kwon, K.H.; Lee, S. Determination of pectolinarin in Cirsium spp. using HPLC/UV analysis. J. Appl. Biol. Chem. 2016, 59, 107–112. [Google Scholar] [CrossRef]
  41. Chan, F.L.; Choi, H.; Chen, Z.; Chan, P.S.; Huang, Y. Induction of apoptosis in prostate cancer cell lines by a flavonoid, baicalin. Cancer Lett. 2000, 160, 219–228. [Google Scholar] [CrossRef]
  42. Ku, K.-L.; Tsai, C.-T.; Chang, W.-M.; Shen, M.-L.; Wu, C.-T.; Liao, H.-F. Hepatoprotective Effect of Cirsium arisanense Kitamura in Tacrine-Treated Hepatoma Hep 3B Cells and C57BL Mice. Am. J. Chin. Med. 2008, 36, 355–368. [Google Scholar] [CrossRef] [PubMed]
  43. Orhan, D.D.; Ergun, F.; Yesilada, E.; Tsuchiya, K.; Takaishi, Y.; Kawazoe, K. Antioxidant activity of two flavonol glycosides from Cirsium hypoleucum DC. through bioassay-guided fractionation Wound Healing View project Studies on Biological Activities of Vitis vinifera Leaves. Turkish J. Pharm. Sci. 2007, 4, 1–14. [Google Scholar]
  44. Singh, V.; Chauhan, N.S. Traditional practices of herbal medicines in the Lahaul valleys, Himachal Himalayas. Indian J. Tradit. Knowl. 2005, 4, 208–220. [Google Scholar]
  45. Lee, S.J. Korean Folk Medicine; Seoul National University Press: Seoul, Korea, 1966. [Google Scholar]
  46. Demirtas, I.; Tufekci, A.R.; Yaglioglu, A.S.; Elmastas, M. Studies on the antioxidant and antiproliferative potentials of Cirsium arvense subsp. vestitum. J. Food Biochem. 2016, 41, e12299. [Google Scholar] [CrossRef]
  47. Sahli, R.; Rivière, C.; Dufloer, C.; Beaufay, C.; Neut, C.; Bero, J.; Hennebelle, T.; Roumy, V.; Ksouri, R.; Leclercq, J.; et al. Antiproliferative and Antibacterial Activities of Cirsium scabrum from Tunisia. Evid. Based Complement. Altern. Med. 2017, 2017, 1–9. [Google Scholar] [CrossRef]
  48. Vizgirdas, R.S.; Rey-Vizgirdas, E.M. Wild Plants of the Sierra Nevada; University of Nevada Press: Reno, NV, USA, 2009. [Google Scholar]
  49. Fernández-Martínez, E.; Díaz-Espinoza, R.; Villavicencio-Nieto, M.A.; Pérez-Escandón, B.E.; Pérez-Hernández, N.; Macías, A.; Ortíz, M.I.; A Ponce-Monter, H. Preliminary phytochemical and biological study of Cirsium ehrenbergii. Proc. West. Pharmacol. Soc. 2007, 50, 162–164. [Google Scholar]
  50. Nazaruk, J.; Jakoniuk, P. Flavonoid composition and antimicrobial activity of Cirsium rivulare (Jacq.) All. flowers. J. Ethnopharmacol. 2005, 102, 208–212. [Google Scholar] [CrossRef]
  51. Khan, H.U.Z.; Ali, F.; Khan, U.S.; Ali, I. Phytochemical study on the constituents from Cirsium arvense. Mediterr. J. Chem. 2011, 2, 64–69. [Google Scholar] [CrossRef]
  52. Hossain, L.M.; Hossain, A.M.; Sadhu, K.S. HPLC Profiling and Evaluation of In-vitro Antioxidant Activity of Cirsium arvense L. (Family: Asteraceae). J. Pharmacogn. Phytochem. 2016, 5, 272–277. [Google Scholar]
  53. Liu, S.; Zhang, J.; Li, D.; Liu, W.; Luo, X.; Zhang, R.; Li, L.; Zhao, J. Anticancer activity and quantitative analysis of flavone of Cirsium japonicum DC. Nat. Prod. Res. 2007, 21, 915–922. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, J.; Rodriguez, J.P.; Lee, K.H.; Park, J.Y.; Kang, K.S.; Hahm, D.-H.; Huh, C.K.; Lee, S.C.; Lee, S. Determination of flavonoids from Cirsium japonicum var. maackii and their inhibitory activities against aldose reductase. Appl. Biol. Chem. 2017, 60, 487–496. [Google Scholar] [CrossRef]
  55. Kim, S.-J.; Kim, G.-H. Identification for Flavones in Different Parts of Cirsium japonicum. Prev. Nutr. Food Sci. 2003, 8, 330–335. [Google Scholar] [CrossRef]
  56. Malejko, J.; Nalewajko-Sieliwoniuk, E.; Nazaruk, J.; Siniło, J.; Kojło, A. Determination of the total polyphenolic content in Cirsium palustre (L.) leaves extracts with manganese(IV) chemiluminescence detection. Food Chem. 2014, 152, 155–161. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, S.; Luo, X.; Li, D.; Zhang, J.; Qiu, D.; Liu, W.; She, L.; Yang, Z. Tumor inhibition and improved immunity in mice treated with flavone from Cirsium japonicum DC. Int. Immunopharmacol. 2006, 6, 1387–1393. [Google Scholar] [CrossRef]
  58. Vázquez, M.M.; Apan, T.O.R.; Lastra, A.L.; Bye, R. A Comparative Study of the Analgesic and Anti-Inflammatory Activities of Pectolinarin Isolated from Cirsium subcoriaceum and Linarin Isolated from Buddleia cordata. Planta Med. 1998, 64, 134–137. [Google Scholar] [CrossRef]
  59. Lim, H.; Son, K.H.; Chang, H.W.; Bae, K.; Kang, S.S.; Kim, H.P. Anti-inflammatory Activity of Pectolinarigenin and Pectolinarin Isolated from Cirsium chanroenicum. Biol. Pharm. Bull. 2008, 31, 2063–2067. [Google Scholar] [CrossRef]
  60. Yoo, Y.M.; Nam, J.H.; Kim, M.Y.; Choi, J.; Park, H.J. Pectolinarin and Pectolinarigenin of Cirsium setidens Prevent the Hepatic Injury in Rats Caused by D-Galactosamine via an Antioxidant Mechanism. Biol. Pharm. Bull. 2008, 31, 760–764. [Google Scholar] [CrossRef]
  61. Syrchina, A.I.; Semenov, A.A.; Zinchenko, S.V. Chemical investigation of Cirsium setosum. Chem. Nat. Compd. 1997, 33, 212. [Google Scholar] [CrossRef]
  62. Miyaichi, Y.; Matsuura, M.; Tomimori, T. Phenolic Compound from the Roots of Cirsium japonicum DC. Nat. Med. 1995, 49, 92–94. [Google Scholar]
  63. Kozyra, M.; Biernasiuk, A.; Malm, A.; Chowaniec, M. Chemical compositions and antibacterial activity of extracts obtained from the inflorescences of Cirsium canum (L.) all. Nat. Prod. Res. 2015, 29, 2059–2063. [Google Scholar] [CrossRef] [PubMed]
  64. Ganzera, M.; Pöcher, A.; Stuppner, H. Differentiation of Cirsium japonicum and C. setosum by TLC and HPLC-MS. Phytochem. Anal 2005, 16, 205–209. [Google Scholar] [CrossRef] [PubMed]
  65. Kozyra, M.; Mardarowicz, M.; Kochmańska, J. Chemical composition and variability of the volatile components from inflorescences of Cirsium species. Nat. Prod. Res. 2015, 29, 1942–1944. [Google Scholar] [CrossRef] [PubMed]
  66. Christensen, L.P. Aplotaxene derivatives from Cirsium helenioides. Phytochemistry 1992, 31, 2039–2041. [Google Scholar] [CrossRef]
  67. Gnonlonfin, G.J.B.; Sanni, A.; Brimer, L. Review Scopoletin–A Coumarin Phytoalexin with Medicinal Properties. Crit. Rev. Plant Sci. 2012, 31, 47–56. [Google Scholar] [CrossRef]
  68. Ma, Q.; Jiang, J.-G.; Yuan, X.; Qiu, K.; Zhu, W. Comparative antitumor and anti-inflammatory effects of flavonoids, saponins, polysaccharides, essential oil, coumarin and alkaloids from Cirsium japonicum DC. Food Chem. Toxicol. 2019, 125, 422–429. [Google Scholar] [CrossRef]
  69. Kuhnau, J. Flavonoids. A class of semi-essential food components: Their role in human nutrition. J. Nutr. Diet. 1976, 24, 117–191. [Google Scholar]
  70. Yuan, Z.; Duan, H.; Xu, Y.; Wang, A.; Gan, L.; Li, J.; Liu, M.; Shang, X. α-Tocospiro C, a novel cytotoxic α-tocopheroid from Cirsium setosum. Phytochem. Lett. 2014, 8, 116–120. [Google Scholar] [CrossRef]
  71. Choi, H.S. Chemical Composition of the Essential Oil from Cirsium setidens, a Korean Medicinal Plant. TACL 2015, 5, 94–102. [Google Scholar]
  72. Nazaruk, J.; Jablonski, J. Chemical constituents of chloroform and petroleum extracts from Cirsium palustre flower heads. Chem. Nat. Compd. 2011, 47, 654–655. [Google Scholar] [CrossRef]
  73. Nazaruk, J.E.; Karna Kalemba, D. The chemical composition of the essential oils of Cirsium palustre and C. rivulare and their antiproliferative effect. Nat. Prod. Commun. 2012, 7, 269–272. [Google Scholar] [CrossRef] [PubMed]
  74. Mallick, S.; Dutta, A.; Chaudhuri, A.; Mukherjee, D.; Dey, S.; Halder, S.; Ghosh, J.; Sultana, S.S.; Biswas, G.; Lai, T.K.; et al. Successful Therapy of Murine Visceral Leishmaniasis with Astrakurkurone, a Triterpene Isolated from the Mushroom Astraeus hygrometricus, Involves the Induction of Protective Cell-Mediated Immunity and TLR9. Antimicrob. Agents Chemother. 2016, 60, 2696–2708. [Google Scholar] [CrossRef] [PubMed]
  75. Kozyra, M.; Głowniak, K. Phenolic acids in extracts obtained from the flowering herbs of Cirsium vulgare (Savi) Ten. growing in Poland. Acta Soc. Bot. Pol. 2013, 82, 325–329. [Google Scholar] [CrossRef]
  76. Arnason, T.; Swain, T.; Wat, C.K.; Graham, E.A.; Partington, S.; Towers, G.H.N.; Lam, J. Mosquito larvicidal activity of polyacetylenes from species in the Asteraceae. Biochem. Syst. Ecol. 1981, 9, 63–68. [Google Scholar] [CrossRef]
  77. Bohlmann, F.; Burkhardt, T.; Zdero, C. Naturally Occurring Acetylenes; Academic Press: London, UK, 1973. [Google Scholar]
  78. Baek, N.; Park, J.D.; Lee, Y.H.; Jeong, S.Y.; Kim, S.I. A Novel Polyacetylene from Cirsium spp. Pharm. Soc. Korea 1995, 39, 268–275. [Google Scholar]
  79. Guillet, G.; Philogène, B.J.R.; O’Meara, J.; Durst, T.; Arnason, J.T. Multiple modes of insecticidal action of three classes of polyacetylene derivatives from Rudbeckia hirta. Phytochemistry 1997, 46, 495–498. [Google Scholar] [CrossRef]
  80. Harborne, J.B. Classes and functions of secondary products from plants. In Chemicals from Plants; Walton, N.J., Brown, D.E., Eds.; Imperial College Press: London, UK, 1999; pp. 1–26. [Google Scholar]
  81. Shuqing, W.; Hanhong, X.; Shanhuan, Z.; Zhizhen, S.; Zhun, L. Phototoxicity of synthetic polyacetylenes against mosquito larvae (Culex quinquefasciatus), Kun Chong Xue Bao. Acta Entomol. Sin. 2000, 43, 264–270. [Google Scholar]
  82. Ying, W.; Masao, T.; Felicitas, K.; Matthias, H.; Kurt, H. Polyacetylenes fromArtemisia borealis and their biological activities. Phytochemistry 1990, 29, 3101–3105. [Google Scholar] [CrossRef]
  83. Wat, C.-K.; Prasad, S.; Graham, E.; Partington, S.; Arnason, T.; Towers, G.; Lam, J. Photosensitization of invertebrates by natural polyacetylenes. Biochem. Syst. Ecol. 1981, 9, 59–62. [Google Scholar] [CrossRef]
  84. Amiri, N.; Yadegari, M.; Hamedi, B. Essential Oil Composition of Cirsium arvense L. Produced in Different Climate and Soil Properties. Rec. Nat. Prod. 2018, 12, 251–262. [Google Scholar] [CrossRef]
  85. Dehjurian, A.; Lari, J.; Motavalizadehkakhky, A. Anti-Bacterial Activity of Extract and the Chemical Composition of Essential Oils in Cirsium arvense from Iran. J. Essent. Oil Bear. Plants 2017, 20, 1162–1166. [Google Scholar] [CrossRef]
  86. Zeng, Q.-H.; Zhao, J.-B.; Wang, J.-J.; Zhang, X.-W.; Jiang, J.-G. Comparative extraction processes, volatile compounds analysis and antioxidant activities of essential oils from Cirsium japonicum Fisch. ex DC and Cirsium setosum (Willd.) M.Bieb. LWT 2016, 68, 595–605. [Google Scholar] [CrossRef]
  87. Borawska, M.H.; Czechowska, S.K.; Markiewicz, R.; Socha, K.; Nazaruk, J.; Pałka, J.; Isidorov, V.A. Enhancement of antibacterial effects of extracts from Cirsium species using sodium picolinate and estimation of their toxicity. Nat. Prod. Res. 2010, 24, 554–561. [Google Scholar] [CrossRef] [PubMed]
  88. Strawa, J.; Wajs-Bonikowska, A.; Leszczyńska, K.; Ściepuk, M.; Nazaruk, J.; Jakub, S.; Anna, W.B.; Leszczyńska, K.; Ściepuk, M.; Nazaruk, J. Chemical composition and antioxidant, antibacterial activity of Cirsium rivulare (Jacq) All. roots. Nat. Prod. Res. 2016, 30, 2730–2733. [Google Scholar] [CrossRef] [PubMed]
  89. Lee, S.H.; Heo, S.-I.; Li, L.; Lee, M.J.; Wang, M.-H. Antioxidant and Hepatoprotective Activities of Cirsium setidens NAKAI against CCl4-Induced Liver Damage. Am. J. Chin. Med. 2008, 36, 107–114. [Google Scholar] [CrossRef] [PubMed]
  90. Jiang, L.; Ji, Z. Chemical constituents from the aerial parts of Cirsium japonicum and their antifungal activities. Chin. J. Pestic. Sci. 2018, 20, 316–322. [Google Scholar]
  91. Nazaruk, J.; Czechowska, K.S.; Markiewicz, R.; Borawska, H.M. Polyphenolic compounds and in vitro antimicrobial and antioxidant activity of aqueous extracts from leaves of some Cirsium species. Nat. Prod. Res. 2008, 22, 1583–1588. [Google Scholar] [CrossRef]
  92. Shin, S.; Saravanakumar, K.; Sathiyaseelan, A.; Mariadoss, A.V.A.; Park, S.; Park, S.; Han, K.; Wang, M.-H. Phytochemical profile and antidiabetic effect of the bioactive fraction of Cirsium setidens in streptozotocin-induced type 2 diabetic mice. Process Biochem. 2022, 116, 60–71. [Google Scholar] [CrossRef]
  93. Kim, M.J.; Lee, S.; Kim, H.Y.; Cho, E.J. Cirsium japonicum var. maackii inhibits hydrogen peroxide-induced oxidative stress in SH-SY5Y cells. Korean J. Agric. Sci. 2021, 48, 119–131. [Google Scholar]
  94. Nalewajko-Sieliwoniuk, E.; Nazaruk, J.; Kotowska, J.; Kojło, A. Determination of the flavonoids/antioxidant levels in Cirsium oleraceum and Cirsium rivulare extracts with cerium(IV)–rhodamine 6G chemiluminescence detection. Talanta 2012, 96, 216–222. [Google Scholar] [CrossRef] [PubMed]
  95. Ma, Q.; Wang, L.H.; Jiang, J.G. Hepatoprotective effect of flavonoids from Cirsium japonicum DC on hepatotoxicity in comparison with silymarin. Food Funct. 2016, 7, 2179–2184. [Google Scholar] [CrossRef]
  96. Noh, Y.-H.; Hong, J.; Lee, J.-W.; Kim, S.-S.; Lee, J.Y.; Kang, I.-J.; Won, M.-H.; Jeong, Y.; Whang, W.K.; Myung, S.-C.; et al. A Complex of Cirsium japonicum var. maackii (Maxim.) Matisum. and Thymus vulgaris L. Improves Menopausal Symptoms and Supports Healthy Aging in Women. J. Med. Food 2022, 25, 281–292. [Google Scholar] [CrossRef] [PubMed]
  97. Jung, H.A.; Abdul, Q.A.; Byun, J.S.; Joung, E.-J.; Gwon, W.-G.; Lee, M.-S.; Kim, H.-R.; Choi, J.S. Protective effects of flavonoids isolated from Korean milk thistle Cirsium japonicum var. maackii (Maxim.) Matsum on tert -butyl hydroperoxide-induced hepatotoxicity in HepG2 cells. J. Ethnopharmacol. 2017, 209, 62–72. [Google Scholar] [CrossRef] [PubMed]
  98. Zhao, Z.-W.; Chang, H.-C.; Ching, H.; Lien, J.-C.; Huang, H.-C.; Wu, C.-R. Antioxidant Effects and Phytochemical Properties of Seven Taiwanese Cirsium Species Extracts. Molecules 2021, 26, 3935. [Google Scholar] [CrossRef]
  99. Shin, M.-S.; Park, J.Y.; Lee, J.; Yoo, H.H.; Hahm, D.-H.; Lee, S.C.; Lee, S.; Hwang, G.S.; Jung, K.; Kang, K.S. Anti-inflammatory effects and corresponding mechanisms of cirsimaritin extracted from Cirsium japonicum var. maackii Maxim. Bioorganic Med. Chem. Lett. 2017, 27, 3076–3080. [Google Scholar] [CrossRef]
  100. Ghil, S.-H.; Kim, D.-Y.; Kang, S.-H. Ghil Cirsium japonicum extract induces apoptosis and anti-proliferation in the human breast cancer cell line MCF-7. Mol. Med. Rep. 2010, 3, 427–432. [Google Scholar] [CrossRef]
  101. Lin, P.-C.; Ji, L.-L.; Zhong, X.-J.; Li, J.-J.; Wang, X.; Shang, X.-Y.; Lin, S. Taraxastane-type triterpenoids from the medicinal and edible plant Cirsium setosum. Chin. J. Nat. Med. 2019, 17, 22–26. [Google Scholar] [CrossRef]
  102. Kim, B.R.; Seo, H.S.; Ku, J.M.; Kim, G.J.; Jeon, C.Y.; Park, J.H.; Jang, B.H.; Park, S.J.; Shin, Y.C.; Ko, S.G. Silibinin inhibits the production of pro-inflammatory cytokines through inhibition of NF-κB signaling pathway in HMC-1 human mast cells. Inflamm. Res. 2013, 62, 941–950. [Google Scholar] [CrossRef]
  103. Zhao, Z.-W.; Chang, J.-C.; Lin, L.-W.; Tsai, F.-H.; Chang, H.-C.; Wu, C.-R. Comparison of the Hepatoprotective Effects of Four Endemic Cirsium Species Extracts from Taiwan on CCl4-Induced Acute Liver Damage in C57BL/6 Mice. Int. J. Mol. Sci. 2018, 19, 1329. [Google Scholar] [CrossRef] [Green Version]
  104. Park, H.S.; Shim, S.M.; Kim, G.H. Silydianin in chloroform soluble fraction of Cirsium japonicum leaf inhibited adipocyte differentiation by regulating adipogenic transcription factors and enzymes. Appl. Biol. Chem. 2013, 56, 709–713. [Google Scholar] [CrossRef]
  105. Gurovic, M.S.V.; Castro, M.J.; Richmond, V.; Faraoni, M.B.; Maier, M.S.; Murray, A.P. Triterpenoids with acetylcholinesterase inhibition from Chuquiraga erinacea D. Don. subsp. erinacea (Asteraceae). Planta Med. 2010, 76, 607–610. [Google Scholar] [CrossRef] [PubMed]
  106. Cho, C.; Kang, L.J.; Jang, D.; Jeon, J.; Lee, H.; Choi, S.; Han, S.J.; Oh, E.; Nam, J.; Kim, C.S.; et al. Cirsium japonicum var. maackii and apigenin block Hif-2α-induced osteoarthritic cartilage destruction. J. Cell Mol. Med. 2019, 23, 5369–5379. [Google Scholar] [CrossRef] [PubMed]
  107. Park, J.Y.; Kim, H.Y.; Shibamoto, T.; Jang, T.S.; Lee, S.C.; Shim, J.S.; Hahm, D.-H.; Lee, H.-J.; Lee, S.; Kang, K.S. Beneficial effects of a medicinal herb, Cirsium japonicum var. maackii, extract and its major component, cirsimaritin on breast cancer metastasis in MDA-MB-231 breast cancer cells. Bioorganic Med. Chem. Lett. 2017, 27, 3968–3973. [Google Scholar] [CrossRef] [PubMed]
  108. Liao, Z.; Wu, Z.; Wu, M. Cirsium japonicum Flavones Enhance Adipocyte Differentiation and Glucose Uptake in 3T3-L1 Cells. Biol. Pharm. Bull. 2012, 35, 855–860. [Google Scholar] [CrossRef] [PubMed]
  109. Jung, H.A.; Park, J.J.; Min, B.S.; Jung, H.J.; Islam, M.N.; Choi, J.S. Inhibition of advanced glycation endproducts formation by Korean thistle, Cirsium maackii. Asian Pac. J. Trop. Med. 2015, 8, 1–5. [Google Scholar] [CrossRef]
  110. Luan, N.; Di Wei, W.; Wang, A.; Wu, X.L.; Qi, Y.; Li, J.J.; Zheng, J.Q.; Shang, X.Y. Four new taraxastane-type triterpenoic acids from Cirsium setosum. J. Asian Nat. Prod. Res. 2016, 18, 1015–1023. [Google Scholar] [CrossRef]
  111. Joy dela Pena, I.; Lee, H.L.; Yoon, S.Y.; de la Peña, J.B.; Kim, K.H.; Hong, E.Y.; Cheong, J.H. The ethanol extract of Cirsium japonicum increased chloride ion influx through stimulating GABA A receptor in human neuroblastoma cells and exhibited anxiolytic-like effects in mice. Drug Discov. Ther. 2013, 7, 18–23. [Google Scholar]
  112. De La Peña, J.B.I.; Kim, C.A.; Lee, H.L.; Yoon, S.Y.; Kim, H.J.; Hong, E.Y.; Kim, G.H.; Ryu, J.H.; Lee, Y.S.; Kim, K.M.; et al. Luteolin mediates the antidepressant-like effects of Cirsium japonicum in mice, possibly through modulation of the GABAA receptor. Arch. Pharmacal Res. 2013, 37, 263–269. [Google Scholar] [CrossRef]
  113. Liao, Z.; Chen, X.; Wu, M. Antidiabetic effect of flavones from Cirsium japonicum DC in diabetic rats. Arch. Pharmacal Res. 2010, 33, 353–362. [Google Scholar] [CrossRef]
  114. Meng, Y.H.; Wang, Q.H.; Yang, L. Pharmacological effects of Cirsium setosum in Hei Long Jiang. Inf. Tradit. Chin. Med. 2011, 28, 17–18. [Google Scholar]
  115. Chen, Q. Study on the Material Basis of Hemostatic Efficacy and the Mechanism of Hemostatic Synergism of Large Thistle Charcoal; Beijing University of Traditional Chinese Medicine: Beijing, China, 2014. [Google Scholar]
  116. Pang, Q.Q.; Kim, J.H.; Choi, J.M.; Song, J.L.; Lee, S.; Cho, E.J. Cirsium japonicum var. Maackii Improves Cognitive Impairment under Amyloid β25-35-Induced Alzheimer’s Disease Model. BioMed Res. Int. 2022, 2022, 1–11. [Google Scholar]
  117. Wang, Y. Study on the Hemostatic Material Basis and Mechanism of Cirsium Japonicum Charcoal; Beijing University of traditional Chinese Medicine: Beijing, China, 2018. [Google Scholar]
  118. Takaishi, Y.; Okuyama, T.; Nakano, K.; Murakami, K.; Tomimatsu, T.; Yamahara, J. Polyolefinic compounds from Cirsium nipponicum roots. Phytochemistry 1991, 30, 1539–1542. [Google Scholar] [CrossRef]
  119. Funk, C.D.; Hoshiko, S.; Matsumoto, T.; Rdmark, O.; Samuelsson, B. Characterization of the human 5-lipoxygenase gene. Proc. Natl. Acad. Sci. USA 1989, 86, 2587–2591. [Google Scholar] [CrossRef] [PubMed]
  120. Özçelik, B.; Deliorman Orhan, D.; Karaoğlu, T.; Ergun, F. Antimicrobial activities of various Cirsium hypoleucum extracts. Ann. Microbiol. 2005, 55, 135–138. [Google Scholar]
  121. Xu, Y.; Tang, J.; Guo, Q.; Xu, Y.; Yan, K.; Wu, L.; Xie, K.; Zhu, A.; Rong, X.; Ye, D.; et al. Traditional Chinese Medicine formula FTZ protects against polycystic ovary syndrome through modulating adiponectin-mediated fat-ovary crosstalk in mice. J. Ethnopharmacol. 2021, 268, 113587. [Google Scholar] [CrossRef]
  122. Zhang, Y.; Sun, M.; Wang, D.; Hu, Y.; Wang, R.; Diao, H.; Guo, J. FTZ protects against cardiac hypertrophy and oxidative injury via microRNA-214/SIRT3 signaling pathway. Biomed Pharm. 2022, 148, 112696. [Google Scholar] [CrossRef]
  123. Luo, D.; Li, J.; Chen, K.; Rong, X.; Guo, J. Untargeted Metabolomics Reveals the Protective Effect of Fufang Zhenshu Tiaozhi (FTZ) on Aging-Induced Osteoporosis in Mice. Front. Pharmacol. 2019, 9, 1483. [Google Scholar] [CrossRef]
  124. Wang, L.; Tao, W.; Luo, D.; Hu, Y.; Bei, W.; Guo, J. Potential synergistic effects of Chinese herbal prescription FTZ components detected in blood towards hepatic lipid-modulating targets. Complement. Ther. Med. 2014, 22, 887–893. [Google Scholar] [CrossRef]
  125. Hu, X.; Wang, M.; Bei, W.; Han, Z.; Guo, J. The Chinese herbal medicine FTZ attenuates insulin resistance via IRS1 and PI3K in vitro and in rats with metabolic syndrome. J. Transl. Med. 2014, 12, 47. [Google Scholar] [CrossRef]
  126. Song, L.; Zhang, D.; Guo, C.; Gu, Z.; Wang, L.; Yao, Y.S.; Wang, H.; Zeng, Z.; Wang, W.; Yang, Y.; et al. The traditional Chinese medicine formula Fufang-Zhenzhu-Tiaozhi protects myocardia from injury in diabetic minipigs with coronary heart disease. Biomed. Pharmacother. 2021, 137, 111343. [Google Scholar] [CrossRef] [PubMed]
  127. Shenghua, P.; Ziqin, Z.; Shuyu, T.; Huixia, Z.; Xianglu, R.; Jiao, G. An integrated fecal microbiome and metabolome in the aged mice reveal anti-aging effects from the intestines and biochemical mechanism of FuFang zhenshu TiaoZhi (FTZ). Biomed. Pharm. 2020, 121, 109421. [Google Scholar] [CrossRef] [PubMed]
  128. Cai, J.; Zhang, J.; Li, S.; Lin, Y.; Xiao, X.; Guo, J. Comprehensive chemical analysis of Zhenshu Tiaozhi formula and its effect on ameliorating glucolipid metabolic disorders in diabetic rats. Biomed. Pharmacother. 2021, 133, 111060. [Google Scholar] [CrossRef] [PubMed]
  129. Diao, H.; Cheng, J.; Huang, X.; Huang, B.; Shao, X.; Zhao, J.; Lan, D.; Zhu, Q.; Yan, M.; Zhang, Y.; et al. The Chinese medicine Fufang Zhenzhu Tiaozhi capsule protects against atherosclerosis by suppressing EndMT via modulating Akt1/β-catenin signaling pathway. J. Ethnopharmacol. 2022, 293, 115261. [Google Scholar] [CrossRef] [PubMed]
  130. Yang, Y.-Q.; Tan, H.-B.; Zhang, X.-Y.; Zhang, Y.-Z.; Lin, Q.-Y.; Huang, M.-Y.; Lin, Z.-Y.; Mo, J.-Z.; Zhang, Y.; Lan, T.; et al. The Chinese medicine Fufang Zhenzhu Tiaozhi capsule protects against renal injury and inflammation in mice with diabetic kidney disease. J. Ethnopharmacol. 2022, 292, 115165. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, L.; Xiang, L.; Piao, S.; Gong, X.; Zhou, W.; Feng, W.; Li, H.; Li, L.; Wei, A.; Zhu, Q.; et al. The Efficacy and Safety of Chinese Medicine Fufang Zhenzhu Tiaozhi Capsule (FTZ) in the Treatment of Diabetic Coronary Heart Disease: Study Protocol for Multicenter, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Diabetes Metab. Syndr. Obesity Targets Ther. 2021, 14, 2651–2659. [Google Scholar] [CrossRef]
  132. Yoon, S.; Kim, M.; Shin, S.; Woo, J.; Son, D.; Ryu, D.; Yoo, J.; Park, D.; Jung, E. Effect of Cirsium japonicum Flower Extract on Skin Aging Induced by Glycation. Molecules 2022, 27, 2093. [Google Scholar] [CrossRef]
  133. Kim, K.M.; Son, H.D.; Shin, W.S.; Jung, S.E.; Park, H. Composition Containing Cirsium japonicum Extract as Active Ingredient for Stimulating Melanogenesis. U.S. Patent 2022/0287956, 15 September 2022. [Google Scholar]
  134. Oku, H.; Kamiyama, Y.; Inafuku, M. Method for Treating Fatty Liver. U.S. Patent 15/903,823, 19 May 2020. [Google Scholar]
  135. Mori, S.; Ichii, Y.; Tanaka, N.; Yorozu, H.; Kanazawa, S.; Nishizawa, Y. Lipolysis Acceleration Method. U.S. Patent 5,698,199, 16 December 1997. [Google Scholar]
  136. Kim, J.W.; Park, Y.C.; Lee, K.H.; Hyungwoo, K.I.M. Composition for Preventing, Ameliorating or Treating Acne Symptoms Using Natural Extracts as Active Ingredients. U.S. Patent 11,154,580, 26 October 2021. [Google Scholar]
  137. Shimotoyodome, Y.; Ito, S.; Sugai, Y.; Hashimoto, H.; Ishikawa, J. Ceramide Production Enhancer and Moisturizer. U.S. Patent 9,682,029, 20 June 2017. [Google Scholar]
  138. Wang, H.C.; Bao, Y.R.; Wang, S.; Li, T.J.; Meng, X.S. Simultaneous determination of eight bioactive components of Cirsium setosum flavonoids in rat plasma using triple quadrupole LC/MS and its application to a pharmacokinetic study. Biomed. Chromatogr. 2019, 33, e4632. [Google Scholar] [CrossRef]
  139. Zhang, X.; Liao, M.; Cheng, X.; Liang, C.; Diao, X.; Zhang, L. Ultrahigh-performance liquid chromatography coupled with triple quadrupole and time-of-flight mass spectrometry for the screening and identification of the main flavonoids and their metabolites in rats after oral administration of Cirsium japonicum DC extract. Rapid Commun. Mass Spectrom. 2018, 32, 1451–1461. [Google Scholar]
  140. Zhang, Z.; Jia, P.; Zhang, X.; Zhang, Q.; Yang, H.; Shi, H.; Zhang, L. LC–MS/MS determination and pharmacokinetic study of seven flavonoids in rat plasma after oral administration of Cirsium japonicum DC. extract. J. Ethnopharmacol. 2014, 158, 66–75. [Google Scholar] [CrossRef]
  141. Lee, J.S.; Kim, Y.-H.; Kim, D.-B.; Bang, W.-S.; Lee, O.-H. Acute and 4-Week Repeated-Dose Oral Toxicity Studies of Cirsium setidens in Rats. Molecules 2014, 19, 7138–7151. [Google Scholar] [CrossRef] [PubMed]
  142. Shim, S.M.; Bae, E.; Kim, G.H. Single-Dose Oral Acute Toxicity and Mutagenic Effects of Methanol Extracts of Cirsium japonicum. J. Food Biochem. 2011, 35, 845–858. [Google Scholar] [CrossRef]
  143. Shin, S.; Saravanakumar, K.; Mariadoss, A.V.A.; Hu, X.; Sathiyaseelan, A.; Wang, M.H. Functionalization of selenium nanoparticles using the methanolic extract of Cirsium setidens and its antibacterial, antioxidant, and cytotoxicity activities. J. Nanostructure Chem. 2022, 12, 23–32. [Google Scholar] [CrossRef]
  144. Barbinta-Patrascu, M.E.; Ungureanu, C.; Besliu, D.; Lazea-Stoyanova, A.; Iosif, L. Bio-active nanomaterials phyto-generated from weed herb Cirsium arvense. Optoelectron. Adv. Mater. Rapid Commun. 2020, 14, 459–465. [Google Scholar]
  145. Tahir, K.; Nazir, S.; Li, B.; Khan, A.U.; Khan, Z.U.H.; Ahmad, A.; Khan, Q.U.; Zhao, Y. Enhanced visible light photocatalytic inactivation of Escherichia coli using silver nanoparticles as photocatalyst. J. Photochem. Photobiol. B Biol. 2015, 153, 261–266. [Google Scholar] [CrossRef] [PubMed]
  146. Fallahi, M.; Norouzi, B. Synthesis of cobalt oxide nanoparticles using Cirsium vulgare leaves extract and evaluation of electrocatalytic effects on oxidation of l-cysteine. Ionics 2020, 26, 1951–1961. [Google Scholar] [CrossRef]
  147. Khan, Z.U.H.; Khan, A.; Shah, A.; Wan, P.; Chen, Y.; Khan, G.M.; Khan, A.U.; Tahir, K.; Muhammad, N.; Khan, H.U. Enhanced photocatalytic and electrocatalytic applications of green synthesized silver nanoparticles. J. Mol. Liq. 2016, 220, 248–257. [Google Scholar] [CrossRef]
  148. Rehman, K.U.; Khan, A.U.; Tahir, K.; Nazir, S.; Albalawi, K.; Hassan, H.M.; Alabbad, E.A.; Refat, M.S.; Al-Shehri, H.S.; Aldawsari, A.M. Facile synthesis of copper oxide nanoparticles (CuONPs) using green method to promote photocatalytic and biocidal applications. J. Mol. Liq. 2022, 360, 119453. [Google Scholar] [CrossRef]
  149. Luo, J.; Zhang, M.; Cheng, J.; Wu, S.; Xiong, W.; Kong, H.; Zhao, Y.; Qu, H. Hemostatic effect of novel carbon dots derived from Cirsium setosum Carbonisata. RSC Adv. 2018, 8, 37707–37714. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The botanical classification of the genus Cirsium.
Figure 1. The botanical classification of the genus Cirsium.
Chemistry 04 00079 g001
Figure 2. (A) Leaves of Cirsium (B) Cirsium Whole Plant (C) Aboveground parts of Cirsium.
Figure 2. (A) Leaves of Cirsium (B) Cirsium Whole Plant (C) Aboveground parts of Cirsium.
Chemistry 04 00079 g002
Figure 3. Commonly found flavonoids in different species of Cirsium.
Figure 3. Commonly found flavonoids in different species of Cirsium.
Chemistry 04 00079 g003
Figure 4. Infrequently distributed flavonoids in Cirsium species.
Figure 4. Infrequently distributed flavonoids in Cirsium species.
Chemistry 04 00079 g004
Figure 5. Major flavone glucoside detected in different species of Cirsium.
Figure 5. Major flavone glucoside detected in different species of Cirsium.
Chemistry 04 00079 g005
Figure 6. Other polyphenolic compounds isolated from Cirsium species.
Figure 6. Other polyphenolic compounds isolated from Cirsium species.
Chemistry 04 00079 g006
Figure 7. Terpenoids found in different species of Cirsium.
Figure 7. Terpenoids found in different species of Cirsium.
Chemistry 04 00079 g007
Figure 8. Major sterols identified in different species of Cirsium.
Figure 8. Major sterols identified in different species of Cirsium.
Chemistry 04 00079 g008
Figure 9. Major phenolic acids found in different species of Cirsium.
Figure 9. Major phenolic acids found in different species of Cirsium.
Chemistry 04 00079 g009
Figure 10. Polyacetylenes, acetylenes, and hydrocarbons found in Cirsium species.
Figure 10. Polyacetylenes, acetylenes, and hydrocarbons found in Cirsium species.
Chemistry 04 00079 g010
Figure 11. Fatty acids, aldehydes, and ketones found in Cirsium species.
Figure 11. Fatty acids, aldehydes, and ketones found in Cirsium species.
Chemistry 04 00079 g011
Figure 12. Mode of action of Silybin against gastric cancer.
Figure 12. Mode of action of Silybin against gastric cancer.
Chemistry 04 00079 g012
Figure 13. Mode of action of Cirsium species against Alzheimer’s disease.
Figure 13. Mode of action of Cirsium species against Alzheimer’s disease.
Chemistry 04 00079 g013
Table 1. Distribution and pharmacological potential of some species of Cirsium.
Table 1. Distribution and pharmacological potential of some species of Cirsium.
S. NoSpecies of CirsiumGeographical DistributionTraditional UsesCommon NameMajor PhytoconstituentsPharmacological ApplicationsReferences
1Cirsium arvenseEurope, Asia, Northern Africa,
India
Pharyngitis, astringent, tonic, tumor, diuretic, toothache, diaphoreticCanada thistle, Creeping thistle, Field thistle, Californian thistleAcacetin, Ciryneol C, Hispidulin, Pectolinarigenin, Luteolin, Tracin, Scopoletin, Apigenin, CitronellolAntimicrobial, antifungal, anticancer,
antidiabetic,
neuroprotective,
anti-inflammatory
[5,8]
2Cirsium oleraceumEurope to West Siberia and KazakhstanAnxiolytic, diuretic, astringent, antiphlogistic, AntitumorCabbage thistle, Siberian thistleThymol, Carvacrol, Luteolin, Apigenin, MethylkaempferolAntioxidant, antimicrobial, anti-glioma effect[20]
3Cirsium englerianumEthiopiaDermal infections, cough, snake bite, hematuria, diarrhea
anthrax,
anti-scabies
-Alkaloids, Quinones, Terpenoids, Phenolics, and Flavonoids Antioxidant, antimicrobial[5]
4Cirsium eriophorumChina South-Central, East Himalaya, Myanmar, TibetDetoxification and cure of hepatic infectionsWoolly thistleVanillic acid, Balanophonin, Apigenin, Kaempferol, Taraxasterol, Sitosterol, Linoleic acidAntioxidant, acetylcholinesterase inhibitory activity[21]
5Cirsium wallichiiAfghanistan, East Himalaya, Nepal, Pakistan, West HimalayaPyrexia, bleeding relief, burning sensation, and stomach inflammationWallich’s Thistle, Plume thistlesAcetyljacoline, Fumaric acidAntimicrobial, antifungal, antioxidant[5]
6Cirsium verutumAssam, East Himalaya, Myanmar, Nepal, Pakistan, Tibet, Vietnam, West HimalayaTyphoid, bleeding, chest pain, measles, purgative, pharyngitis,
dyspepsia,
dysentery,
tuberculosis
Common thistle, Creeping thistle, Plume thistleLupeol, Taraxasterol acetate, Pectolinarigenin,
Cirsitakaoside,
Cirsitakaogenin,
Pectolinarin
Antimicrobial,
antifungal
[22]
7Cirsium
setidens
KoreaPyrexia, detoxify, and improve blood circulationUngungqwui’ in Korea,
Thistles in English
Linarin, Phytol, Syringin, Pectolinarigenin, Cyclocitral, Pentylfuran,
Trans-β-Ionone
Rutin, Setidenosides, Isorhamnetin
Antimicrobial, antifungal, anticancer, neuroprotective,
anti-inflammatory
Antidiabetic,
osteogenic agent
[23]
8Cirsium tenoreanumItalyTreatment of varicoseCardo di TenoreKaempferol, Apigenin, Quercetin-3-O-galactosideAntimicrobial,
antiproliferative
[24]
9Cirsium vulgareEurope to Siberia and Arabian Peninsula, West HimalayaAnxiolyticSpear or bull thistleQuercetin, Apigenin,
Kaempferol, and Luteolin
Antioxidant,
antimicrobial
[9]
10Cirsium
japonicum
China, Korea, JapanHemorrhages, cancer, hypertension, and hepatitisJapanese thistleLinarin, Luteolin, Coumaric acid Pectolinarin, Ciryneol, Syringin, Cirsimaritin Pectolinarigenin, Lupenyl acetateAnticancer,
anti-Alzheimer.
anti-inflammatory,
antimicrobial
[25]
Table 2. Major phytoconstituents present in various species of Cirsium.
Table 2. Major phytoconstituents present in various species of Cirsium.
Category of PhytoconstituentName of PhytoconstituentsSpeciesReferences
FlavanoidsLinarinC. arvense[46]
C. japonicum[62,64]
C. setosum[64]
C. rivulare[65]
C. canum[63]
ScopoletinC. arvense[51]
PectolinarigeninC. chanroenicum[59]
C. setidens[60]
Pectolinarigenin-7-O-glucopyranosideC. arvense[51]
AcacetinC. arvense[51]
6,7-DimethoxycoumarinC. arvense[51]
TracinC. arvense[51]
HispidulinC. arvense[35]
C. japonicum[54,55]
C. rivulare[30]
Hispidulin-7-neohesperidosideC. japonicum[64]
LuteolinC. arvense[35]
C. japonicum[55,64]
C. canum[63]
C. palustre[56]
C. rivulare[62]
Luteolin 7-O-β-D-glucuronideC. scabrum[26]
Luteolin 7-O-β-D-glucosideC. scabrum[26]
C. canum[63]
C. palustre[56]
Eriodictyol 7-O-glucosideC. palustre[56]
PectolinarinC. japonicum[53,57,64]
C. rivulare[50]
C. subcoriaceum[58]
C. chanroenicum[59]
C. setidens[60]
C. japonicum[25]
Isokaempferide 7-O-β-D-(6″-methylglucuronideC. rivulare[50]
Isokaempferide 7-glucuronideC. rivulare[50]
ApigeninC. canum[63]
C. setosum[61]
C. rivulare[30]
C. japonicum[55]
Apigenin 7-(6″-methylglucuronide)C. rivulare[30,50]
Apigenin 7-glucosideC. canum[63]
C. rivulare[30]
KaempferolC. canum[63]
Kaempferol 3-galactosideC. rivulare[50]
Kaempferol 3-glucosideC. canum[63]
Kaempferol 3- β-D-glucopyranoside (Astragalin)C. setosum[61]
4-Vinyl guaiacolC. creticum[17]
4-Ethyl guaiacolC. creticum[17]
5,7-Dihydroxy-6,4′-dimethoxyflavoneC. japonicum[53,57]
CirsimaritinC. japonicum[49]
CirsimarinC. japonicum[49]
RutosideC. canum[68]
6-Hydroxyluteolin 7-O-glucosideC. palustre[56]
TricinC. rivulare[69]
IsorhamnetinC. helenioides[66]
SteroidsStigmasterolC. arvense[46]
Steroidal glucosideDaucosterolC. arvense[46]
AlkaloidsBenzymidazole C. arvense[46]
Terpenesα-TocopherolC. setidens[23]
C. arvense[35]
α-Tocospiro A, B and CC. setosum[70]
4(15),10(14)-Guaiadien-12,6-olide C. setidens[23]
Trans-PhytolC. setidens[23,71]
DihydroactinidiolideC. creticum[17]
TriterpenesLupeolC. scabrum[47]
Lupeol acetateC. palustre[72]
Taraxasterol acetateC. scabrum[47]
25-Hydroperoxycycloart-23-en-3β-olC. scabrum[47]
C. setidens[23]
β-AmyrinC. palustre[72]
FaradiolC. palustre[72]
SesquiterpenesCaryophyllene oxide C.setidens[71]
β-Caryophyllene alcoholC.setidens[71]
Cyclic etherCiryneolC. arvense[51]
1,2,15,16-DiepoxyhexadecaneC. setidens[71]
Fatty acids9, 12, 15-Octadecatrienoic acidC. setidens[23]
9, 12-Octadecadienoic acidC. setidens[23]
Hexadecanoic acidC. setidens[23]
C. creticum[17]
C. palustre[72]
Palmitic acidC. japonicum[41]
SterolsAcylglycosyl β-sitosterolC. setidens[23]
β-Sitosterol glucosideC. setidens[23]
TaraxasterolC. setosum[61]
GlycerolMonogalactosyldiacyl glycerolC. setidens[23]
C. helenioides[66]
C. palustre[73]
C. rivulare[73]
DihydroaplotaxeneC. helenioides[66]
TetrahydroaplotaxeneC. helenioides[66]
PentacosaneC. setidens[71]
AldehydesSinapaldehydeC. helenioides[66]
Ketones6,10,14-Trimethyl-2-pentadecanoneC. setidens[71]
Phenolic acidsChlorogenic acidC. canum[63]
C. palustre[56]
Caffeic acidC. canum[63]
p-Coumaric acidC. canum[63]
Protocatechuic acidC. canum[63]
p-Hydroxybenzoic acidC. canum[63]
Vanillic acidC. canum[63]
Syringic acidC. canum[63]
Trans-Cinnamic acidC. canum[63]
Table 3. Antimicrobial and antioxidant potential of different Cirsium species.
Table 3. Antimicrobial and antioxidant potential of different Cirsium species.
S. NoCirsium
Species
ApplicationModelDetailed InformationReferences
1.C. scabrumIn vitroS. aureus,
Dermabacter
hominis
Moderate activity[47]
2.C. canumIn vitroGram-positive
Bacteria
Inhibitory activity against S. aureus and S. pneumoniae[63]
3.C. arvenseIn vitroS. aureus, S. typhiZone of inhibition:
9–32 mm
[35]
4.C. oleraceum
C. palustre
C. rivulare
C. vulgare
C. arvense
In vitroS. aureus
P. aeruginosa
B. subtilis
C. albicans
Micrococcus luteus
E. coli
Minimum inhibitory concentration range from 3.12–50 mg/mL[50,89]
5.C. hypoleucumIn vitroS. aureusInhibitory activity against S. aureus at 32 μg/mL[92]
6.C. setidensIn vitro-DPPH activity: IC50 value of 45.14 g/mL[93]
7.C. japonicumIn vitroNeuronal cellsMore levels of heme oxygenase, thioredoxin reductase, antioxidative enzymes[94]
8.C. arvenseIn vitro-DPPH activity:118 µg/mL[46,52]
9.C. palustreIn vitro-CAF(Flower) > CAR (Root) > CAL (Leaf) > CAS (Stem)[56]
10.C. leucopsis
C. sipyleum
C. eriophorum
In vitro-DPPH inhibition: 4–38.98 %[95]
11.C. oleraceum
C. rivulare
In vitro-ABTS scavenging activity: >85%[96]
12.C. setidensIn vivoWister albino ratsDPPH inhibition: 2.15–30%[21]
13.C. arvense
C. oleraceum
C. palustre
C. rivulare
In vitro-Total antioxidant activity: 0.98 to 2.71 mM/L[39]
Table 4. Anticancer and anti-inflammatory activities of Cirsium species.
Table 4. Anticancer and anti-inflammatory activities of Cirsium species.
S. NoCirsium SpeciesApplicationModelDetailed InformationReferences
1.C. scabrumIn vitroJ774 cancerous cell lineIC50 = 11.53 μg/mL[47]
2.C. rivulareIn vitroMCF-7 and MDA-MBA-breast cancer cell lineIC50 = 110 to 140 μg/mL[73]
3.C. setosumIn vitroHCT8 colon cancer cellsIC50 = 0.03 μM[70]
4.C. tenoreanumIn vitroMCF7 breast cancer cells73% cell death[24]
5.C. arvenseIn vitroHeLa and C6 cell linesCAR > CAF > CAL [46]
6.C. setidensIn vitroLung, skin, ovarian, and colon cancer cellsED50 = 2.66 to 11.25 μM[23]
7.C. japonicumIn vitroBreast cancer cellsReduction in angiogenesis by lowering the production of VEGF, Akt, and ERK in MDA-MB-231 cells[100]
8.C. japonicumIn vitroMCF-7 cellsArresting the cell cycle in the G1 phase and induced apoptosis[101]
9.C. chanroenicumIn vitroRAW macrophage cells and murine leukemia cellsInhibition of cyclooxygenase and leukotriene production [59]
10.C. subcoriaceumIn vivoMurine modelED30 = 25 mg/kg[58]
11.C. japonicumIn vitroMacrophage cell line
Mast cell line
Reduction in pro-inflammatory cytokines, NO and NF-κB in Mast Cells[102,103]
Table 5. Other pharmacological activities of Cirsium species.
Table 5. Other pharmacological activities of Cirsium species.
ActivityCirsium
Species
ApplicationModelDetailed InformationReferences
Oviposition
stimulatory
C. japonicumIn vitroOstrinia zealisExtract potently induced oviposition by females[41]
AllelopathyC. creticumIn vivoRadish Lettuce
Cress
Inhibitory activity on germination[17]
Enzyme inhibition
activity
C. japonicumIn vitro and In vivoChondrocytesDecrease the levels of Hif-2α, metalloproteinases, and cyclooxygenases[104]
Flavonoids of C. japonicumIn vitroAldose reductase inhibitorIC50 values of 0.21 μg/mL and 0.77 μM[54]
C. leucopsis
C. sipyleum
C. eriophorum
In vitroAcetyl-and butyryl-cholinesterase inhibitory activity16–57% Inhibition[105,106]
C. japonicumIn vivoMurine modelReduction in the levels of lipoprotein lipase and fatty acid synthetase[106]
HepatoprotectiveC. japonicum
Cirsii herba
In vivoC57BL/6 MiceDecrease in liver necrosis restored the hepatic antioxidant enzymes and malondialdehyde[107]
C. arisanenseIn vitro and In vivoHep 3B Cells and MiceReduction in Hepatitis B surface antigen. Declined the levels of SGOT and SGPT[42]
C. setidensIn vivoMiceDecrement in hepatic damage in rats induced due to CCl4 and hepatic ballooning degeneration[21,83]
NephroprotectiveC. japonicumIn vivoMurine ModelDecrease the levels of Cholesterol and triglycerides[108]
C. japonicumIn vitro3T3-L1 CellsEnhancement in insulin-stimulated glucose uptake[109]
ImmunomodulatoryC. japonicumIn vivoMurine modelInduction of humoral and cellular immune responses.
Activation of complement pathway and Natural killer cell activity
[57]
Table 6. Details of patents of Cirsium.
Table 6. Details of patents of Cirsium.
S. NoTitle of PatentApplicantPublished
Application
Number
1.Composition containing Cirsium japonicum extract as active ingredient for stimulating melanogenesisBiospectrum Inc.US20210361559A1
2.Immunoregulatory composition containing Cirsium maritimum extractKochi Prefectural University Corp KochiJP6882730B2
3.Organic extract of plant of genus Cirsium, and application and composition thereofZhejiang Wolwo Bio Pharmaceutical Co., Ltd.CN112022892A
4.Method for treating fatty liverNPO Amami Functional Foods Study Group University of the Ryukyus Amino UP Co., Ltd.US10653740B2
5.Ceramide production enhancer and moisturizerKao CorpUS9682029B2
6.Composition for preventing, ameliorating, or treating acne symptoms using natural extracts as active ingredientsCelim Biotech Co., Ltd.US11154580B1
7.Preparation composition for external use for skin and bath agent compositionKao CorpJPH09208483A
8.Composition for bubble bathKao CorpJPH10147516A
9.Adiponectin secretion promoting agentNPO Amami Functional Foods Study Group Tokunoshima Town Osaka University NUCUS20210283207A1
10.Lipolysis acceleration methodKao CorpUS5698199A
11.Fat accumulation inhibitor, drug, prophylactic or therapeutic agent for fatty liver, food or drink, and method for producing fat accumulation inhibitorNPO Amami Functional Foods Study Group Amino UP Chemical Co., Ltd. University of the RyukyusUS20160184378A1
Table 7. Pharmacokinetic information of Cirsium in vivo.
Table 7. Pharmacokinetic information of Cirsium in vivo.
ModelAdministration
Method
Quantitative MethodDetailsReferences
Sprague-Dawley ratsOral UHPLC-Q-TOF-MSQuercetin, Luteolin, Diosmetin, Cirsimarin, Linarin, Apigenin, Cirsimaritin, Pectolinarin, Tilianin, Hispedulin, Pectolinarigenin, Acacetin were detected[139]
Sprague-Dawley ratsOralLC-MSMaximum Cmax for quercetin = 513.2 ng/mL, while the minimum Cmax of diosmetin = 231.2 ng/mL
AUC0–t value of Compounds (Higher Bioavailability)
Quercetin = 6071 ng·h/mL
Wogonin = 3789 ng·h/mL
Naringin = 2808 ng·h/mL
Acacetin = 2636 ng·h/mL
Rutin = 1884 ng·h/mL
AUC0–t value of Compounds (Lower Bioavailability)
Diosmetin = 238.0 ng·h/mL
[138]
Sprague-Dawley ratsOralLC-MS/MSCmax of detected Compounds (ng/mL)
Pectolinarin = 876
Diosmetin = 37
Pectolinarigenin = 6
Linarin = 86
Hispidulin = 32
Acacetin = 19
Apigenin = 148
Tmax of Pectolinarin, linarin, pectolinarigenin, hispidulin, diosmetin, acacetin = 5 min
Tmax of Apigenin = 360 min
[140]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Aggarwal, G.; Kaur, G.; Bhardwaj, G.; Mutreja, V.; Sohal, H.S.; Nayik, G.A.; Bhardwaj, A.; Sharma, A. Traditional Uses, Phytochemical Composition, Pharmacological Properties, and the Biodiscovery Potential of the Genus Cirsium. Chemistry 2022, 4, 1161-1192. https://doi.org/10.3390/chemistry4040079

AMA Style

Aggarwal G, Kaur G, Bhardwaj G, Mutreja V, Sohal HS, Nayik GA, Bhardwaj A, Sharma A. Traditional Uses, Phytochemical Composition, Pharmacological Properties, and the Biodiscovery Potential of the Genus Cirsium. Chemistry. 2022; 4(4):1161-1192. https://doi.org/10.3390/chemistry4040079

Chicago/Turabian Style

Aggarwal, Gaurav, Gurpreet Kaur, Garima Bhardwaj, Vishal Mutreja, Harvinder Singh Sohal, Gulzar Ahmad Nayik, Anikesh Bhardwaj, and Ajay Sharma. 2022. "Traditional Uses, Phytochemical Composition, Pharmacological Properties, and the Biodiscovery Potential of the Genus Cirsium" Chemistry 4, no. 4: 1161-1192. https://doi.org/10.3390/chemistry4040079

Article Metrics

Back to TopTop