Next Article in Journal
Development and Validation of a LC–MS/MS-Based Assay for Quantification of Free and Total Omega 3 and 6 Fatty Acids from Human Plasma
Previous Article in Journal
Tripleurin XIIc: Peptide Folding Dynamics in Aqueous and Hydrophobic Environment Mimic Using Accelerated Molecular Dynamics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Traditional Uses, Botany, Phytochemistry, Pharmacology, Pharmacokinetics and Toxicology of Xanthium strumarium L.: A Review

1
School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
2
Sichuan Neautus Traditional Chinese Herb Limited Company, Chengdu 611731, China
*
Authors to whom correspondence should be addressed.
Molecules 2019, 24(2), 359; https://doi.org/10.3390/molecules24020359
Submission received: 28 December 2018 / Revised: 10 January 2019 / Accepted: 16 January 2019 / Published: 19 January 2019
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
Xanthium strumarium L. (Asteraceae) is a common and well-known traditional Chinese herbal medicine usually named Cang-Er-Zi, and has been used for thousands of years in China. The purpose of this paper is to summarize the progress of modern research, and provide a systematic review on the traditional usages, botany, phytochemistry, pharmacology, pharmacokinetics, and toxicology of the X. strumarium. Moreover, an in-depth discussion of some valuable issues and possible development for future research on this plant is also given. X. strumarium, as a traditional herbal medicine, has been extensively applied to treat many diseases, such as rhinitis, nasal sinusitis, headache, gastric ulcer, urticaria, rheumatism bacterial, fungal infections and arthritis. Up to now, more than 170 chemical constituents have been isolated and identified from X. strumarium, including sesquiterpenoids, phenylpropenoids, lignanoids, coumarins, steroids, glycosides, flavonoids, thiazides, anthraquinones, naphthoquinones and other compounds. Modern research shows that the extracts and compounds from X. strumarium possess wide-ranging pharmacological effects, including anti- allergic rhinitis (AR) effects, anti-tumor effects, anti-inflammatory and analgesic effects, insecticide and antiparasitic effects, antioxidant effects, antibacterial and antifungal effects, antidiabetic effects, antilipidemic effects and antiviral effects. However, further research should focus on investigating bioactive compounds and demonstrate the mechanism of its detoxification, and more reasonable quality control standards for X. strumarium should also be established.

1. Introduction

Since 1963, the fruits of Xanthium strumarium L. have been listed in the Pharmacopoeia of the People’s Republic of China (CH.P), and currently over 60 formulas containing the fruits of X. strumarium have been applied for treating various diseases, including rhinitis, nasal sinusitis, headache, gastric ulcer, urticarial, rheumatism, bacterial and fungal infections, and arthritis [1,2,3]. So far, many studies have been devoted to the pharmacological and phytochemical studies of X. strumarium, and more than 170 chemical compounds have been isolated and identified from this plant, including sesquiterpene lactones, phenols, glycoside, alkaloids, fatty acid and others [4]. In addition, increasing evidence has indicated that X. strumarium possesses a wide spectrum of pharmacological activities including analgesic and anti-inflammatory, antioxidant, hypoglycemic, anti-cancer, antibacterial and antifungal, anti-trypanosomal, anti-tussive activities, and effects on nervous and digestive systems, as well as other effects [1]. Nowadays, the fruits of X. strumarium remains a common Traditional Chinese Medicine (TCM) listed in the CH.P, and atractyloside and chlorogenic acid are used as the quality indicator agents for evaluating quality of the fruits of X. strumarium [5].
In this paper, we systematically summarize the traditional uses, botany, phytochemistry, pharmacology, pharmacokinetics as well as the safety aspects of X. strumarium, hoping that it could propel the research forward for applying the medicinal values of this plant completely. Moreover, potential research directions and emphasis on Xanthium strumarium L. are discussed as well.

2. Traditional Usages

X. strumarium has a long history for utilization as a medicinal plant in China due to its extensive biological and pharmacological activities. In particular, the fruit is the predominant medicinal part of X. strumarium, and is one of the most common used herbal medicines to treat rhinitis and headache for thousands years [6]. Before clinical use, the fruits of X. strumarium are often processed by stir-baking to a yellowish color, which aims to reduce toxicity and enhance efficacy. The first record of the pharmacological effects of this plant can be traced back to ShenNong BenCaoJing, which is the earliest monograph of TCM during the Eastern Han dynasty. In this monograph, it was used for the treatment of anemofrigid headache and rheumatic arthralgia. Then, in Mingyi Bielu which is another known TCM monograph, X. strumarium was recorded as an effective herbal medicine with the function of curing gonyalgia. In Yaoxinglun, X. strumarium was described as an agent for treating hepatic heat and eye diseases. Subsequently, another famous monograph, Xinxiu Bencao, described X. strumarium with improving eyesight, antiepileptic and antirheumatic properties. Besides, X. strumarium was also listed in some other classical monographs of materia medica in China, such as Bencao Shiyi, Bencao Mengquan, Depei Bencao, Caomu Bianfang, Tianbao Bencao and others.
Currently, the fruits of X. strumarium have become an important traditional Chinese medicine commonly used in clinic for the treatment of nasal diseases (including acute and chronic rhinitis, allergic rhinitis (AR), nasosinusitis, and nasal obstruction), itching diseases, and painful diseases. In order to meet clinical needs better, various forms of formulas are developed, such as pills, tablets, granules, oral liquid, powders and others (Table 1). Furthermore, in India, X. strumarium, commonly known as Chotagokhru or Chotadhatura, are usually used to cure leucoderma, poisonous bites of insects, epilepsy, and biliousness [7]. In addition, several North American Indian tribes and Zuni tribes apply this plant to relieve constipation, diarrhoea and vomiting [1]. Besides, X. strumarium is also reported as a folk herbal medicine in Bangladesh for the treatment of urinary disorder, ear infection, diabetic, and gastric disorder [8].
Apart from clinical application, its potential capacity as a biodiesel feedstock has been proven. X. strumarium has very strong environmental adaptability and thus has numerous wild resources. The seed has a high oil content (42.34%) which gives potential annual output of 100,000 tons just in China [9]. Furthermore, the research in Pakistan also found the prospects of non-edible seed oils for use as biodiesel to solve the serious energy crisis [10].

3. Botany

Xanthium, belonging to the Asteraceae family, is a taxonomically complex genus, which includes more than 20 species in the world and three species and one varietas in China [8]. Xanthium strumarium L. (Figure 1) is an annual herb approximately 20–90 cm in height, its stems are erect, branched, often speckled with purple and have short white hairs scattered across the surface. Leaves are green, cauline, mostly alternate (proximal 2–6 sometimes opposite) with petiole, which are 5–20 cm long and 4–16 cm wide; the shape of blades are lanceolate, linear, ovate, orbicular-deltate, or suborbicular, and both surfaces are hirtellous or strigose, usually with gland-dotted, margin entire or toothed. The capitula are discoid, whose female (proximal) or functionally male (distal) are in racemiform to spiciform arrays or borne singly (in axils). The female capitula are elliptic, 2–5 mm in diameter; Male capitula are saucer-shaped, 3–5 mm in diameter. The achenes are black, fusiform, obovoid, enclosed in the hardened involucre, with two hooked beaks and hooked bristles [11,12].
This plant is widely distributed all over the world, including Russia, Iran, India, North Korea and Japan. It is native to China and widely distributed in the area of Northeast China, Southwest China, North China, East China and South China. It often grows in plains, hills, mountains and wilderness roadsides. The flowering time ranges from July to August, and fruiting stage lasts from September to October in China [1].

4. Phytochemistry

So far, many phytochemical studies of X. strumarium have been conducted, and more than 170 compounds have been isolated and identified from this plant. Among them, sesquiterpenes and phenylpropanoids are the most abundant and major bioactive constituents in X. strumarium, and are considered as the characteristic constituents of this plant. In addition to the chemical constituents found in fruits, constituents in other parts of X. strumarium were also comprehensively reported, including leaves, roots and stems, etc. In this section, the identified compounds are listed in the following table and the corresponding structures are also comprehensively presented. (Table 2, Figure 2, Figure 3, Figure 4, Figure 5, Figure 6, Figure 7, Figure 8, Figure 9, Figure 10, Figure 11 and Figure 12).

4.1. Sesquiterpenoids and Triterpenoids

Sesquiterpenoids have many important biological functions and physiological activities, which are abundant in X. strumarium. Sesquiterpene lactones, the main characteristic components of plants in the Asteraceae family, exhibit strong activities with anti-microbial, antiviral, anti-tumor and anti-inflammation [55,56]. The predominant sesquiterpene lactones are the guaiane type and seco-guaiane type, of which xanthanolides are the important active constituent. In 2015, eight sesquiterpenes were isolated from the fruits of X. strumarium, including sibirolide A (1), sibirolide B (2) and norxanthantolide A–F (3–8) [13]. In addition, 1β-hydroxyl-5α-chloro-8-epi-xanthatin (9) and 11α, 13-dihydro-8-epi-xanthatin (10) were isolated from the aerial parts of X. strumarium [14]. Moreover, xanthinin (11), xanthumin (12), xanthanol (13), xanthanol acetate (14), isoxanthanol (13), xanthumanol (16), deacetoxylxanthumin (17), xanthatin (18), xanthinosin (19), tomentosin (20) were isolated from the leaves of X. strumarium [15,16]. Furthermore, other sesquiterpenoids were isolated and identified from the fruits, leaves and aerial parts of X. strumarium, including 8-epi-tomentosin (21) [17], 11α,13-dihydroxanthuminol (22), desacetylxanthanol (23) [18], (2E,4E,1’S,2’R,4’S,6’R)-dihydrophaseic acid (24) [19], 8-epi-xanthatin (25) [20], 2-hydroxy xanthinosin (26) [21], lasidiol p-methoxybenzoate (27) [18], 1β,4β, 4α,5α-diepoxyxanth-11(13)-en-12-oic acid (28), 11α,13-dihydroxanthatin (29), 4β, 5β-epoxyxanthatin-1α,4α-endoperoxide (30), 4-epi-xanthanol (31), 4-epi-isoxanthanol (32), 4-oxo-bedfordia acid (33) [22], 2-hydroxytomentosin (34), 2-hydroxytomentosin-1β,5β-epoxide (35) [20], xanthnon (36) [21], 6β,9β-dihydroxy-8-epi-xanthatin (37) [25], inusoniolide (38) [21], (3S,5R,6S,7E)-5,6-epoxy-3-hydroxy-7-megastigmene-9-one (39) [24], pungiolide E (40), pungiolide A (41), pungiolide D (42) [25], 5-azuleneacetic acid (43) [21], dihydrophaseic acid sodium salt 4’-O-β-d-glucopyranoside (44) [26], (3S,5R,6R,7E,9S)-megastigman-7ene-3,5,6,9-tetrol-3-O-β-d- glucopyranoside (45) [27].
Triterpenoids are another important kind of biomolecule found in X. strumarium. Nine triterpenoids including betulinic acid (46), botulin (47), erythrodiol (48) [28], lup-20(29)-en-3β-ol (49) [27], lupenyl acetate (50) [29], lupeol acetate (51) [30], β-amyrin (52), oleanolic acid (53) [31] and α-amyrin (54) [32] are reported from this plant. The chemical structures of these sesquiterpenoids and triterpenoids isolated from X. strumarium are shown in Figure 2 and Figure 3.

4.2. Phenylpropenoids

Phenylpropenoids are also important active constituents found in X. strumarium. To date, 45 phenylpropenoids have been reported in this plant. Phenolic acids, mainly chlorogenic acid, are considered to be the main anti-inflammatory and analgesic active ingredients and the highest content of organic acids [57]. The phenolic acids in X. strumarium contain caffeic acid, ferulic acid, and protocatechuic acid, etc. However, studies have shown that factors such as origin, harvesting time, processing time and temperature have obvious effects on the content of phenolic acid in X. strumarium [58]. Thirteen caffeoylquinic acids (CQA) derivatives were isolated from X. strumarium, including 1,3,5-tri-O-caffeoylquinic acid (55), 3,5-di-O-caffeoylquinic acid (56), neochlorogenic acid methyl ester (57), 1,3-di-O-caffeoylquinic acid (58), methyl-3,5-di-O-caffeoylquinic acid (59), chlorogenic acid (60), 1,4-di-O-caffeoylquinic acid (61), 4,5-di-O-caffeoylquinic acid (62), 5-O-caffeoylquinic acid (63), 1,5-di-O-caffeoylquinic acid (64), 3,4-di-caffeoylquinic acid methyl ester (65), 3,5-di-caffeoylquinic acid methyl ester (66), 4-O-caffeoyl quinic acid methyl ester (67) [33,34,35,36,37,38]. In addition, in 2017, N-trans-feruloyl tyramine (68) and 9,9’-O-di-(E)-feruloyl-(-)-secoisolariciresinol (69) were firstly reported in this plant [39].
Besides, some other phenylpropanoids were also isolated and identificated from this plan, such as xanthiumnolic A (70), xanthiumnolic C (71) [40], 2,3-dihydroxy-1-(4-hydroxy-3-methoxyphenyl)-propan-1-one (72) [41], threo-guaiacylglycerol-8-O-4’-(coniferyl alcohol) ether (73), erythro-guaiacylglycerol-8-O-4’-(coniferyl alcohol) ether (74), threo-1-phenyl-(4-hydroxy-3-methoxy)-2-phenyl-(4’’-hydroxy-3’’-methoxy)-1,3-propanediol (75), (1S,2R)-1,2-bis(4-hydroxy-3- methoxyphenyl)-1,3-propanediol (76), threo-guaiacylglycerol-β-coniferyl aldehyde ether (77), erythro-guaiacylglycerol-β-coniferyl aldehyde ether (78) [42], xanthiumnolic D (79), xanthiumnolic E (80) [40], ferulic acid (81) [43], caffeic acid (82) [36], protocatechuic acid (83) [19], isovanillic acid (84) [30], 7-(4-hydroxy-3-methoxyphenyl)-1-phenylhept-4-en-3-one (85) [28], xanthiazone-(2-O-caffeoyl)-β-d-glucopyranoside (86) [44], rel-(2α,3β)-7-O-methylcedrusin (87) [42], caffeic acid choline ester (88) [38], icariside D1 (89) [45], 3-methoxy-4-hydroxy-transcinnamaldehyde (90) [24], methylchlorogenate (91) [46], icariside F2 (92), arbutin (93), coniferine (94) [45], 3-hydoxy-1-(4-hydroxy-phenyl)-propan-1-one (95) [47], ω-hydroxypropioguaiacone (96) [45], caffeic acid ethyl ester (97) [19], 4-hydroxy-3-methoxycinnamaldehyde (98) [37], p-hydroxybenzaldehyde (99) [24], The chemical structures of these phenylpropenoids isolated from X. strumarium are shown in Figure 4.

4.3. Lignanoids and Coumarins

In recent years, some studies found that X. strumarium contain lignanoids and coumarins, moreover, 21 lignanoids and four coumarins have been discovered in this plant and are displayed in Figure 5 and Figure 6. In 2017, xanthiumnolic B (100) was found from the fruits of X. strumarium and its anti-inflammatory activity has been demonstrated [40]. Later, 14 lignanoids were also isolated from the fruits of X. strumarium, including (-)-1-O-d-glucopyranosyl-2-{2-methoxy-4-[1-(E)-propen-3-ol]phenoxyl} -propane-3-ol (101), leptolepisol D (102), dihydrodehydrodiconiferyl alcohol (103), chushizisin E (104), (-)-(2R)-1-O-d-glucopyranosyl-2-{2-methoxy-4-[(Eformylvinyl]phenoxyl}propane-3-ol (105), (-)-7R,8S-dehydrodiconiferyl alcohol (106), (-)-simulanol (107), 2-(4-hydroxy-3-methoxyphenyl)-3-(2-hydroxy-5-methoxyphenyl)-3-oxo-1-propanol (108), diospyrosin (109), dehydrodiconiferyl alcohol (110), balanophonin A (111), threo-dihydroxydehydrodiconiferyl alcohol (112), 1-(4-hydroxy-3-methoxy)-phenyl-2-[4-(1,2,3-trihydroxypropyl)-2-methoxy]-phenoxy-1,3-propandiol (113), 7R,8S-dihydrodehydrodiconiferyl alcohol 4-O-d-glucopyranoside (114) [48]. Furthermore, syringaresinol (115) [39], fructusol A (116) [42], balanophonin (117) [24], 4-oxopinoresinol (118) [28], pinoresinol (119) [24] were identified from the plant.
In 2011, Kan et al. isolated four coumarins from the roots of X. strumarium for the first time, including scopoletin (120), Jatrocin B (121), cleomiscosin A (122), cleomiscosin C (123) [39].

4.4. Steroids

A few studies have been conducted investigating the steroids in X. strumarium. In 2010, β-sitostenone (124), β-sitosterol (125), daucosterol (126), stigmast-4-en-β-ol-3-one (127), and 5α,8α-epidioxy-22E-ergosta-6,22-dien-3β-ol (128) were isolated from X. strumarium [39]. Furthermore, Chen et al. found 6β-hydroxy-stigmast-4,22-dien-3-one (129), 6β-hydroxy-stigmast-4-en-3-one (130), 3-oxo-Δ4,5-sitostenone (131), β-daucosterol (132), β-stigmastero (133) and 7-ketositosterol (134) from the roots of X. strumarium [28].
Lately, stigmasterol (135), β-sitosterol-3-O-β-d-glucopyranoside (136) [31], ergosterol (137), taraxasteryl acetate (138) [30], 7α-hydroxy-β-sitosterol(stigmast-5-ene-3β,7α-diol) (139), stigmast-4-ene-3β,6α-diol (140) [24] and 14-methyl-12,13-dehydro-sitosterol-heptadeconate (141) [32] were isolated and identified in X. strumarium. The chemical structures of these steroids isolated from X. strumarium are shown in Figure 7.

4.5. Glycosides

In 1962, Song et al. isolated a toxic glycoside component named AA2 from the fruits of X. strumarium, which has been authenticated as atractyloside (142) by Wang in 1983 [49,59]. Subsequently, John et al. found another toxic ingredient known as carboxyatractyloside (143) in 1975 [50]. Research showed that the content of atractyloside in X. strumarium could be reduced after stir-flying, and its toxicity could be reduced. [60] Lately, seven other glycosides were separated from the fruits of X. strumarium, such as 3β-norpinan-2-one 3-O-d-apiofuranosyl-(1→6)-β-d-glucopyranoside (144), (6Z)-3-hydroxymethyl-7-methylocta-1,6-dien-3-ol 8-O-d-glucopyranoside (145), (6E)-3-hydroxymethyl-7-methylocta-1,6-dien-3-ol 8-O-d-glucopyranoside (146), 7-[(β-d-apiofuranosyl-(1→6)-β-d-glucopyranosyl)oxymethy]-8,8-dimethyl-4,8-dihydrobenzo[1,4]thiazine-3,5-dione (147) [41], 3’,4’-dedisulphated-atractyloside (148) [46], 2-methyl-3-buten-2-ol-phated-atractylosideimethy-d-glucopyranoside (149), everlastoside C (150) [51], and all glycosides are displayed in Figure 8.

4.6. Flavonoids

Flavonoids are common chemical components in plants all over the world. Six flavonoids including ononin (151) [43], quercetin (152), allopatuletin (153) [37], patuletin-3-glucuronide (154), quercetin-3-O-glucuronide (155) [34], formononetin (156) [43] have been isolated from this plant and are presented in Figure 9.

4.7. Thiazides

To this day, six thiazides from X. strumarium have been reported. In 1997, xanthiazone (157) was isolated from the aqueous acetone extract of the fruits [36]. Furthermore, 2-hydroxy-xanthiazone (158) [42], 7-hydroxymethyl-8,8-dimethyl-4,8-dihydrobenzol[1,4] thiazine-3,5-dione-11-O-d-glucopyranoside (159), 2-hydroxy-7-hydroxymethyl-8,8-dimethyl-4,8-dihydrobenzol[1,4]thiazine-3,5-dione-11-O-d-glucopyranoside (160) [43], 7-Hydroxymethyl-8,8-dimethyl-4,8-dihydrobenzol[1,4]thiazine-3,5-dione-(2-O-caffeoyl)-β-d-glucopyranoside (161) [52], and xanthialdehyde (162) [53] were identified from this plant (Figure 10).
A few studies have been focused on anthraquinones in X. strumarium. In one report in 2005, Huang et al. found chrysophanic acid (163), emodin (164) and aloe emodin (165) in the fruits of X. strumarium [54]. Then, the 5-hydroxy-3,6-dimethoxy-7-methyl-1,4-naphthalenedione (166), a new naphthoquinone, was isolated from the roots of X. strumarium [28] (Figure 11).

4.8. Other Compounds

Apart from these major types of phytochemical compounds mentioned above, there are some other chemical ingredients isolated from X. strumarium, including 5-methyluracil (167), uracil (168) [39], sibiricumthionol (169) [19], indole-3-carbaldehyde (170) [45], N-(1’-d-deoxyxylitolyl)-6,7-dimethyl-1,4-dihydro-2,3-quinoxalinedione (171) [38], nonadecanoic acid (172) [39], hexadecanoic acid (173) [32] (Figure 12).

5. Pharmacology

5.1. Anti-AR Effect

X. strumarium is a traditional medicine widely used in the treatment of nasal diseases, especially allergic rhinitis (AR). In modern pharmacological study, the mechanism of X. strumarium in treating AR has been studied extensively. In 2003, it was reported that WEX inhibited compound 48/80 (C 48/80)-induced systemic anaphylaxis in mice (0.01 to 1 g/kg, p.o.), and the mechanism may be related to the inhibition of histamine and TNF-α released from rat peritoneal mast cells (RPMC) [61,62]. In 2008, Zhao et al. found that WEX (0.25–1 mg/mL) can modulate the human mast cell-mediated and peripheral blood mononuclear cell (PBMNC)-mediated inflammatory and immunological reactions which induced by pro-inflammatory cytokines including interleukin (IL)-4, IL-6, IL-8, GM-CSF and TNF-α [63]. Furthermore, the MEX is found to possess the inhibitory effect on the activation of C 48/80 stimulated mast cells, and the mechanism was correlated to inhibit Ca2+ uptake and histamine release, and increase cAMP in RPMC [64]. In addition, in 2014, Peng et al. demonstrated that the caffeoylxanthiazonoside (CXT) (5, 10, 20 mg/kg, p.o.) isolated from the fruits of X. strumarium was helpful to alleviate the nasal symptoms of ovalbumin (OVA) induced AR rats via anti-allergic, down-regulating IgE, anti-inflammatory and analgesic properties [65].

5.2. Anti-Tumor Effect

Anti-tumor effects are also regarded as primary pharmacological properties of X. strumarium, and have been extensively investigated in lung cancer, breast cancer, cervical cancer, colon cancer, liver cancer, meningioma, and leukemia.
Tao et al. studied the inhibitory effect of xanthatin (1–40 μM), an active agent in X. strumarium, against lung cancer cells (Cell lines of A549, H1975, H1299, H1650 and HCC827) and its potential mechanisms [66,67]. It found that xanthatin could downregulate the STAT3, GSK3β and β-catenin, moreover, xanthatin could also trigger Chk1-mediated DNA damage and destabilize Cdc25C via lysosomal degradation [66,67,68]. In 1995, Ahn et al. isolated three cytotoxic compounds from the leaves of X. strumarium, among them, xanthatin and 8-epi-xanthatin possessed obvious anti-tumor activity on A549 cells with IC50 (half maximal inhibitory concentration) values of 1.3 and 1.1 μg/mL, respectively [17]. Later, in 2002, it was reported that 1,8-epi-xanthatin epoxide has notable anti-tumor effect against A549 cells with IC50 value of 3.0 μM [69]. Furthermore, Wang et al. and Ferrer et al. reported that 8-epi-xanthatin-1α,5α-epoxide, 1β-hydroxyl-5α-chloro-8-epi-xanthatin and EEXA can inhibit the proliferation of A549 cells (IC50 = 9.5 μM, 20.7 μM and 52.2 μg/mL, respectively) [25,70].
In 2007, by using CellTiter 96 assay in vitro, Ramı’rez-Erosa et al. found that xanthatin and xanthinosin, two sesquiterpene lactones isolated from the burs of X. strumarium, obviously restrain the proliferation of breast cancer MDA-MB-231 cells with the IC50 values of 13.9 and 4.8 μg/mL, respectively [71]. Furthermore, Takeda et al. studied the mechanism of xanthatin against breast cancer MDA-MB-231 cells in 2011, and the results indicated that xanthatin (5–25 μM) inhibits cell growth via inducing caspase independent cell death which were irrelevant with FTase inhibition [72]. In addition, xanthatin (2.5–10 μM) can also up-regulate GADD45 γ tumor suppressor gene, and induce the prolonged expression of c-Fos via N-acetyl-l-cysteine-sensitive mechanism [73,74]. In 2016, the anti-tumor activity of EEXA on MFC7 cells was reported as well, with an IC50 value of 70.6 μg/mL [70].
In 2015, Vaishnav et al. demonstrated that WEX with a concentration of 12.5–50 μg/mL were able to induce death in HeLa cervical cancer cells by altering the antioxidant levels [75]. Recently, Liu et al. revealed that xanthatin (5–20 µM) targeted the selenocysteine (Sec) residue of thioredoxin reductase (TrxR) and inhibited the enzyme activity irreversibly [76]. Meanwhile, the inhibition of TrxR by xanthatin promoted oxidative stress-mediated apoptosis of HeLa cells.
In 1995, Ahn et al. reported that xanthatin and 8-epi-xanthatin were remarkably cytotoxic to colon cancer HCT-15 cells with ED50 (median effective dose) values of 1.1 and 0.1 μg/mL, respectively [17]. Later, in 2007, Ramı’rez-Erosa et al. (2007) found that xanthatin (IC50 = 6.15 μg/mL) and xanthinosin (IC50 = 6.15 μg/mL) possessed the function of inhibiting WiDr cells growth [71]. Furthermore, eremophil-1(10),11(13)-dien-12,8β-olide,8-epi-xanthatin-1β,5β-epoxide and tomentosin were isolated from the aerial parts of X. strumarium, and their anti-tumor activities on BGC-823 cells and KE-97 cells were aslo determined. The related results showed that the IC50 values of three compounds on BGC-823 cells are 13.22, 2.43, and 4.54 µM, respectively. Similarly, IC50 values of three compounds on BGC-823 cells are 4.41, 1.44, and 3.47 µM, respectively [77]. Moreover, Zhang et al. reported that xanthatin (3.9–18.6 µM) inhibited the proliferation of MKN-45 cells by inducing G2/M cell cycle arrest and apoptosis [78]. Later, in 2015, Karmakar et al. found that xanthinosin (8 µM) and lasidiol p-methoxybenzoate (16 µM) potentiate both extrinsic and intrinsic TRAIL-mediated apoptosis pathways and also decreased the level of cell survival protein Bcl-2 in AGS cells [20]. Simultaneously, fructusnoid C (IC50 = 7.6 µM) also reported to exhibit cytotoxic effects on AGS cells [79]. EEXA and CFEEXA have been identified as the active ingredients against the growth of CT26 cells with IC50 values of 58.9 and 25.3 μg/mL, respectively [70].
Furthermore, the anti-tumor effects of X. strumarium on liver cancers have also been reported in recent years. In 2013, Wang et al. found that the 1β-hydroxyl-5α-chloro-8-epi-xanthatin possessed significant in vitro cytotoxicity with an IC50 value of 5.1 µM against SNU387 cells [25]. Later, in 2017, the cytotoxic effects of MEX and EAFMEX on HepG2 cells were verified as LC50 (Lethal Concentration 50) values of 112.9 and 68.739 μg/mL [80]. Furthermore, Liu et al. demonstrated that xanthatin (5–40 μM) can induce HepG2 cells apoptosis by inhibiting thioredoxin reductase and eliciting oxidative stress [76].
Additionally, an investigation in 1995 indicated that Xanthatin and 8-epi-xanthatin both have cytotoxic effects on SK-MEL-2 cells with ED50 values 0.5 and 0.2 μg/mL, respectively [17]. In 2012, the EEXS showed notable inhibitory activity on Mel-Ab cells through downregulation of tyrosinase via GSK3β phosphorylation at concentrations of 1–50 μg/mL [81]. Later, in 2013, Li et al. reported the anti-tumor effects of xanthatin both in vitro and in vivo. Previous results showed that xanthatin (2.5–40 μM) possess a remarkable anti-proliferative effect against B16-F10 cells, and the related mechanism probably associated with activation of Wnt/β-catenin pathway as well as inhibition of angiogenesis. Meanwhile, the in vivo evidence in mice (xanthatin, 0.1–0.4 mg/10 g, i.p.) also verified the results mentioned above [82].
In 1994, DFEEXA was reported to be toxic to leukemia P-388 cells with an IC50 value of 1.64 μg/mL [83]. In addition, results of Nibret et al. showed that xanthatin has significant cytotoxic on HL-60 cells in 2011 [84]. Another report in 2017 reported that both MEX and EAFMEX have inhibitory effects on Jurkat cells, and EAFMEX showed higher toxicity to Jurkat cells when compared to MEX [80].
Besides, in 1995, Ahn et al. found that xanthatin and 8-epi-xanthatin have cytotoxic effects on CNS carcinoma XF-498 cells, and the ED50 values were 1.7 and 1.3 μg/mL, respectively [17]. In 2013, Pan et al. reported that WEX can cause significant cytotoxic effects on arcoma S180 cells in vivo (S180 cells bearing mice, 5–20 g/kg) [85]. The in vitro anti-proliferative activity of CEXR and MEXR on laryngeal cancer HEP-2 cells were implemented at doses of 12.5–100 µg/mL, and the two extracts of X. strumarium showed potent cytotoxic activities against the HEP-2 cells [86].

5.3. Anti-Inflammatory and Analgesic Effects

In 2004, it was reported that WEX (10, 100 and 1000 µg/mL) inhibited inflammatory responses in Lipopolysaccharide (LPS)-stimulated mouse peritoneal macrophages via decreasing IFN-γ, LPS-induced NO production and TNF-α production in a dose dependent manner [87]. Furthermore, in 2005, Kim et al. evaluated the anti-inflammatory and anti-nociceptive activities of MEX both in vitro and in vivo, it showed that the MEX (30, 60 and 90 mg/mL) can down-regulate the production of NO, PGE 2 and TNF-α, and MEX treatment (100 and 200 mg/kg/day, p.o.) clearly reduced carrageenan induced hind paw edema in rats [88]. In addition, MEX (100 and 200 mg/kg/day, p.o.) significantly reduced the amount of writhing induced by acetic acid, and increased jumping response latency in a hot plate test. Later, in 2008, xanthatin and xanthinosin were reported to inhibit LPS-induced inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) expression in microglial BV-2 cells with IC50 values of 0.47 and 11.2 μM, respectively [89]. By using LPS inhibition assay and animal model of inflammation (carrageenan induced hind paw edema), the MEXL (100, 200 and 400 mg/kg) showed obvious anti-inflammatory activity both in vitro (IC50 = 87 μg/mL) and in vivo [90]. A report in 2015 showed that MEXR (50–400μg/mL) can suppress inflammatory responses via the inhibition of nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) in LPS-induced murine macrophages [91]. Moreover, the WEX was found to restrain LPS-induced inflammatory responses through suppressing NF-κB activation, inhibiting JNK/p38 MAPK phosphorylation, and enhancing HO-1 expression in macrophages [92]. In 2016, Hossen et al. demonstrated that the inhibitory effect of MEX on the inflammatory disease possibly related to signaling inhibition of MAPK and AP-1 [93]. In another study, Hossen et al. found the potential anti-inflammatory activity of MEXA on LPS-treated macrophages and an HCl/EtOH-induced mouse model of gastritis by inhibiting PDK1 kinase activity and blocking signaling to its downstream transcription factor, NF-κB [94]. Later, in 2017, Jiang et al. found a new phenylpropanoid derivative named Xanthiumnolic E isolated from X. strumarium, which has notable inhibitory effect on LPS-induced nitric oxide (NO) production with IC50 value of 8.73 μM [26].
Additionally, X. strumarium was confirmed to inhibit some other kinds of inflammatory and painful diseases. In 2011, Huang et al. suggested that WEX inhibited the development of paw edema induced by carrageenan, and exhibited inhibitory activity on acetic acid effect and reduced the formalin effect at the late-phase (0.1, 0.5 and 1.0 g/kg, p.o.) [95]. In addition, the NFEEX at doses of 0.5, 0.75 and 1.0 mg/ear showed strong anti-inflammatory activity in the croton-oil-induced ear edema test, and reduced the amount of writhing induced by acetic acid in mice in a dose-dependent manner (100, 200 and 400 mg/kg) [96]. A report in 2011 demonstrated the anti-inflammatory activity of xanthatin by inhibiting both PGE 2 synthesis and 5-lipoxygenase activity at doses of 100 and 97 mg/mL, respectively [84]. Furthermore, Park et al. first explained the anti-inflammatory mechanism of EEX, which inhibited TNF-α/IFN-γ-induced expression of Th2 chemokines (TARC and MDC) by blocking the activation of the NF-κB, STAT1 and ERK-MAPK pathways in HaCaT keratinocytes [97]. The hot plate test, acetic acid induced writhing test and formalin test were applied to evaluate the analgesic activity of EEX, and it showed significant analgesic activity at concentrations of 250 and 500 mg/kg body weight [98].

5.4. Insecticide and Antiparasitic Effects

In 1995, Talakal et al. reported that EEXL possess anti-plasmodial activity against Trypanosoma evansi both in vitro and in vivo. The EEXL exhibited trypanocidal activity at all the four tested doses at 5, 50, 500 and 1000 µg/mL in vitro, and it can significantly prolong the survival period of the T. evansi infected mice at concentrations of 100, 300 and 1000 mg/kg [99]. In 2011, xanthatin was demonstrated to be the dominating insecticidal active compound against Trypanosoma brucei brucei with an IC50 value of 2.63mg/mL and a selectivity index of 20 [84]. In addition, Go¨kce et al. showed that MEX exhibited both ingestion toxicity and ovicidal activity to Paralobesia viteana with an LC50 of 11.02% (w/w) [100]. In 2012, by using schizont inhibition assay, the anti-plasmodial activity of EEXL against Plasmodium berghei was assessed, and it showed significant activity (IC50 = 4 µg/mL) and high selectivity index in vitro [101]. Later, in 2014, Roy et al. found that WEXL had distinct insecticidal properties against Callosobruchus chinensis with strong toxicity, repellent properties, inhibited fecundity and adult emergence of the insects at 1%, 2% and 4% concentrations [102]. Moreover, it is reported that EEX revealed anti-nematode activity against Meloidogyne javanica in inhibiting egg hatching and inducing mortality among second stage juveniles (J2s) [103]. Furthermore, the effect of MEX on the mortality rates of Aedes caspius and Culex pipiens were investigated, and the results revealed that the LC50 values of MEX were found to be 531.07 and 502.32 μg/mL against A. caspius and C. pipiens, respectively [80].

5.5. Antioxidant Effect

In 2010, it was reported that CEXR and MEXR showed significant free radical scavenging activity by 1,1-diphenyl-2-picrylhydrazyl (DPPH) method with LC50 values of 10.28 and 40.40 µg/mL, respectively [86]. After administration of PEEXW (250 and 500 mg/kg, p.o., for 20 days), the contents of superoxide dismutase, glutathione peroxidase, glutathione reductase and catalase significantly increased in rats’ brain [104]. Later, in 2011, Huang et al. found that WEX exhibited 70.6% to 76.4% and 35.2% to 79.1% scavenging activity on 2,2’-Azinobis-(3-ethylbenzthiazoline-6-sulphonate) (ABTS) radicals and DPPH radical scavenging in the concentration of 0.05–0.2 mg/mL; simultaneously, the reducing activity of WEX increased and liposome protection effect enhanced in a concentration-dependent manner with the same doses [95]. In the treatment with the MEXS (100 and 200 mg/kg, p.o. for 10 days), the contents of SOD, CAT, GSH and GPx were obviously increased in the diabetic rats’ tissues [105]. Moreover, in 2011, Sridharamurthy et al. evaluated the antioxidant effect of EEXR and CEXR by the scavenging activity of free radicals such as DPPH, super oxide, nitric oxide, and hydrogen peroxide [106]. Results showed that the IC50 values of EEXR were 29.81, 495.30, 395.20 and 10.18 µg/mL, respectively, and the IC50 values of CEXR were 24.85, 418.30, 415.18 and 9.23 µg/mL, respectively. In addition, Kamboj et al. demonstrated that EEXL possessed strong scavenging capacity against DPPH, nitric oxide and hydrogen peroxide with IC50 values of 85, 72 and 62 µg/mL. In addition, the antioxidant activity was possibly due to the presence of compounds in the extracts like flavonoid and phenolic [107]. In 2015, hexadecanoic acid, α-amyrin and 14-methyl-12,13-dehydro-sitosterol-heptadeconate were isolated from the leaves of X. strumarium, and their antioxidant potential was also evaluated. These three chemical components showed significant antioxidant activity in a dose dependent manner by DPPH and hydroxyl radical assay methods with the IC50 values of 106.4, 64.16, 76.18 µg/mL and 127.4, 83.96 and 84.4 µg/mL, respectively [32]. A study in 2017 revealed that the EOX displayed notable activity for DPPH radicals with an IC50 value of 138.87 μg/mL [108]. Furthermore, the antioxidant effects of the MEX obtained by the response surface methodology were measured by the scavenging activity towards the DPPH radical and Ferric ion reducing antioxidant power (FRAP). These results showed that methanol concentration and solid to solvent ratio were demonstrated to possess obvious effects on DPPH and FRAP values [28].

5.6. Antibacterial and Antifungal Effects

In 1983, Mehta et al. reported that the WEXFT possessed antimicrobial properties against Vibrio cholera [109]. Later, a study in 1997 revealed that the xanthatin isolated from the leaves of X. strumarium had notable potent activities against Staphylococus epidermidis, Bacillus cereus, Klebsiella pneumoniae, Pseudomonas aeruginosa and Salmonella fyphi with minimum inhibitory concentration (MIC) values of 31.3, 62.5, 31.3, 125 and 125 µg/mL, respectively [110]. In addition, it is reported that MEXL (500 and 100 mg/mL) exhibited strong activity against K. pneumoniae, Proteus vulgaris, P. aeruginosa, Pseudomonas putida, Salmonella typhimurium, B. cereus, Bacillus subtilis and S. epidermidis [111]. In 2015, Chen et al. also reported that β-sitosterol and β-daucosterol isolated from the X. strumarium have significant inhibitory effects against Escherichia coli, with MIC values of 0.17 and 0.35 mg/mL, respectively [112]. By using the disc diffusion method, Devkota et al. determined the antibacterial activity of MEXL and WEXL, and results showed that the two extracts inhibited growth towards K. pneumoniae, Proteus mirabilis, E. coli, B. subtilis, Enterococcus faecalis and Staphylococcus aureus at concentrations of 50, 100, 150, 200 and 250 mg/mL [113]. Moreover, Sharifi-Rad et al. revealed that EOXL can significantly suppress the growth of S. aureus, B. subtilis, K. pneumoniae and P. aeruginosa with MIC values of 0.5, 1.3, 4.8 and 20.5 µg/mL, respectively; additionally, EOXL (30, 60 and 120 mg/mL) also exhibited obvious antibacterial activity against Shiga toxin-producing Escherichia coli [114,115]. Furthermore, Wang et al. revealed that WEX possessed antibacterial potentials against S. aureus and E. coli with MIC values of 31.25 and 7.81 mg/mL, respectively [116]. Using the disk diffusion, the antibacterial activity of EOXF on Rathayibacter toxicus and Pyricularia oryzae was evaluated, and the MIC values were 25 and 12.5 µg/mL, respectively [108].
Similar to the antibacterial potentials, the antifungal activities of X. strumarium were also deeply investigated. In the year of 2002, Kim et al. found an antifungal constituent from X. strumarium, which was named deacetylxanthumin. It can inhibit mycelial growth and zoospore germination of Phytophthora drechsleri with a MIC value of 12.5 µg/mL [117]. In 2011, Yanar et al. used radial growth technique to test the antifungal activities of MEX against Phytophthora infestans, and the MEX showed the lowest MIC value of 2.0% w/v which was lower than the standard fungicide (Metalaxyl 4% + Mancuzeb 64%, MIC value was 2.5%, w/v) [118]. Later, in 2015, Sharifi-Rad et al. investigated the antifungal ability of EOXL on Candida albicans and Aspergillus niger, and the MIC values were 55.2 and 34.3 µg/mL, respectively [114]. In vitro, using the disk diffusion method, the EOXL exhibited strong inhibition against Pyricularia oryzae and Fusarium oxysporum with MIC values of 12.5 and 50 µg/mL, respectively [108]. Furthermore, the EOXL showed remarkable growth inhibition of a wide spectrum of fungal strains, such as A. niger, Aspergillus flavus, F. oxysporum, Fusarium solani, Alternaria alternata and Penicillium digitatum with both MIC and MBC (minimum bactericidal concentration) values of 8 µg/mL [119].

5.7. Antidiabetic Effect

In 1974, Kupiecki et al. found that the WEX (15 and 30 mg/kg, i.p.) exhibited potent hypoglycemic activity in normal rats in a dose-dependent manner [120]. In 2000, the antidiabetic effect of caffeic acid isolated from X. strumarium was investigated on both streptozotocin-induced and insulin-resistant rat models. The results showed that caffeic acid (0.5–3.0 mg/kg, i.v.) can decrease the plasma glucose level via increasing the glucose utilization [121]. In 2011, Narendiran et al. found that MEXS at the doses of 100 and 200 mg/kg (p.o., for 30 days) had remarkable diabetic activity in normal-glycemic and streptazocin induced hyperglycemic rats [105]. A report in 2013 demonstrated that the methyl-3,5-di-O-caffeoylquinate showed strong ability to counteract diabetic complications via competitive inhibition of aldose reductase (AR) and galactitol formation in rat lenses [47]. In addition, it is reported that the CFMEXL exhibited notable inhibitory activity on α-glucosidase enzyme with the IC50 value of 72 µg/mL [122]. Similarly, another study found that MEX also had a strong α-glucosidase inhibitory effect with IC50 value of 15.25 µg/mL [28].

5.8. Antilipidemic Effect

Recently, investigations into the antilipidemic effects of X. strumarium have been conducted. In 2011, the CEXR and EEXR were evaluated for anti-lipidemic activity in Triton WR-1339 induced hyperlipidemia in Swiss albino rats. The results showed that CEXR and EEXR (200 and 400 mg/kg p.o.) can significantly decrease the contents of plasma cholesterol, TG, LDL, and VLDL and increase plasma HDL levels, which was possiblely related to their significant antioxidant activity [106]. Later, in 2016, Li et al. found that WEX (570 and 1140 mg/kg, p.o., for 6 weeks) could improve the synthesis of fatty acid and TG, thus decreased the circulating free fatty acid (FFA) levels, indicating that WEX is involved in solving the abnormality of FFA in the circulation, which is executed by promoting the storage of the excess fat, rather than the elimination of added fat [123]. Furthermore, after treatment with WEX (3.7 and 11.11 g/kg, p.o., for 4 weeks), the blood glucose, TC, TG, LDLC levels decreased and HDLC levels increased in diabetic mice [124].

5.9. Antiviral Activity

In 2009, it was reported that the WEX (0.01, 0.1 and 1.0 g/kg, i.g., for 10 days) possessed antiviral activity against duck hepatitis B virus, and it can delay pathological changes [125]. In addition, five compounds were isolated from the fruits of X. strumarium, and their antiviral abilities were also evaluated. The results indicated that norxanthantolide F, 2-desoxy-6-epi-parthemollin, xanthatin, threo-guaiacylglycerol-8′-vanillic acid ether and caffeic acid ethyl ester exhibited notable activity against influenza A virus with IC50 values of 6.4, 8.6, 8.4, 8.4 and 3.7 µM, respectively by a cytopathic effect (CPE) inhibition method [13].

5.10. Other Pharmacological Effects

Apart from the pharmacological effects displayed above, X. strumarium also possesses some other activities. In 2016, the CXT (10, 20, and 40 mg/kg, i.p.) isolated from fruits of X. strumarium showed significant anti-septic activity in animal models of Cecal ligation and puncture (CLP) operation. Meanwhile, the CXT can increase survival rates of septic mice induced by CLP and decrease TNF-α and IL-6 levels induced by LPS in serum of mice [126]. After treatment with WEX (570 and 1140 mg/kg p.o., for 6 weeks), the glucose tolerance and insulin sensitivity improved, meanwhile, lipogenesis increases and lipid oxidation decreased in the liver of high-fat diet rats [127]. In 2014, Lin et al. demonstrated that the EEX (75 and 300 mg/kg, p.o.) can significantly inhibit paw swelling and arthritic score and increase body weight loss and decrease the thymus index in animal model of rheumatoid arthritis induced by Complete Freund’s Adjuvant (CFA) [128]. Moreover, the overproduction of TNF-α and IL-1β was notably suppressed in the serum of all EEX-treated rats. The anti-pyretic activity of MEXW (200 and 400 mg/kg, p.o.) was estimated on yeast induced hyperpyrexia, and it showed significant reduction in elevated body temperature [129]. Using Maximal Electroshock (MES) and Pentylenetetrazole (PTZ) induced seizures models, the anticonvulsant activity of PEEXW was tested, and results showed that PEEXW can reduce the mean duration of extensor phase and delay onset of myoclonic spasm and clonic convulsion of treated groups at doses of 250 and 500 mg/kg [130]. In 2016, Panigrah et al. explored the antiurolithiatic effect of HEEXB, and showed that HEEXB can restore the impairment induced by ethylene glycol including hyperoxaluria, crystalluria, hypocalciuria, polyurea, raised serum urea, creatinine, erythrocytic lipid peroxidise and nitric oxide, kidney calcium content as well as crystal deposition. The mechanism may be related to inhibition of various pathways involved in renal calcium oxalate formation, antioxidant property and down regulation of matrix glycoprotein, osteopontin (OPN) [131]. A report in 2012 indicated the antiulcer effect of EEXL in pylorus ligation induced gastric ulcers, and its gastro-protective mechanism may be due to DNA repair, free radical scavenging and down regulation of oxidativenitrosative stress along with cytokines [132]. In an in vivo study, with the CXT treatment (10, 20 and 40 mg/kg, p.o.), the cardiac hypertrophy reduced and fractional shortening (FS), ejection fraction (EF), cardiac output (CO) and heart rate (HR) reversed via suppressing the expression of pro-inflammatory cytokines and the NF-κB signaling pathway [133].

5.11. Summary of Pharmacologic Effects

In conclusion, X. strumarium has a wide range of pharmacological effects including anti-AR effects, anti-tumor effects, anti-inflammatory and analgesic effects, insecticide and antiparasitic effects, antioxidant effects, antibacterial and antifungal effects, antidiabetic effects, antilipidemic effects, and antiviral effects. (Table 3). It is noteworthy that the research areas of modern pharmacy primarily focus on chemical components and extracts, which indicated the promising potential of X. strumarium for treating disease. Nevertheless, the chemical constituents and corresponding pharmacological effects of X. strumarium are not systematically sorted out and analyzed. Therefore, it is necessary to investigate the pharmacological activity, structure-activity relationship and mechanism of X. strumarium both in vitro and in vivo experiments in the future.

6. Pharmacokinetics

Up to now, there are few reports on the pharmacokinetics of the extracts or monomers of X. strumarium. Previous pharmacokinetics studies of X. strumarium mainly focused on its active compounds including xanthatin, cryptochlorogenic acid, and toxic ingredient such as atractyloside. In 2014, a sensitive, specific and rapid ultra-high performance liquid chromatography (UHPLC) tandem mass spectrometry (UHPLC-MS/MS) method was applied to research pharmacokinetic properties of xanthatin in rat plasma. After intravenous injection of xanthatin at a dose of 2.4 mg/200 g, 4.8 mg/200 g and 9.6 mg/200 g, respectively. The t1/2 of three concentrations were found to be 108.58 ± 32.82, 123.50 ± 66.69, and 181.71 ± 148.26 min, respectively; and the peak plasma concentration (Cmax) values were 418.72 ± 137.51, 904.89 ± 193.53, and 1773.46 ± 1733.10 ng/mL, respectively. As the dose increased, the AUC0–t and AUC0–∞ were gradually enlarged, and the AUC0–t of three doses were 14,340.20 ± 7122.41, 32,149.52 ± 11,259.44, and 49,524.28 ± 28,520.88 n gh/mL, respectively; furthermore, the AUC0–∞ of three levels are 15,538.97 ± 7733.12, 36,431.22 ± 14,498.16, and 61,885.45 ± 30,704.80 n gh/mL, respectively. In addition, the total body CL were 0.13 ± 0.14, 0.17 ± 0.11, 0.22 ± 0.13 mL/min and Vd were 46.85 ± 20.19, 159.99 ± 30.49, and 208.22 ± 85.97 mL of three concentrations [134].
After intragastric administration of the atractyloside at doses of 11.4, 22.8, and 45.6 mg/kg, the peak time (Tmax) values were determined to be 0.38, 1.85, 0.27 h, respectively, the t1/2 were 13.64, 9.62, 8.61 h, respectively, and the peak plasma concentration (Cmax) values were 41.98, 24.61, 263.40 µg/mL, respectively. In addition, the area under the concentration-time curve (AUC) was also determined, and the AUC0–t was 132.70, 222.90, and 345.20 µ gh/L. The results showed that the toxicokinetic behavior of atractyloside in rats was non-linear within the experimental dose range [135].
Furthermore, Shen et al. studied the pharmacokinetics of neochlorogenic acid and cryptochlorogenic acid in X. strumarium and its processed products after intragastric administration in rats. The results showed that the Tmax of neochlorogenic acid and cryptochlorogenic acid in processed fruits of X. strumarium were 2.94 ± 0.18, and 3.00 ± 0.46 h, respectively; the t1/2 of neochlorogenic acid and cryptochlorogenic acid in processed fruits of X. strumarium were 2.35 ± 1.11, 1.97 ± 0.66 h. Moreover, the Tmax of neochlorogenic acid and cryptochlorogenic acid in raw fruits of X. strumarium were 3.75 ± 0.46, 2.75 ± 0.27 h, and the t1/2 of neochlorogenic acid and cryptochlorogenic acid in raw fruits of X. strumarium were 1.70 ± 0.61, 2.12 ± 0.68 h. The neochlorogenic acid in fruits of X. strumarium, after being processed, takes effect quickly and lasts for a long time, while the cryptochlorogenic acid takes effect slowly and has a short action time [136].

7. Toxicity

In 1990, it was reported that X. strumarium has medium to strong allergenic effects and is poisonous to mammals, and atractyloside and carboxyatractyloside are considered to be the major toxic compounds [137]. X. strumarium is prudently ranked into the medium grade with less toxicity in the Shennong Bencao Jing, a monograph of materia medica. Some other Chinese materia medicas aslo record that X. strumarium possessed mild toxicity, such as Bencao Pinhui Jingyao, Bencao Huiyan. Thus, it is obvious that the ancient Chinese people have had a clear understanding of the toxicity of X. strumarium for a long time [138].
In recent years, many investigations have indicated the toxic effects and related mechanisms of the extracts and monomers of X. strumarium (Table 4). In 2005, Li et al. found that the median lethal concentration (LD50) value of the WEX in mice was 201.14 g/kg (i.g., crude herbs mass equal) [139]. In addition, a report in 2012 suggested that the LD50 value of the WEX in mice was 167.60 g/kg (crude herbs mass equal, i.g.), however the LD50 value was 194.15 g/kg (i.g., crude herb mass equivalent) in Fu’s research report [140,141]. These changes can be attributed to the toxicity of X. strumarium which varied with the processing method, genetic characteristics and growing conditions [138]. Furthermore, the LD50 value of the EEX in mice was 275.41 g/kg (crude herbs mass equal, i.g.), which was higher than WEX [140]. Another study showed that the carboxyatractyloside (10–100 mg, i.v.) can induce death in swine [142].
Recently, animal experiments and clinical studies on X. strumarium showed that hepatotoxicity is the main toxicity. In 2011, Wang et al. demonstrated that kaurene glycosides including atractylosid (50–200 mg/kg, i.p.) and carbxyatractyloside (50–150 mg/kg, i.p.) induced hepatotoxicity in mice by way of its induction of oxidative stress as lipid peroxidation in liver [143]. Besides, the chief mechanism of atractyloside poisoning is deemed to be inhibition of the mitochondrial ADP transporter [144]. Furthermore, the WFEEX and NFEEX (0.06, 0.3, 0.7 g/kg, i.g., for 28 days), which have marked hepatotoxicity to rats, can cause pathological changes, such as enlarged hepatic cell space, karyolysis, and inflammatory cell infiltration [145]. Moreover, it has been reported that WEX (21.0 g/kg i.g., for 28 days) significantly increased the content of ALT, AST in mice serum and decreased weight loss [146]. In addition, a study in 2014 found that WEX (7.5, 15.0 and 30.0 g/kg, i.g., for 5 days) can increased the serum ALT, AST, ALP, TBIL levels and the contents of LDL/vLDL, β-HB, glutamate, choline, acetate, glucose in male rats [147]. Finally, in 2018, Zeng et al. indicated that the contents of GLDH, α-GST increased and miRNA-122 decreased after administered WEX (16.7 g/kg i.g., for 7 days), which can be used as sensitive biomarkers for studying the regularity of hepatotoxicity of X. strumarium [148]. Apart from hepatotoxicity, Mandal et al. studied the neurotoxicity of the MEXA in mice and results show that MEXA (100, 200, 300 mg/kg) can obviously depress the action of central nervous system [149].
Many other studies have demonstrated that different medicinal parts and extraction parts are also cytotoxic to normal cells including hepatocytes, nephrocytes, ovary cells, etc. The cell inhibition ability of atractyloside on rat hepatocytes was investigated, and the results demonstrated that atractyloside (0.01–0.05 g/L) induced dose-dependent hepatotoxicity according to obvious decreases of cell viability, intracellular gluta-thione (GSH) content and albumin secretion [150]. Furthermore, atractyloside and carbxyatractyloside was reported to improve LDH activity and inhibit cell proliferation at the concentration of 100 μmol/L [147]. In 2013, Yu et al. indicated that WEX at concentrations 100 μg/mL can inhibit growth of HK-2 cells [151]. Moreover, HEXA (25–100 μg/mL) also causes in vitro DNA damage at cytotoxic concentrations through sister chromatid exchanges, chromosome aberrations, and comet assay, meanwhile, it also shows significant reduction in CHO cell viability [152]. In 2016, Su et al. compared the cytotoxicities of the components with different polarities, and study indicated that EAFEEX (IC50 = 231.1 μg/mL) was the most toxic part [153].
In recent years, few investigations have focused on the toxic effects of X. strumarium on reproduction. In 2014, it was reported that the WEX possessed reproductive toxicity to zebrafish embryos, including decreases in hatch rate, and increases in mortality rate, heart rate and swimming speed [154].

8. Future Perspectives and Conclusions

In summary, X. strumarium, which possesses anti-AR effects, anti-inflammatory and analgesic effects and anti-tumor effects, has been widely applied to clinical practice in many countries. In the meantime, many modern studies on X. strumarium were also carried out, and its pharmacological activities and chemical compositions have been preliminarily investigated. Nevertheless, how to find out the mechanism of pharmacological activities and its related compounds, develop clinical efficacy of X. strumarium and ensure medication safety are still extremely crucial now.
First, the chemical compounds and pharmacological activity studies of X. strumarium mainly focused on its fruits, but there are few investigations on the roots, leaves, stems and other parts of X. strumarium. In order to enlarge the source domain of the active compounds and maximize the plant utilization rate, it is very critical for researchers to conduct a comprehensive evaluation of other parts of this plant. Second, the fruits of X. strumarium are officially recognized as Cang-Er-Zi in the Chinese Pharmacopoeia (2015 Edition), but many other Xanthium species such as X. mongolicum Kitag, Xanthium spinosum L. and Xanthium canadens Mill were used as X. strumarium alternatives in many areas of China. Therefore, the physical properties, chemical compositions and pharmacological activities should be used to identify and differentiate the different varieties, and it is important to guarantee the safety and efficacy with these herbs to ensure its suitability for clinical use. Third, in China, X. strumarium is commonly used after processing in clinical medicine, but the mechanism of its detoxification still needs further study. The degree of processing depends mainly on the subjective experience of people, and it is difficult to ensure the consistency of the quality of Chinese Medicine. Thus, the intelligent sensory technology combined with artificial intelligence technology, such as machine vision, electronic nose and electronic tongue can be applied to standardize processing methods. Fourth, on the basis of current research progress in vivo and in vitro, many active compounds of X. strumarium have been found and identified, which are probably developed into effective drugs. Among them, xanthatin possessed strong anticancer activity against many kinds of tumors, which means that it has the potential to become an anticancer drug in the future. However, systematic investigations on pharmacokinetics, target-organ toxicity and clinical research of xanthatin will help to develop its bioactive constituents as novel drugs. Fifth, traditional Chinese medicine has the characteristics of multi-component, multi-target and multi-channel, and a single component cannot completely reveal its pharmacological activity. Recently, quality marker (Q-Markers) technologies have started to contribute to scientifically interpreting the correlation degree of effectiveness-material basis-quality control of significant components in traditional Chinese Medicine. For X. strumarium, Q-Markers technologies are able to clarify its possible action, toxicity mechanism and symbolic components, and it is helpful to establish the whole quality control and quality traceability system of X. strumarium.

Author Contributions

Conceptualization, W.P. and C.W.; writing—original draft preparation, W.F., L.F., C.P., Q.Z., L.W., L.L., J.W.; writing—review and editing, W.P. and D.Z.; funding acquisition, W.P. and C.W.

Funding

This work was funded by the China Postdoctoral Science Foundation (no. 2018M631071); Innovative Research Team of Chinese Medicine Discipline in Chengdu University of Traditional Chinese Medicine (no.030041007) and Sichuan Science and Technology Project (no.2018JY0435).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

WEXwater extracts of fruit of Xanthium strumarium
MEXmethanol extracts of fruits of X. strumarium
EEXAethanol extracts of aerial parts of X. strumarium
EEXSethanol extracts of stems of X. strumarium
WFEEXwater fraction of ethanol extracts of fruits of X. strumarium
NFEEXn-butanol fraction of ethanol extracts of fruits of X. strumarium
MEXAmethanol extracts of aerial parts of X. strumarium
HEXAhydroalcoholic extracts of aerial parts of X. strumarium
EAFEEXethylacetate fraction of ethanol extracts of fruits of X. strumarium
CFEEXAchloroform fraction of ethanol extracts of aerial parts of X. strumarium
CEXRchloroform extracts of roots of X. strumarium
MEXRmethanol extracts of roots of X. strumarium
EAFMEXethylacetate fraction of methanol extracts of fruits of X. strumarium
DFEEXAdichloromethane fraction of ethanol extracts of aerial parts of X. strumarium
EEXethanol extracts of fruits of X. strumarium
MEXLmethanol extracts of leaves of X. strumarium
WEXLwater extracts of leaveas of X. strumarium
EEXLethanol extracts of leaves of X. strumarium
EEXLethanol extracts of leaves of X. strumarium
PEEXWpetroleum ether extracts of whole plant of X. strumarium
MEXSmethanol extracts of stems of X. strumarium
EEXRethanol extracts of roots of X. strumarium
EOXessential oil of fruits of Xanthium strumarium
EOXLessential oil of leaves of Xanthium strumarium
WEXFTwater extract of flowering twigs of Xanthium strumarium
CFMEXLchloroform fraction of methanol extracts of leaves of X. strumarium
MEXWmethanol extracts of whole plant of X. strumarium
HEEXBhydro-ethanol extracts of burs of X. strumarium
HEEXAhydro-ethanol extracts of aerial parts of X. strumarium
EFEEXethylacetate fraction of ethanol extracts of X. strumarium

References

  1. Kamboj, A.; Saluja, A.K. Phytopharmacological review of Xanthium strumarium L. (Cocklebur). Int. J. Green Pharm. 2010, 4, 129–139. [Google Scholar] [CrossRef]
  2. Chinese Pharmacopoeia Commission. Pharmacopoeia of the People’s Republic of China Part I; People’s Medical Publishing House: Beijing, China, 1963; p. 130. (In Chinese) [Google Scholar]
  3. Amin, S.; Barkatullah; Khan, H. Pharmacology of Xanthium species. A review. J. Phytopharmacol. 2016, 5, 126–127. [Google Scholar]
  4. Zhuang, Y.S.; Hu, J.; Cai, H.; Qin, K.M.; Yang, B.; Liu, X.; Cai, B.C. advanced study on chemical constituents and pharmaceutical activities of Xanthium strumarium. J. Nanjing Univ. Tradit. Chin. Med. 2017, 33, 428–432. (In Chinese) [Google Scholar]
  5. Chinese Pharmacopoeia Commission. Pharmacopoeia of the People’s Republic of China Part I; People’s Medical Publishing House: Beijing, China, 2015; p. 162. (In Chinese) [Google Scholar]
  6. Nanjing University of Traditional Chinese Medicine. Traditional Chinese Medicine Dictionary; Shanghai Science and Technology Press: Shanghai, China, 1986; p. 1071. (In Chinese) [Google Scholar]
  7. Chopra, R.N.; Nayar, S.L.; Chopra, I.C. Glossary of Indian Medicinal Plants; Council of Scientific and Industrial Research: New Delhi, India, 1986; p. 259. [Google Scholar]
  8. Islam, M.R.; Uddin, M.Z.; Rahman, M.S.; Tutul, E.; Rahman, M.Z.; Hassan, M.A.; Faiz, M.A.; Hossain, M.; Hussain, M.; Rashid, M.A. Ethnobotanical, phytochemical and toxicological studies of Xanthium strumarium L. Bangladesh Med. Res. Counc. Bull. 2009, 35, 84–90. [Google Scholar] [CrossRef] [PubMed]
  9. Chang, F.; Hanna, M.A.; Zhang, D.J.; Li, H.; Zhou, Q.; Song, B.A.; Yang, S. Production of biodiesel from non-edible herbaceous vegetable oil: Xanthium sibiricum Patr. Bioresour. Technol. 2013, 140, 435–438. [Google Scholar] [CrossRef] [PubMed]
  10. Rozina; Asif, S.; Ahmad, M.; Zafar, M.; Ali, N. Prospects and potential of fatty acid methyl esters of some non-edible seed oils for use as biodiesel in Pakistan. Renew. Sustain. Energy Rev. 2017, 74, 687–702. [Google Scholar] [CrossRef]
  11. Chinese Flora Commission. Flora of China; Science Publishing House: Beijing, China, 1975; p. 325. (In Chinese) [Google Scholar]
  12. State Administration of Traditional Chinese Medicine. Chinese Materia Medica; Shanghai Science and Technology Press: Shanghai, China, 1998; pp. 1010–1013. (In Chinese) [Google Scholar]
  13. Shi, Y.S.; Liu, Y.B.; Ma, S.G.; Li, Y.; Qu, J.; Li, L.; Yuan, S.P.; Hou, Q.; Li, Y.H.; Jiang, J.D.; et al. Bioactive Sesquiterpenes and Lignans from the Fruits of Xanthium sibiricum. J. Nat. Prod. 2015, 78, 1526–1535. [Google Scholar] [CrossRef] [PubMed]
  14. Han, T.; Zhang, H.; Li, H.l.; Zhang, Q.H.; Zheng, H.C.; Qin, L.P. Composition of supercritical fluid extracts of some Xanthium species from China. Chem. Nat. Compd. 2008, 6, 814–816. [Google Scholar] [CrossRef]
  15. Winters, T.E.; Theodore, A.; Geissman, D.S. Sesquiterpene lactones of Xanthium species. Xanthanol and isoxanthanol, and correlation of xanthinin with ivalbin. J. Org. Chem. 1969, 34, 153–155. [Google Scholar] [CrossRef]
  16. McMillan, C.; Chavez, P.I.; Mabry, T.J. Sesquiterpene lactones of Xanthium strumarium in a texas population and in experimental hybrids. Biochem. Syst. Ecol. 1975, 3, 137–141. [Google Scholar] [CrossRef]
  17. Ahn, J.W.; No, Z.; Ryu, S.Y.; Zee, O.P. Isolation of cytotoxic compounds from the leaves of Xanthium strumarium L. Nat. Prod. Sci. 1995, 1, 1–4. [Google Scholar]
  18. Karmakar, U.K.; Ishikawa, N.; Toume, K.; Arai, M.A.; Sadhu, S.K.; Ahmed, F.; Ishibashi, M. Sesquiterpenes with TRAIL-resistance overcoming activity from Xanthium strumarium. Bioorg. Med. Chem. 2015, 23, 4746–4754. [Google Scholar] [CrossRef] [PubMed]
  19. Shi, Y.S.; Li, L.; Liu, Y.B.; Ma, S.G.; Li, Y.; Qu, J.; Liu, Q.; Shen, Z.F.; Chen, X.G.; Yu, S.S. A new thiophene and two new monoterpenoids from Xanthium sibiricum. J. Asian Nat. Prod. Res. 2015, 17, 1039–1047. [Google Scholar] [CrossRef] [PubMed]
  20. Malik, M.S.; Sangwan, N.K.; Dhindsa, K.S. Xanthanolides from Xanthium strumarium. Phytochemistry 1992, 32, 206–207. [Google Scholar] [CrossRef]
  21. Hu, D.Y.; Yang, S.Y.; Yuan, C.S.; Han, G.T.; Shen, H.M. Isolation and identification of chemical constituents in Xanthium sibiricum. Chin. Tradit. Herbal Drugs 2012, 43, 640–644. (In Chinese) [Google Scholar]
  22. Mahmoud, A.A. Xanthanolides and xanthane epoxide derivatives from Xanthium strumarium. Planta Med. 1998, 64, 724–727. [Google Scholar] [CrossRef] [PubMed]
  23. Saxena, V.K.; Mondal, S.K. A xanthanolide from Xanthium strumarium. Phytochemistry 1994, 35, 1080–1082. [Google Scholar] [CrossRef]
  24. Chen, J.; Wang, R.; Shi, Y.P. Chemical constituents from Xanthii Fructus. Chin. Tradit. Herbal Drugs 2013, 44, 1717–1720. (In Chinese) [Google Scholar]
  25. Wang, L.; Wang, J.; Li, F.; Liu, X.; Chen, B.; Tang, Y.X.; Wang, M.K. Cytotoxic sesquiterpene lactones from aerial parts of Xanthium sibiricum. Planta Med. 2013, 79, 661–665. [Google Scholar] [CrossRef]
  26. Jiang, H.; Yang, L.; Xing, X.D.; Yan, M.L.; Guo, X.Y.; Su, X.L.; Sun, Y.P.; Yang, B.Y.; Wang, Q.H.; Kuang, H.X. Chemical constituents of terpenoids from Xanthium strumarium. Chin. Tradit. Pat. Med. 2018, 40, 2461–2466. [Google Scholar]
  27. Cui, W.P. Chemical Constituents from Three Medicinal Plants and Their Bioactivities. Ph.D. Thesis, East China Normal University, Shanghai, China, 2013. (In Chinese). [Google Scholar]
  28. Ingawale, A.S.; Sadiq, M.B.; Nguyen, L.T.; Ngan, T.B. Optimization of extraction conditions and assessment of antioxidant, α-glucosidase inhibitory and antimicrobial activities of Xanthium strumarium L. fruits. Biocatal. Agric. Biotechnol. 2018, 14, 40–47. [Google Scholar] [CrossRef]
  29. Wahab, A.; Sultana, A.; Khan, K.M.; Irshad, A.; Ambreen, N.; Ali, M.; Bilal, M. Chemical investigation of Xanthium strumarium Linn and biological activity of its different fractions. J. Pharm. Res. 2012, 5, 1984–1987. [Google Scholar]
  30. Li, N.; Zhang, W.Z. Studies on Chemical Constituents of Xanthium sibiricum Patrin ex Widder. J. Qiqihar Univ. 2016, 32, 51–53. (In Chinese) [Google Scholar]
  31. Sultana, A. Phytochemical Studies on the Chemical Constituents of Xanthium strumarium Linn., Synthesis in addition Bioactivities of 2, 3-Diaminonaphthalenimidazole Derivatives and Amides of Piperic Acid. Ph.D. Thesis, Federal Urdu University of Arts, Science and Technology, Karachi, Pakistan, 2014. [Google Scholar]
  32. Kaur, M.; Kamboj, A.; Rathour, A.; Saluja, A.K. Isolation and Characterization of Constituents from the Leaves of Xanthium strumarium and their Evaluation for Antioxidant and Antimicrobial Potential. Nat. Prod. Chem. Res. 2015, 3, 168–173. [Google Scholar] [CrossRef]
  33. Agata, I.; Goto, S.; Hatano, T.; Nishibe, S.; Okuda, T. 1, 3, 5-tri-O-caffeoylquinic acid from Xanthium strumarium. Phytochemistry 1993, 33, 508–509. [Google Scholar] [CrossRef]
  34. Hwang, S.H.; Wang, Z.Q.; Yoon, H.N.; Lim, S.S. Xanthium strumarium as an Inhibitor of α-Glucosidase, Protein Tyrosine Phosphatase 1β, Protein Glycation and ABTS+ for Diabetic and Its Complication. Molecules 2016, 21, 1241. [Google Scholar] [CrossRef]
  35. Han, T.; Li, H.L.; Hu, Y.; Zhang, Q.Y.; Huang, B.K.; Zheng, H.C.; Rahman, K.; Qin, L.P. Phenolic acids in Fructus Xanthii and determination of contents of total phenolic acids in different species and populations of Xanthium in China. J. Chin. Intergr. Med. 2006, 4, 194–198. (In Chinese) [Google Scholar] [CrossRef]
  36. Tian, J.; Xia, Y.F.; Fang, K.H. Simultaneous determination of eight phenolic acids in Xanthium sibiricum by HPLC. Chin. Tradit. Pat. Med. 2013, 36, 1623–1626. (In Chinese) [Google Scholar]
  37. Yuan, H.E. Study on the Chemical Constituents of Herba Commelinae and Fructus Xanthii. Master’s Thesis, Jinan University, Guangzhou, China, 2014. (In Chinese). [Google Scholar]
  38. Cheng, Z.; Wang, L.; Chen, B.; Li, F.; Wang, M.K. Chemical Constituents from Fructus Xanthii. Chin. J. Appl. Environ. Biol. 2011, 17, 350–352. (In Chinese) [Google Scholar] [CrossRef]
  39. Kan, S.Q.; Chen, G.Y.; Han, C.R.; Chen, Z.; Song, X.M.; Ren, M.; Jiang, H. Chemical constituents from the roots of Xanthium sibiricum. Nat. Prod. Res. 2011, 25, 1243–1249. [Google Scholar] [CrossRef]
  40. Jiang, H.; Yang, L.; Ma, G.X.; Xing, X.D.; Yan, M.L.; Zhang, Y.Y.; Wang, Q.H.; Yang, B.Y.; Kuang, H.X.; Xu, X.D. New phenylpropanoid derivatives from the fruits of Xanthium sibiricum and their anti-inflammatory activity. Fitoterapia 2017, 117, 11–15. [Google Scholar] [CrossRef] [PubMed]
  41. Jiang, H.; Yang, L.; Liu, C.; Hou, H.; Wang, Q.; Wang, Z.; Yang, B.; Kuang, H. Four new glycosides from the fruit of Xanthium sibiricum Patr. Molecules 2013, 18, 12464–12473. [Google Scholar] [CrossRef] [PubMed]
  42. Yin, R.H.; Bai, X.; Feng, T.; Dong, Z.J.; Li, Z.H.; Liu, J.K. Two new compounds from Xanthium strumarium. J. Asian Nat. Prod. Res. 2016, 18, 354–359. [Google Scholar] [CrossRef] [PubMed]
  43. Han, T.; Li, H.L.; Zhang, Q.Y.; Zheng, H.C.; Qin, L.P. New thiazinediones and other components from Xanthium strumarium. Chem. Nat. Compd. 2006, 42, 567–570. [Google Scholar] [CrossRef]
  44. Pandey, D.P.; Rather, M.A. Isolation and Identification of Phytochemicals from Xanthium strumarium. Int. J. ChemTech Res. 2012, 4, 266–271. [Google Scholar]
  45. Jiang, H.; Yang, L.; Xing, X.D.; Zhang, Y.Y.; Yan, M.L.; Yang, B.Y.; Wang, Q.H.; Kuang, H.X. Chemical constituents from fruits of Xanthium sibiricum. Chin. Tradit. Herbal Drugs 2017, 48, 47–51. (In Chinese) [Google Scholar]
  46. Qiu, Y.L.; Dai, Y.H.; Wang, D.; Cui, Z. Chemical constituents in the fruits of Xanthium sibiricum. Chin. J. Med. Chem. 2010, 20, 214–216. (In Chinese) [Google Scholar]
  47. Yoon, H.N.; Lee, M.Y.; Kim, J.K.; Suh, H.W.; Lim, S.S. Aldose Reductase Inhibitory Compounds from Xanthium strumarium. Arch. Pharmacal. Res. 2013, 36, 1090–1095. [Google Scholar] [CrossRef]
  48. Jiang, H.; Yang, L.; Xing, X.D.; Yan, M.L.; Guo, X.Y.; Su, X.L.; Sun, Y.P.; Yang, B.Y.; Wang, Q.H.; Kuang, H.X. Study on lignans from Xanthii Fructus. Chin. J. Chin. Mater. Med 2018, 43, 2097–2103. (In Chinese) [Google Scholar]
  49. Wang, S.X.; Ren, L.J.; Sun, Z.R.; Pei, Y.H.; Zhu, T.R. Toxic Constituents in Seeds of Xanthium mongolicum. Chin. J. Med. Chem. 1983, 14, 529–531. (In Chinese) [Google Scholar]
  50. Craig, J.C.; Mole, M.L.; Billets, S.; El-Feraly, F. Isolation and identification of the hypoglycemic agent, carboxyatracrylate from Xanthium strumarium. Phytochemistry 1976, 15, 1178. [Google Scholar] [CrossRef]
  51. Jiang, H.; Zhang, Y.Y.; Zhang, Y.; Yang, L.; Wang, Q.H.; Kuang, H.X. Isolation and Identification of Chemical Constituents from the Fruit of Xanthium Sibiricum Patr. Inf. Tradit. Chin. Med. 2016, 33, 8–10. (In Chinese) [Google Scholar]
  52. Ma, Y.T.; Huang, M.C.; Hsu, F.L.; Chang, H.F. Thiazinedione from xanthium strumarium. Phytochemistry 1998, 48, 1083–1085. [Google Scholar] [CrossRef]
  53. Lee, C.L.; Huang, P.C.; Hsieh, P.W.; Hwang, T.L.; Hou, Y.Y.; Chang, F.R.; Wu, Y.C. (-)-Xanthienopyran, a new inhibitor of superoxide anion generation by activated neutrophils, and further constituents of the seeds of Xanthium strumarium. Planta Med. 2008, 74, 1276–1279. [Google Scholar] [CrossRef] [PubMed]
  54. Huang, W.H.; Yu, J.G.; Sun, L.; Guo, B.L.; Li, D.Y. Studies on Chemical Constituents of Xanthium sibiricum. Chin. J. Chin. Mater. Med. 2005, 30, 1027–1028. (In Chinese) [Google Scholar]
  55. Vasas, A.; Hohmann, J. Xanthane sesquiterpenoids: Structure, synthesis and biological activity. Nat. Prod. Rep. 2011, 28, 824–842. [Google Scholar] [CrossRef] [PubMed]
  56. Seaman, F.C. Sesquiterpene lactones as taxonomic characters in the asteraceae. Bot. Rev. 1982, 48, 121–594. [Google Scholar] [CrossRef]
  57. Hong, Y.; Han, Y.Q.; Xia, L.Z.; Gui, J.; Chen, X.; Sun, Y.H. Simultaneous Determination of Nine Phenolic Acid Components in Xanthii Fructus. Chin. Pharm. J. 2013, 13, 1109–1112. (In Chinese) [Google Scholar]
  58. Jiang, H.; Yang, L.; Xing, X.; Yan, M.; Guo, X.; Yang, B.; Wang, Q.; Kuang, H. HPLC-PDA Combined with Chemometrics for Quantitation of Active Components and Quality Assessment of Raw and Processed Fruits of Xanthium strumarium L. Molecules 2018, 23, 243. [Google Scholar] [CrossRef] [PubMed]
  59. Song, Z.Y.; Zhang, L.Y.; Xie, M.Z.; Li, Z.H.; Guo, W.W. Toxic Constituents and Pharmacological Effect of Xanthium sibiricum. Acta Pharm. Sin. 1962, 9, 678–683. (In Chinese) [Google Scholar]
  60. Duo, R.; Chen, Y.; Liu, Y.H.; Huang, Z.F.; Liu, Y.H.; Yi, J.H. Influence of processing on contents of carboxyatractyloside and atractyloside in Xanthii Fructus. Chin. Tradit. Pat. Med. 2013, 35, 353–356. (In Chinese) [Google Scholar]
  61. Hong, S.H.; Jeong, H.J.; Kim, H.M. Inhibitory effects of Xanthii Fructus extract on mast cell-mediated allergic reaction in murine model. J. Ethnopharmacol. 2003, 88, 229–234. [Google Scholar] [CrossRef]
  62. Hong, S.H.; Oh, M.J.; Lee, E.J.; Park, J.H.; Kim, N.H.; Rhee, H.K.; Kim, H.M.; Jung, S.K. Processed Xanthii Fructus increases cell viability of mast cell line, RBL-2H3. Orient. Pharm. Exp. Med. 2004, 4, 60–64. [Google Scholar]
  63. Zhao, Y.; Yang, H.; Zheng, Y.B.; Wong, Y.O.; Leung, P.C. The Effects of Fructus Xanthii Extract on Cytokine Release from Human Mast Cell Line (HMC-1) and Peripheral Blood Mononuclear Cells. Immunopharmacol. Immunotoxicol. 2008, 30, 543–552. [Google Scholar] [CrossRef] [PubMed]
  64. Yan, G.H.; Jin, G.Y.; Li, G.Z.; Cui, C.A.; Quan, G.H.; Jin, D.S.; Jin, D.Z. The possible mechanism of inhibitory effect of xanthium strumarium on mast cells activated by compound 48/80. Prog. Anat. Sci. 2010, 16, 164–166. (In Chinese) [Google Scholar]
  65. Peng, W.; Ming, Q.L.; Han, P.; Zhang, Q.Y.; Jiang, Y.P.; Zheng, C.J.; Han, Y.; Qin, L.P. Anti-allergic rhinitis effect of caffeoylxanthiazonoside isolated from fruits of Xanthium strumarium L. in rodent animals. Phytomedicine 2014, 21, 824–829. [Google Scholar] [CrossRef]
  66. Tao, L.; Fan, F.T.; Liu, Y.P.; Li, W.D.; Zhang, L.; Ruan, J.S.; Shen, C.S.; Sheng, X.B.; Zhu, Z.J.; Wang, A.Y.; et al. Concerted suppression of STAT3 and GSK3β is involved in growth inhibition of non-small cell lung cancer by Xanthatin. PLoS ONE 2013, 8, e81945. [Google Scholar] [CrossRef]
  67. Tao, L.; Sheng, X.; Zhang, L.; Li, W.; Wei, Z.; Zhu, P.; Zhang, F.; Wang, A.; Woodgett, J.R.; Lu, Y. Xanthatin anti-tumor cytotoxicity is mediated via glycogen synthase kinase-3β and β-catenin. Biochem. Pharmacol. 2016, 115, 18–27. [Google Scholar] [CrossRef]
  68. Tao, L.; Cao, Y.; Wei, Z.; Jia, Q.; Yu, S.; Zhong, J.; Wang, A.; Woodgett, J.R.; Lu, Y. Xanthatin triggers Chk1-mediated DNA damage response and destabilizes Cdc25C via lysosomal degradation in lung cancer cells. Toxicol. Appl. Pharmacol. 2017, 337, 85–94. [Google Scholar] [CrossRef]
  69. Kim, Y.S.; Kim, J.S.; Park, S.H.; Choi, S.U.; Lee, C.O.; Kim, S.K.; Kim, Y.K.; Kim, S.H.; Ryu, S.Y. Two cytotoxic sesquiterpene lactones from the leaves of Xanthium strumarium and their in vitro inhibitory activity on farnesyltransferase. Planta Med. 2003, 69, 375–377. [Google Scholar] [CrossRef]
  70. Ferrer, J.P.; Zampini, I.C.; Cuello, A.S.; Francisco, M.; Romero, A.; Valdivia, D.; Gonzalez, M.; Carlos, S.; Lamar, A.S.; Isla, M.I. Cytotoxic Compounds from Aerial Organs of Xanthium strumarium. Nat. Prod. Commun. 2016, 11, 371–374. [Google Scholar] [PubMed]
  71. Ramírez-Erosa, I.; Huang, Y.; Hickie, R.A.; Sutherland, R.G.; Barl, B. Xanthatin and xanthinosin from the burs of Xanthium strumarium L. as potential anticancer agents. Can. J. Physiol. Pharmacol. 2007, 85, 1160–1172. [Google Scholar] [CrossRef] [PubMed]
  72. Takeda, S.; Matsuo, K.; Yaji, K.; Okajima-Miyazaki, S.; Harada, M.; Miyoshi, H.; Okamoto, Y.; Amamoto, T.; Shindo, M.; Omiecinski, C.J.; et al. (-)-Xanthatin selectively induces GADD45γ and stimulates caspase-independent cell death in human breast cancer MDA-MB-231 cells. Chem. Res. Toxicol. 2011, 24, 855–865. [Google Scholar] [CrossRef] [PubMed]
  73. Takeda, S.; Noguchi, M.; Matsuo, K.; Yamaguchi, Y.; Kudo, T.; Nishimura, H.; Okamoto, Y.; Amamoto, T.; Shindo, M.; Omiecinski, C.J.; et al. (-)-Xanthatin up-regulation of the GADD45γ tumor suppressor gene in MDA-MB-231 breast cancer cells: Role of topoisomerase IIα inhibition and reactive oxygen species. Toxicology 2013, 305, 1–9. [Google Scholar] [CrossRef] [PubMed]
  74. Takeda, S.; Nishimura, H.; Koyachi, K.; Matsumoto, K.; Yoshida, K.; Okamoto, Y.; Amamoto, T.; Shindo, M.; Aramaki, H. (-)-Xanthatin induces the prolonged expression of c-Fos through an N-acetyl-l-cysteine (NAC)-sensitive mechanism in human breast cancer MDA-MB-231 cells. J. Toxicol. Sci. 2013, 38, 547–557. [Google Scholar] [CrossRef] [PubMed]
  75. Vaishnav, K.; George, L.B.; Highland, H.N. Induction of cell death through alteration of antioxidant activity in HeLa cervical cancer cells by Xanthium strumarium L. extract. IOSR J. Pharm. Biol. Sci. 2015, 10, 33–42. [Google Scholar]
  76. Liu, R.; Shi, D.; Zhang, J.; Li, X.; Han, X.; Yao, X.; Fang, J. Xanthatin Promotes Apoptosis via Inhibiting Thioredoxin Reductase and Eliciting Oxidative Stress. Mol. Pharm. 2018, 15, 3285–3296. [Google Scholar] [CrossRef]
  77. Bui, V.B.; Liu, S.T.; Zhu, J.J.; Xiong, J.; Zhao, Y.; Yang, G.X.; Xia, G.; Hu, J.F. Sesquiterpene lactones from the aerial parts of Xanthium sibiricum and their cytotoxic effects on human cancer cell lines. Phytochem. Lett. 2012, 5, 685–689. [Google Scholar] [CrossRef]
  78. Zhang, L.; Tao, L.; Ruan, J.; Li, W.; Wu, Y.; Yan, L.; Zhang, F.; Fan, F.; Zheng, S.; Wang, A.; et al. Xanthatin induces G2/M cell cycle arrest and apoptosis in human gastric carcinoma MKN-45 cells. Planta Med. 2012, 78, 890–895. [Google Scholar] [CrossRef]
  79. Jiang, H.; Ma, G.X.; Yang, L.; Xing, X.D.; Yan, M.L.; Zhang, Y.Y.; Wang, Q.H.; Kuang, H.X.; Xu, X.D. Rearranged ent-kauranoid glycosides from the fruits of Xanthium strumarium and their antiproliferative activity. Phytochem. Lett. 2016, 18, 192–196. [Google Scholar] [CrossRef]
  80. Al-Mekhlafi, F.A.; Abutaha, N.; Mashaly, A.M.A.; Nasr, F.A.; Ibrahim, K.E.; Wadaan, M.A. Biological activity of Xanthium strumarium seed extracts on different cancer cell lines and Aedes caspius, Culex pipiens (Diptera: Culicidae). Saudi, J. Biol. Sci. 2017, 24, 817–821. [Google Scholar] [CrossRef] [PubMed]
  81. Li, H.; Min, Y.S.; Park, K.C.; Kim, D.S. Inhibition of melanogenesis by Xanthium strumarium L. Biosci. Biotechnol. Biochem. 2012, 76, 767–771. [Google Scholar] [CrossRef] [PubMed]
  82. Li, W.D.; Wu, Y.; Zhang, L.; Yan, L.G.; Yin, F.Z.; Ruan, J.S.; Chen, Z.P.; Yang, G.M.; Yan, C.P.; Zhao, D.; et al. Characterization of xanthatin: Anticancer properties and mechanisms of inhibited murine melanoma in vitro and in vivo. Phytomedicine 2013, 20, 865–873. [Google Scholar] [CrossRef] [PubMed]
  83. Roussakis, C.; Chinou, I.; Vayas, C.; Harvala, C.; Verbist, J.F. Cytotoxic activity of xanthatin and the crude extracts of Xanthium strumarium. Planta Med. 1994, 60, 473–474. [Google Scholar] [CrossRef] [PubMed]
  84. Nibret, E.; Youns, M.; Krauth-Siegel, R.L.; Wink, M. Biological activities of xanthatin from Xanthium strumarium leaves. Phytother. Res. 2011, 25, 1883–1890. [Google Scholar] [CrossRef] [PubMed]
  85. Pan, J.H.; Wang, Y.L.; Xie, M.R.; Yu, F.R. Inhibitory effect of xanthium extract on S180 cells growth and the impact on immune functions in tumor-bearing mice. Chin. J. Clin. Res. 2013, 26, 317–319. (In Chinese) [Google Scholar]
  86. Ishwarya, S.; Singh, M.K. Evaluation of antioxidant and invitro cytotoxicity of crude fractions from the roots of Xanthiuim strumarium. Int. J. Pharmtech Res. 2010, 2, 2219–2223. [Google Scholar]
  87. An, H.J.; Jeong, H.J.; Lee, E.H.; Kim, Y.K.; Hwang, W.J.; Yoo, S.J. Xanthii Fructus Inhibits Inflammatory Responses in LPS-Stimulated Mouse Peritoneal Macrophages. Inflammation 2004, 28, 263–270. [Google Scholar] [CrossRef]
  88. Kim, I.T.; Park, Y.M.; Won, J.H.; Jung, H.J.; Park, H.J.; Choi, J.W.; Lee, K.T. Methanol extract of Xanthium strumarium L. possesses anti-inflammatory and anti-nociceptive activities. Biol. Pharm. Bull. 2005, 28, 94–100. [Google Scholar] [CrossRef]
  89. Yoon, J.H.; Lim, H.J.; Lee, H.J.; Kim, H.D.; Jeon, R.; Ryu, J.H. Inhibition of lipopolysaccharide-induced inducible nitric oxide synthase and cyclooxygenase-2 expression by xanthanolides isolated from Xanthium strumarium. Bioorg. Med. Chem. Lett. 2008, 18, 2179–2182. [Google Scholar] [CrossRef]
  90. Khuda, F.; Iqbal, Z.; Khan, A.; Shah, Z.Y.; Ahmad, L.; Nasir, F.; Hassan, M.; Shah, I.; Shah, W.A. Evaluation of anti-inflammatory activity of selected medicinal plants of Khyber Pakhtunkhwa, Pakistan. Pak. J. Pharm. Sci. 2014, 27, 365–368. [Google Scholar] [PubMed]
  91. Ju, A.; Cho, Y.C.; Cho, S. Methanol extracts of Xanthium sibiricum roots inhibit inflammatory responses via the inhibition of nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) in murine macrophages. J. Ethnopharmacol. 2015, 174, 74–81. [Google Scholar] [CrossRef] [PubMed]
  92. Yeom, M.; Kim, J.H.; Min, J.H.; Hwang, M.K.; Jung, H.S.; Sohn, Y. Xanthii Fructus inhibits inflammatory responses in LPS-stimulated RAW 264.7 macrophages through suppressing NF-κB and JNK/p38 MAPK. J. Ethnopharmacol. 2015, 176, 394–401. [Google Scholar] [CrossRef] [PubMed]
  93. Hossen, M.J.; Kim, M.Y.; Cho, J.Y. MAPK/AP-1-Targeted Anti-Inflammatory Activities of Xanthium strumarium. Am. J. Chin. Med. 2016, 44, 1111–1125. [Google Scholar] [CrossRef] [PubMed]
  94. Hossen, M.J.; Cho, J.Y.; Kim, D. PDK1 in NF-κB signaling is a target of Xanthium strumarium methanolic extract-mediated anti-inflammatory activities. J. Ethnopharmacol. 2016, 190, 251–260. [Google Scholar] [CrossRef] [PubMed]
  95. Huang, M.H.; Wang, B.S.; Chiu, C.S.; Amagaya, S.; Hsieh, W.T.; Huang, S.S.; Shie, P.H.; Huang, G.J. Antioxidant, antinociceptive, and anti-inflammatory activities of Xanthii Fructus extract. J. Ethnopharmacol. 2011, 135, 545–552. [Google Scholar] [CrossRef] [PubMed]
  96. Han, T.; Li, H.L.; Zhang, Q.Y.; Han, P.; Zheng, H.C.; Rahman, K.; Qin, L.P. Bioactivity-guided fractionation for anti-inflammatory and analgesic properties and constituents of Xanthium strumarium L. Phytomedicine 2007, 14, 825–829. [Google Scholar] [CrossRef]
  97. Park, J.H.; Kim, M.S.; Jeong, G.S.; Yoon, J. Xanthii Fructus extract inhibits TNF-α/IFN-γ-induced Th2-chemokines production via blockade of NF-κB, STAT1 and p38-MAPK activation in human epidermal keratinocytes. J. Ethnopharmacol. 2015, 171, 85–93. [Google Scholar] [CrossRef]
  98. Hasan, T.; Das, B.K.; Qibria, T.; Morshed, M.A.; Uddin, M.A. Phytochemical Screening and Evaluation of Analgesic Activity of Xanthium Srtumarium L. Asian. J. Biochem. Pharm. Res. 2011, 1, 2231–2560. [Google Scholar]
  99. Talakal, T.S.; Dwivedi, S.K.; Sharma, S.R. In vitro and in vivo antitrypanosomal activity of Xanthium strumarium leaves. J. Ethnopharmacol. 1995, 49, 141–145. [Google Scholar] [CrossRef]
  100. Gökçe, A.; Isaacs, R.; Whalon, M.E. Ovicidal, larvicidal and anti-ovipositional activities of Bifora radians and other plant extracts on the grape berry moth Paralobesia viteana (Clemens). J. Pest. Sci. 2011, 84, 487–493. [Google Scholar] [CrossRef]
  101. Chandel, S.; Bagai, U. Screening of Cytotoxicity and Antiplasmodial Activity of Xanthium strumarium L. Res. J. Pharm. Biol. Chem. Sci. 2012, 3, 625–631. [Google Scholar]
  102. Roy, B.; Amin, M.R.; Jalal, S.; Kwon, Y.J.; Suh, S.J. Evaluation of common cocklebur Xanthium strumarium leaf extract as post-harvest grain protectant of black gram against pulse beetle Callosobruchus chinensis (Coleoptera: Bruchidae) and isolation of crude compound. Entomol. Res. 2014, 44, 254–261. [Google Scholar] [CrossRef]
  103. Kepenekci, I.; Saglam, H.D. Extracts of some indigenous plants affecting hatching and mortality in the root-knot nematode [Meloidogyne javanica (Treub) Chitwood]. Egypt. J. Pest Control 2015, 25, 39–44. [Google Scholar]
  104. Kumar, K.K.S.; Rajkapoor, B. Effect of Xanthium strumarium L. Extracts on Antioxidant Enzymes Levels in Rat Brain after Induction of Epilepsy. Pharmacologyonline 2010, 2, 883–888. [Google Scholar]
  105. Narendiran, S.; Mohanambal, E.; Kumar, P.S.; Shankar, M.; Kuttimani, T.; Vijayakumar, B. Study of anti-diabetic and anti-oxidant activities of methanolic extract of Xanthium strumarium (Linn.) stems on diabetic rats. J. Pharm. Res. 2011, 4, 3728–3732. [Google Scholar]
  106. Sridharamurthy, N.B.; Yogananda, R.; Srinivas, U. In-vitro Antioxidant and Antilipidemic Activities of Xanthium strumarium L. Curr. Trends Biotechnol. Pharm. 2011, 5, 1362–1371. [Google Scholar]
  107. Kamboj, A.; Atri, P.; Saluja, A.K. Phytochemical Screening, In-vitro Evaluation of Antioxidant and Free Radical Scavenging Activity of Leaves, Stems and Roots of Xanthium strumarium L., (Compositae). Br. J. Pharm. Res. 2014, 4, 1–22. [Google Scholar] [CrossRef]
  108. Ghahari1, S.; Alinezhad, H.; Nematzadeh, G.A.; Tajbakhsh, M.; Baharfar, R. Biochemical Composition, Antioxidant and Biological Activities of the Essential Oil and Fruit Extract of Xanthium strumarium Linn. From Northern Iran. J. Agric. Sci. Technol. 2017, 19, 1603–1616. [Google Scholar]
  109. Mehta, P.; Chopra, S.; Mehta, A. Antimicrobial properties of some plant extracts against bacteria. Folia Microbiol. 1983, 28, 467–469. [Google Scholar] [CrossRef]
  110. Sato, Y.; Oketani, H.; Yamada, T.; Singyouchi, K.; Ohtsubo, T.; Kihara, M.; Shibata, H.; Higuti, T. A xanthanolide with potent antibacterial activity against methicillin-resistant Staphylococcus aureus. J. Pharm. Pharmacol. 1997, 49, 1042–1044. [Google Scholar] [CrossRef] [PubMed]
  111. Srinivas, P.; Rajashekar, V. Phytochemical Screening and in vitro Antimicrobial Investigation of the Methanolic Extract of Xanthium strumarium Leaf. Int. J. Drug Dev. Res. 2011, 3, 286–293. [Google Scholar]
  112. Chen, W.H.; Liu, W.J.; Wang, Y.; Song, X.P.; Chen, G.Y. A new naphthoquinone and other antibacterial constituents from the roots of Xanthium sibiricum. Nat. Prod. Res. 2015, 29, 739–744. [Google Scholar] [CrossRef] [PubMed]
  113. Devkota, A.; Das, R.K. Antibacterial activities of Xanthium strumarium L. J. Nat. Hist. Mus. 2015, 29, 70–77. [Google Scholar] [CrossRef]
  114. Sharifi-Rad, J.; Hoseini-Alfatemi, S.M.; Sharifi-Rad, M.; Sharifi-Rad, M.; Iriti, M.; Sharifi-Rad, M.; Sharifi-Rad, R.; Raeisi, S. Phytochemical compositions and biological activities of essential oil from Xanthium strumarium L. Molecules 2015, 20, 7034–7047. [Google Scholar] [CrossRef] [PubMed]
  115. Sharifi-Rad, J.; Soufi, L.; Ayatollahi, S.A.; Iriti, M.; Sharifi-Rad, M.; Varoni, E.M.; Shahri, F.; Esposito, S.; Kuhestani, K.; Sharifi-Rad, M. Anti-bacterial effect of essential oil from Xanthium strumarium against shiga toxin-producing Escherichia coli. Cell. Mol. Biol. 2016, 62, 69–74. [Google Scholar] [PubMed]
  116. Wang, W.; Jiang, H.; Zhiwei, W.U.; Qian, J.; Wang, X.; Jinxiu, X.U. Study on the bacteriostatic effects of 7 kinds of chinese herbal medicines such as ophiopogon japonicus and comb. Agric. Sci. Technol. 2016, 17, 2560. [Google Scholar]
  117. Kim, D.K.; Shim, C.K.; Bae, D.W.; Kawk, Y.S.; Yang, M.S.; Kim, H.K. Identification and Biological Characteristics of an Antifungal Compound Extracted from Cocklebur (Xanthium strumarium) against Phytophthora drechsleri. Plant Pathol. J. 2002, 18, 288–292. [Google Scholar] [CrossRef]
  118. Yanar, Y.; Kadioğlu, L.; Gökçe, A.; Demirtaş, D.; Gören, N.; Çam, H.; Whalon, M. In vitro antifungal activities of 26 plant extracts on mycelial growth of Phytophthora infestans (Mont.) de Bary. Afr. J. Biotechnol. 2011, 10, 2625–2629. [Google Scholar]
  119. Parveen, Z.; Mazhar, S.; Siddique, S.; Manzoor, A.; Ali, Z. Chemical Composition and Antifungal Activity of Essential Oil from Xanthium strumarium L. Leaves. Indian J. Pharm. Sci. 2017, 79, 316–321. [Google Scholar] [CrossRef]
  120. Kupiecki, F.P.; Ogzewalla, C.D.; Schell, F.M. Isolation and characterization of a hypoglycemic agent from Xanthium strumarium. J. Pharm. Sci. 1974, 63, 1166–1167. [Google Scholar] [CrossRef] [PubMed]
  121. Hsu, F.L.; Chen, Y.C.; Cheng, J.T. Caffeic acid as active principle from the fruit of Xanthium strumarium to lower plasma glucose in diabetic rats. Planta Med. 2000, 66, 228–230. [Google Scholar] [CrossRef]
  122. Khuda, F.; Iqbal, Z.; Khan, A.; Zakiullah; Shah, Y.; Khan, A. Report: Screening of selected medicinal plants for their enzyme inhibitory potential—A validation of their ethnopharmacological uses. Pak. J. Pharm. Sci. 2014, 27, 593–596. [Google Scholar] [PubMed]
  123. Li, X.M.; Yang, M.X.; Li, Z.P.; Xue, M.; ShangGuan, Z.S.; Ou, Z.M.; Liu, M.; Liu, S.H.; Yang, S.Y.; Li, X.J. Fructus xanthii improves lipid homeostasis in the epididymal adipose tissue of rats fed a high-fat diet. Mol. Med. Rep. 2016, 13, 787–795. [Google Scholar] [CrossRef] [PubMed]
  124. Li, T.X.; Shen, J.Y.; Li, M.; Wang, G.R. Effects of Fructus Xanthii on Blood Glucose and Lipid in Diabetic Mice before and after Processing. Lishizhen Med. Mater. Med. Res. 2017, 28, 608–609. (In Chinese) [Google Scholar]
  125. Liu, Y.; Wu, Z.M.; Lan, P. Experimental Study on Effect of Fructus Xanthii Extract on Duck Hepatitis B Virus. Lishizhen Med. Mater. Med. Res. 2009, 20, 1776–1777. (In Chinese) [Google Scholar]
  126. Wang, Y.H.; Li, T.H.; Wu, B.Q.; Liu, H.; Shi, Y.F.; Feng, D.Y. Protective effects of caffeoylxanthiazonoside isolated from fruits of Xanthium strumarium on sepsis mice. Pharm. Biol. 2015, 53, 1367–1371. [Google Scholar] [CrossRef] [PubMed]
  127. Li, X.M.; Li, Z.P.; Xue, M.; Ou, Z.M.; Liu, M.; Yang, M.X.; Liu, S.H.; Yang, S.Y.; Li, X.J. Fructus Xanthii Attenuates Hepatic Steatosis in Rats Fed on High-Fat Diet. PLoS ONE 2013, 8, e61499. [Google Scholar] [CrossRef]
  128. Lin, B.; Zhao, Y.; Han, P.; Yue, W.; Ma, X.Q.; Rahman, K.; Zheng, C.J.; Qin, L.P.; Han, T. Anti-arthritic activity of xanthium strumarium L. extract on complete freund’s adjuvant induced arthritis in rats. J. Ethnopharmacol. 2014, 155, 248–255. [Google Scholar] [CrossRef]
  129. Afsar, S.K.; Kumar, K.R.; Raveesha, P.; Sree, K. Evaluation of Anti-pyretic Activity of Methanolic Whole Plant Extract of Xanthium strumarium Against Yeast Induced Pyrexia Model in Wistar Rats. J. Pharm. Res. 2012, 5, 5277–5279. [Google Scholar]
  130. Kumar, K.K.S.; Rajkapoor, B. Evaluation of Anti-epileptic Activity of Xanthium strumarium L. Pharmacologyonline 2010, 2, 850–855. [Google Scholar]
  131. Panigrahi, P.N.; Dey, S.; Sahoo, M.; Choudhary, S.S.; Mahajan, S. Alteration in oxidative/nitrosative imbalance, histochemical expression of osteopontin and antiurolithiatic efficacy of xanthium strumarium (L.) in ethylene glycol induced urolithiasis. Biomed. Pharmacother. 2016, 84, 1524–1532. [Google Scholar] [CrossRef] [PubMed]
  132. Kandhare, A.D.; Kumar, V.S.; Adil, M.; Rajmane, A.R.; Ghosh, P.; Bodhankar, S.L. Investigation of gastro protective activity of Xanthium strumarium L. by modulation of cellular and biochemical marker. Orient. Pharm. Exp. Med. 2012, 12, 287–299. [Google Scholar] [CrossRef]
  133. Yang, B.; Wang, F.; Cao, H.; Liu, G.; Zhang, Y.; Yan, P.; Li, B. Caffeoylxanthiazonoside exerts cardioprotective effects during chronic heart failure via inhibition of inflammatory responses in cardiac cells. Exp. Ther. Med. 2017, 14, 4224–4230. [Google Scholar] [CrossRef] [PubMed]
  134. Yan, C.P.; Li, H.; Wu, Y.; Xie, D.H.; Weng, Z.B.; Cai, B.C.; Liu, X.; Li, W.D.; Chen, Z.P. Determination of xanthatin by ultra high performance liquid chromatography coupled with triple quadrupole mass spectrometry: Application to pharmacokinetic study of xanthatin in rat plasma. J. Chromatogr. B 2014, 974–978, 57–61. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, L.L. The Detection of atractyIoside in Fructus Xanthii and the Pharmacokinetic of Atractyloside in Rat. Master’s Thesis, Guangzhou University of Chinese Medicine, Guangzhou, China, 2013. (In Chinese). [Google Scholar]
  136. Shen, J.Y. Comparative Study of Pharmacodynamics and Pharmacokinetics before and after Processing Xanthii Fructus. Master’s Thesis, Hubei University of Traditional Chinese Medicine, Wuhan, China, 2016. (In Chinese). [Google Scholar]
  137. Witte, S.T.; Osweiler, G.D.; Stahr, H.M.; Mobley, G. Cocklebur toxicosis in cattle associated with the consumption of mature Xanthium strumarium. J. Vet. Diagn. Investig. 1990, 2, 263–267. [Google Scholar] [CrossRef] [PubMed]
  138. Hu, Y.; Wang, J.X.; Zhang, L.; Huang, J.K.; Yang, X.H. Comprehensive evaluation and risk control measures of Xanthii Fructus. Chin. J. Chin. Mater. Med. 2017, 42, 4079–4085. (In Chinese) [Google Scholar]
  139. Li, J.; Gao, T.; Xie, Z.Q.; Tang, Y. The Toxicity Comparison Experiment of the Extractions from Fructus Xanthii. Lishizhen Med. Mater. Med. Res. 2005, 16, 484–487. (In Chinese) [Google Scholar]
  140. Yan, L.C.; Zhang, T.T.; Zhao, J.N.; Song, J.; Hua, H.; Li, L. Comparative study on acute toxicity of four extracts from Xanthii Fructus in mice. Chin. J. Chin. Mater. Med. 2012, 37, 2228–2231. (In Chinese) [Google Scholar]
  141. Fu, S.; Guan, J.H. Experimental Study on the Toxicity and Efficacy of Siegesbeckiae and Cocklebur. World J. Integr. Tradit. West. Med. 2015, 10, 493–496. (In Chinese) [Google Scholar]
  142. Stuart, B.P.; Cole, R.J.; Gosser, H.S. Cocklebur (xanthium strumarium L. var. strumarium) intoxication in swine: Review and redefinition of the toxic principle. Vet. Pathol. 1981, 18, 368–383. [Google Scholar]
  143. Wang, Y.; Han, T.; Xue, L.M.; Han, P.; Zhang, Q.Y.; Huang, B.K.; Zhang, H.; Ming, Q.L.; Peng, W.; Qin, L.P. Hepatotoxicity of kaurene glycosides from Xanthium strumarium L. fruits in mice. Die Pharm. 2011, 66, 445–449. [Google Scholar]
  144. Stewart, M.J.; Steenkamp, V. The Biochemistry and Toxicity of Atractyloside: A Review. Ther. Drug Monit. 2000, 22, 641–649. [Google Scholar] [CrossRef] [PubMed]
  145. Jin, Y.; Liu, S.M.; Liu, Y.; Mou, H. Toxic effects of ethylacetate, n-butanol, and water extracts from alcohol extractions of cocklebur fruit on liver in rats. Advers. Drug React. J. 2010, 12, 17–20. (In Chinese) [Google Scholar]
  146. Cao, M.; Wu, B.; Ma, D.; Bai, Y.; Liu, S.M. Metabolomics study on Fructus Xanthii-induced hepatotoxicity in rats. Advers. Drug React. J. 2011, 13, 287–293. (In Chinese) [Google Scholar]
  147. Xue, L.M.; Zhang, Q.Y.; Han, P.; Jiang, Y.P.; Yan, R.D.; Wang, Y.; Rahman, K.; Jia, M.; Han, T.; Qin, L.P. Hepatotoxic constituents and toxicological mechanism of Xanthium strumarium L. fruits. J. Ethnopharmacol. 2014, 152, 272–282. [Google Scholar] [CrossRef] [PubMed]
  148. Zeng, J.; Tang, S.W.; Liu, Y.H.; Wang, Y.Q.; Hua, Y.; Zhao, J.N. Experimental Study on Liver Toxicity of Raw and Stir-fried Xanthium sibiricum Based on Sensitive Biomarkers. Pharm. Clin. Chin. Mater. Med. 2018, 34, 122–125. (In Chinese) [Google Scholar]
  149. Mandal, S.C.; Dhara, A.K.; Kumar, C.K.A.; Maiti, B.C. Neuropharmacological Activity of Xanthium Strumarium Linn. Extract. J. Herbs Spices Med. Plants 2001, 8, 69–77. [Google Scholar] [CrossRef]
  150. Yin, J.; Li, D.; Hu, W.; Meng, Q. Effects of glycyrrhizic acid on cocklebur-induced hepatotoxicity in rat and human hepatocytes. Phytother. Res. 2008, 22, 395–400. [Google Scholar] [CrossRef]
  151. Yu, J.; Song, M.Z.; Wang, J.; Li, Y.F.; Lin, P.; Que, L.; Bao, Z. In vitro cytotoxicity and in vivo acute and chronic toxicity of Xanthii Fructus and its processed product. Biomed. Res. Int. 2013, 2013, 1–12. [Google Scholar]
  152. Piloto Ferrer, J.; Cozzi, R.; Cornetta, T.; Stano, P.; Fiore, M.; Degrassi, F.; De Salvia, R.; Remigio, A.; Francisco, M.; Quiñones, O.; et al. Xanthium strumarium L. Extracts Produce DNA Damage Mediated by Cytotoxicity in In vitro Assays but Does Not Induce Micronucleus in Mice. Biomed. Res. Int. 2014, 2014, 575197. [Google Scholar] [CrossRef] [PubMed]
  153. Su, T.; Cheng, B.C.; Fu, X.Q.; Li, T.; Guo, H.; Cao, H.H.; Kwan, H.Y.; Tse, A.K.W.; Yu, H.; Cao, H.; et al. Comparison of the toxicities, bioactivities and chemical profiles of raw and processed Xanthii Fructus. BMC Complement. Altern. Med. 2016, 16, 24. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, X.Q.; Hou, H.R.; Liu, K.C.; Wang, X.M.; Peng, W.B.; Han, L.W.; Wang, X.; Chen, W.Y. Toxicity of Fructus Xanthii extract to the growth and motion behavior of zebrafish embryos. Shandong SCI 2014, 27, 10–13. (In Chinese) [Google Scholar]
Figure 1. Xanthium strumarium L. A–D represent the whole plants (A), leaves (B), inflorescence (C) and fruits (D) of X. strumarium L.
Figure 1. Xanthium strumarium L. A–D represent the whole plants (A), leaves (B), inflorescence (C) and fruits (D) of X. strumarium L.
Molecules 24 00359 g001
Figure 2. Chemical structures of the sesquiterpenoids in X. strumarium.
Figure 2. Chemical structures of the sesquiterpenoids in X. strumarium.
Molecules 24 00359 g002aMolecules 24 00359 g002b
Figure 3. Chemical structures of the triterpenoids in X. strumarium.
Figure 3. Chemical structures of the triterpenoids in X. strumarium.
Molecules 24 00359 g003
Figure 4. Chemical structures of the phenylpropenoids in X. strumarium.
Figure 4. Chemical structures of the phenylpropenoids in X. strumarium.
Molecules 24 00359 g004aMolecules 24 00359 g004b
Figure 5. Chemical structures of the lignanoids in X. strumarium.
Figure 5. Chemical structures of the lignanoids in X. strumarium.
Molecules 24 00359 g005
Figure 6. Chemical structures of the coumarins in X. strumarium.
Figure 6. Chemical structures of the coumarins in X. strumarium.
Molecules 24 00359 g006
Figure 7. Chemical structures of the steroids in X. strumarium.
Figure 7. Chemical structures of the steroids in X. strumarium.
Molecules 24 00359 g007
Figure 8. Chemical structures of the glycosides in X. strumarium.
Figure 8. Chemical structures of the glycosides in X. strumarium.
Molecules 24 00359 g008
Figure 9. Chemical structures of the flavonoids in X. strumarium.
Figure 9. Chemical structures of the flavonoids in X. strumarium.
Molecules 24 00359 g009
Figure 10. Chemical structures of the Thiazides in X. strumarium.
Figure 10. Chemical structures of the Thiazides in X. strumarium.
Molecules 24 00359 g010
Figure 11. Chemical structures of the anthraquinones and naphthoquinones in X. strumarium.
Figure 11. Chemical structures of the anthraquinones and naphthoquinones in X. strumarium.
Molecules 24 00359 g011
Figure 12. Chemical structures of other compounds in X. strumarium.
Figure 12. Chemical structures of other compounds in X. strumarium.
Molecules 24 00359 g012
Table 1. The traditional and clinical uses of Xanthium strumarium in China.
Table 1. The traditional and clinical uses of Xanthium strumarium in China.
Preparation NameMain CompositionsTraditional and Clinical UsesReferences
Li Bi TabletsXanthii Fructus, Scutellariae Radix, Magno1iae Flos, Menthae Haplocalycis Herba, Angelicae Dahuricae Radix, Asari Radix Et Rhizoma, Taraxaci HerbaCuring common cold with nasal obstruction, nasosinusitis, turbid nasal discharge“Chinese Pharmacopoeia (2010)” a
Shuang Xin Bi Dou Yan Ke LiXanthii Fructus, Magno1iae Flos, Angelicae Dahuricae Radix, Asari Radix Et Rhizoma, Lonicerae Japonicae Flos, Lonicerae Japonicae Cau1is, Taraxaci Herba, Glycyrrhizae Radix Et Rhizoma, Platycodonis Radix, Chrysanthemi Flos, Scutellariae Radix, Paeoniae Radix Rubra, Coicis Semen, Rehmanniae Radix Treating nasosinusitis“Guo Jia Zhong Cheng Yao Biao Zhun” b
Xiao Er Bi Yan TabletsXanthii Fructus, Ligustici Rhizoma Et Radix, Saposhnikoviae Radix, Angelicae Dahuricae Radix, Polygoni Tinctorii Folium, Taraxaci Herba, Cimicifugae Rhizoma, Glycyrrhizae Radix Et RhizomaCuring chronic rhinitis of child“Zhong Yao Cheng Fang Zhi Ji”c
Yu Yuan WanXanthii Fructus, Scutellariae Radix, Gardeniae Fructus, Scrophulariae Radix, Magno1iae Flos, Ophiopogonis Radix, Lycii Cortex, Paeoniae Radix Rubra, Forsythiae Fructus, Angelicae Dahuricae Radix, Menthae Haplocalycis Herb, Schizonepetae Herba, Glycyrrhizae Radix Et Rhizoma, Platycodonis RadixTreating redness and swelling of the nostrils, swelling and pain in throat“Zhong Yao Cheng Fang Zhi Ji”c
Yi Xuan Ning Jiao NangXanthii Fructus, Chrysanthemi Flos, Arisaema Cum Bile, Scutellariae Radix, Bambusae Caulis in Taenias, Ostreae Concha, Crataegi Fructus, Citri Reticulatae Pericarpium, Paeoniae Radix Alba Poria, Lycii FructusTreating hyperactivity of liver-yang, vertigo due to deficiency of Qi and blood“Xin Yao Zhuan Zheng Biao Zhun” d
Qing Re Zhi Ke Ke LiXanthii Fructus, Scutellariae Radix, Fritillariae Thunbergii Bulbus, Paridis Rhizoma, Commelinae Herba, Anemarrhenae Rhizoma, Gypsum Fibrosum, Citri Reticulatae Pericarpium, Aurantii Fructus, Armeniacae Semen Amarum, Platycodonis RadixCuring cough, phlegm, fever, pharyngalgia, thirst, chest tightness, dry stool, yellow urine due to pulmonary retention of phlegmopyrexia; acute bronchitis, acute exacerbation of chronic bronchitis“Xin Yao Zhuan Zheng Biao Zhun” d
Di Tong Bi Yan LiquidXanthii Fructus, Taraxaci Herba, Asari Radix Et Rhizoma, Scutellariae Radix, Ephedrae Herba, Acori Tatarinowii Rhizoma, Angelicae Dahuricae Radix, Magno1iae FlosCuring common cold with nasal obstruction, chronic rhinitis, allergic rhinitis, nasosinusitis “Zhong Yao Cheng Fang Zhi Ji” c
Di Tong Bi Yan Liquid Pen Wu JiXanthii Fructus, Scutellariae Radix, Taraxaci Herba, Ephedrae Herba, Magno1iae Flos, Angelicae Dahuricae Radix, Asari Radix Et Rhizoma, Acori Tatarinowii RhizomaCuring common cold with nasal obstruction, chronic rhinitis, allergic rhinitis, nasosinusitis“Xin Yao Zhuan Zheng Biao Zhun” d
Fu Yang Chong JiXanthii Fructus, Chuanxiong Rhizoma, Carthami Flos, Kochiae FructusTreating pruritus, eczema, urticaria“Zhong Yao Cheng Fang Zhi Ji” c
Dan Xiang Bi Yan TabletsXanthii Fructus, Pogostemonis Herba, Angelicae Dahuricae Radix, Centipedae Herba, Schizonepetae Herba, Lonicerae Japonicae Flos, Chrysanthemi Indici FlosCuring chronic simple rhinitis, allergic rhinitis, acute and chronic rhinitis, and nasosinusitis“Zhong Yao Cheng Fang Zhi Ji” c
Nao Ning TabletsXanthii Fructus, Polygonati Rhizoma, Epimedii Folium, Ophiopogonis Radix, Ginseng Radix Et Rhizoma Rubra, Polygalae Radix, Ziziphi Spinosae Semen, Schisandrae Chinensis Fructus, Lycii Fructus, Cervi Cornu Pantotrichum, Testudinis Carapax Et Plastrum, Poria, Jujubae Fructus, Rehmanniae Radix Praeparata, Cervi Cornus CollaCuring neurasthenia, forgetfulness and insomnia, dizziness and palpitation, weariness of body, weak health and spontaneous perspiration, impotence and spermatorrhea“Zhong Yao Cheng Fang Zhi Ji” c
Nao Ning Su TabletsXanthii Fructus, Polygonati Rhizoma, Lycii Fructus, Poria, Epimedii Folium, Polygalae Radix, Jujubae Fructus, Schisandrae Chinensis Fructus, Ziziphi Spinosae Semen, Ophiopogonis Radix, Testudinis Carapax Et Plastrum, Cervi Cornu Pantotrichum, Cervi Cornus Colla, Rehmanniae Radix Praeparata, Ginseng Radix Et RhizomaCuring neurasthenia, forgetfulness and insomnia, dizziness and palpitation, weariness of body, weak health and spontaneous perspiration, impotence and spermatorrhea“Zhong Yao Cheng Fang Zhi Ji” c
Qin Zhi Bi Yan Tang JiangXanthii Fructus, Scutellariae Radix, Angelicae Dahuricae Radix, Ephedrae Herba, Magno1iae Flos, Centipedae Herba, Menthae Haplocalycis HerbaTreating acute rhinitis“Chinese Pharmacopoeia (2015)” a
Cang Yi Di Bi YouXanthii Fructus, Angelicae Dahuricae Radix, Borneolum Syntheticum Curing nasosinusitis, nasal obstruction and runny nose“Zhong Yao Cheng Fang Zhi Ji”c
Cang Xin Qi Wu JiXanthii Fructus, Magno1iae Flos, Asari Radix Et Rhizoma, Angelicae Dahuricae Radix, Coptidis Rhizoma Curing nasal obstruction, rhinocnesmus, sneeze, allergic rhinitis, acute and chronic rhinitis“Guo Jia Zhong Cheng Yao Biao Zhun” b
Xin Yi Bi Yan PillsXanthii Fructus, Magno1iae Flos, Menthae Haplocalycis Herba, Perillae Folium, Glycyrrhizae Radix Et Rhizoma, Pogostemonis Herba, Centipedae Herba, Isatidis Radix, Angelicae Dahuricae Radix, Saposhnikoviae Radix, Houttuyniae Herba, Chrysanthemi Flos Treating allergic rhinitis, chronic rhinitis, nervous headache, cold and rhinorrhea, nasal obstruction“Zhong Yao Cheng Fang Zhi Ji” c
Xin Qin Chong JiXanthii Fructus, Asari Radix Et Rhizoma, Scutellariae Radix, Schizonepetae Herba, Saposhnikoviae Radix, Angelicae Dahuricae Radix, Astragali Radix, Atractylodis Macrocephalae Rhizoma, Cinnamomi Ramulus, Acori Tatarinowii RhizomaCuring allergic rhinitis due to deficiency of lung qi“Zhong Yao Cheng Fang Zhi Ji” c
Xin Qin TabletsXanthii Fructus, Asari Radix Et Rhizoma, Scutellariae Radix, Schizonepetae Herba, Saposhnikoviae Radix, Angelicae Dahuricae Radix, Astragali Radix, Atractylodis Macrocephalae Rhizoma, Cinnamomi RamulusCuring allergic rhinitis, deficiency of lung qi, exogenous pathogenic wind“Xin Yao Zhuan Zheng Biao Zhun” d
Xin Qin Ke LiXanthii Fructus, Asari Radix Et Rhizoma, Scutellariae Radix, Schizonepetae Herba, Saposhnikoviae Radix, Angelicae Dahuricae Radix, Astragali Radix, Atractylodis Macrocephalae Rhizoma, Cinnamomi Ramulus, Acori Tatarinowii RhizomaCuring rhinocnesmus, sneeze, rhinorrhea, cold, allergic rhinitis“Chinese Pharmacopoeia (2010)” a
Tong Qiao Bi Yan TabletsXanthii Fructus, Saposhnikoviae Radix, Astragali Radix, Magno1iae Flos, Atractylodis Macrocephalae Rhizoma, Menthae Haplocalycis HerbaCuring nasal obstruction, rhinorrhea, rhinocnesmus, forehead headache, chronic rhinitis, allergic rhinitis, nasosinusitis“Chinese Pharmacopoeia (2010)” a
Tong Qiao Bi Yan Jiao NangXanthii Fructus, Saposhnikoviae Radix, Astragali Radix, Magno1iae Flos, Atractylodis Macrocephalae Rhizoma, Menthae Haplocalycis HerbaCuring nasal obstruction, rhinorrhea, rhinocnesmus, forehead headache, chronic rhinitis, allergic rhinitis, nasosinusitis“Xin Yao Zhuan Zheng Biao Zhun” d
Tong Qiao Bi Yan Ke LiXanthii Fructus, Astragali Radix, Magno1iae Flos, Menthae Haplocalycis Herba, Saposhnikoviae Radix, Angelicae Dahuricae Radix, Atractylodis Macrocephalae RhizomaCuring nasal obstruction, rhinocnesmus, rhinorrhea, forehead headache, chronic rhinitis, allergic rhinitis, nasosinusitis“Chinese Pharmacopoeia (2015)” a
Fang Zhi Bi Yan TabletsXanthii Fructus, Chrysanthemi Indici Flos, Centipedae Herba, Angelicae Dahuricae Radix, Saposhnikoviae Radix, Ecliptae Herba, Paeoniae Radix Alba, Arisaema Cum Bile, Glycyrrhizae Radix Et Rhizoma, Tribuli FructusCuring sneeze, nasal obstruction, headache, allergic rhinitis, nasosinusitis“Zhong Yao Cheng Fang Zhi Ji”c
Bi Yan Qing Du JiXanthii Fructus, Chrysanthemi Indici Flos, Paridis Rhizoma, Zanthoxyli Radix, Prunellae Spica, Gentianae Radix Et Rhizoma, Codonopsis RadixTreating chronic inflammation of nasopharynx, swelling and pain in throat“Zhong Yao Cheng Fang Zhi Ji”c
Bi Yan Qing Du Ke LiXanthii Fructus, Chrysanthemi Indici Flos, Paridis Rhizoma, Zanthoxyli Radix, Prunellae Spica, Gentianae Radix Et Rhizoma, Codonopsis RadixTreating chronic inflammation of nasopharynx“Chinese Pharmacopoeia (2015)” a
Bi Yuan PillsXanthii Fructus, Magno1iae Flos, Lonicerae Japonicae Flos, Rubiae Radix Et Rhizoma, Chrysanthemi Indici FlosCuring nasal obstruction, nasosinusitis, ventilation lack, rhinorrhea, anosmia, headache, pain of superciliary ridge“Chinese Pharmacopoeia (2010)” a
Bi Yuan He JiXanthii Fructus, Magno1iae Flos, Lonicerae Japonicae Flos, Rubiae Radix Et Rhizoma, Chrysanthemi Indici FlosCuring nasal obstruction, nasosinusitis, ventilation lack, rhinorrhea, anosmia, headache, pain of superciliary ridge“Xin Yao Zhuan Zheng Biao Zhun” d
Bi Yuan TabletsXanthii Fructus, Magno1iae Flos, Lonicerae Japonicae Flos, Rubiae Radix Et Rhizoma, Chrysanthemi Indici FlosCuring chronic rhinitis, nasosinusitis“Zhong Yao Cheng Fang Zhi Ji”c
Bi Yuan Shu Kou Fu YeXanthii Fructus, Magno1iae Flos, Menthae Haplocalycis Herba, Angelicae Dahuricae Radix, Scutellariae Radix, Gardeniae Fructus, Bupleuri Radix, Asari Radix Et Rhizoma, Chuanxiong Rhizoma, Astragali Radix, Clematidis Armandii Caulis, Platycodonis Radix, PoriaCuring rhinitis, nasosinusitis“Chinese Pharmacopoeia (2010)” a
Bi Yuan Shu Jiao NangXanthii Fructus, Magno1iae Flos, Menthae Haplocalycis Herba, Angelicae Dahuricae Radix, Scutellariae Radix, Gardeniae Fructus, Bupleuri Radix, Asari Radix Et Rhizoma, Chuanxiong Rhizoma, Astragali Radix, Clematidis Armandii Caulis, Platycodonis Radix, PoriaCuring rhinitis, nasosinusitis“Chinese Pharmacopoeia (2010)” a
Bi Yuan Tong Qiao Ke LiXanthii Fructus, Magno1iae Flos, Ephedrae Herba, Angelicae Dahuricae Radix, Menthae Haplocalycis Herba, Ligustici Rhizoma Et Radix, Scutellariae Radix, Forsythiae Fructus, Chrysanthemi Indici Flos, Trichosanthis Radix, Rehmanniae Radix, Salviae Miltiorrhizae Radix Et Rhizoma, Poria, Glycyrrhizae Radix Et RhizomaCuring acute nasosinusitis, nasal obstruction, headache, fever“Chinese Pharmacopoeia (2015)” a
Bi Yan Ling PillsXanthii Fructus, Magno1iae Flos, Angelicae Dahuricae Radix, Asari Radix Et Rhizoma, Scutellariae Radix, Menthae Haplocalycis Herba, Fritillariae Cirrhosae Bulbus, Sojae Semen PraeparatumCuring nasosinusitis, nasal obstruction, chronic rhinitis“Zhong Yao Cheng Fang Zhi Ji” c
Bi Yan Ling TabletsXanthii Fructus, Magno1iae Flos, Angelicae Dahuricae Radix, Asari Radix Et Rhizoma, Scutellariae Radix, Fritillariae Cirrhosae Bulbus, Sojae Semen PraeparatumTreating chronic nasosinusitis, rhinitis, nasal obstruction and headache, anosmia“Zhong Yao Cheng Fang Zhi Ji” c
Bi Yan TabletsXanthii Fructus, Magno1iae Flos, Saposhnikoviae Radix, Forsythiae Fructus, Chrysanthemi Indici Flos, Schisandrae Chinensis Fructus, Platycodonis Radix, Angelicae Dahuricae Radix, Anemarrhenae Rhizoma, Schizonepetae Herba, Glycyrrhizae Radix Et Rhizoma, Phellodendri Chinensis Cortex, Ephedrae Herba, Asari Radix Et RhizomaTreating acute and chronic rhinitis, nasal obstruction, rhinorrhea, fever, headache“Chinese Pharmacopoeia (2010)” a
Bi Yan Tang JiangXanthii Fructus, Scutellariae Radix, Angelicae Dahuricae Radix, Ephedrae Herba, Magno1iae Flos, Centipedae Herba, Menthae Haplocalycis HerbaTreating acute rhinitis“Zhong Yao Cheng Fang Zhi Ji” c
Bi Dou Yan Kou Fu YanXanthii Fructus, Magno1iae Flos, Menthae Haplocalycis Herba, Platycodonis Radix, Bupleuri Radix, Angelicae Dahuricae Radix, Chuanxiong Rhizoma, Scutellariae Radix, Gardeniae Fructus, Poria, Clematidis Armandii Caulis, Astragali Radix Curing nasal obstruction due to wind-heat affecting lung, acute and chronic rhinitis, nasosinusitis“Chinese Pharmacopoeia (2010)” a
Bi Shu Shi TabletsXanthii Fructus, Chrysanthemi Indici Flos, Centipedae Herba, Angelicae Dahuricae Radix, Saposhnikoviae Radix, Ecliptae Herba, Paeoniae Radix Alba, Arisaema Cum Bile, Glycyrrhizae Radix Et Rhizoma, Tribuli Fructus Curing sneeze, rhinorrhea, nasal obstruction, headache, allergic rhinitis, chronic nasosinusitis“Zhong Yao Cheng Fang Zhi Ji” c
Bi Tong PillsXanthii Fructus, Magno1iae Flos, Angelicae Dahuricae Radix, Centipedae Herba, Menthae Haplocalycis Herba, Scutellariae Radix, Glycyrrhizae Radix Et RhizomaCuring affection of exogenous wind-heat, chronic rhinitis“Zhong Yao Cheng Fang Zhi Ji” c
a Cited from “Chinese Pharmacopoeia”; b Cited from “Guo Jia Zhong Cheng Yao Biao Zhun”; c Cited from “Zhong Yao Cheng Fang Zhi Ji”; d Cited from “Xin Yao Zhuan ZhengBiao Zhun”. Xanthii Fructus means the fruits of Xanthium strumarium L.
Table 2. Chemical constituents isolated from X. strumarium.
Table 2. Chemical constituents isolated from X. strumarium.
ClassificationNo.Chemical ComponentPart of PlantReference
Sesquiterpenoids1sibirolide AFruits[13]
2sibirolide BFruits[13]
3norxanthantolide AFruits[13]
4norxanthantolide B Fruits[13]
5norxanthantolide CFruits[13]
6norxanthantolide D Fruits[13]
7norxanthantolide EFruits[13]
8norxanthantolide FFruits[13]
91β-hydroxyl-5α-chloro-8-epi-xanthatinAerial parts[14]
1011α,13-dihydro-8-epi-xanthatinAerial parts[14]
Sesquiterpenoids11xanthininLeaves[15]
12xanthumin Leaves[15]
13xanthanolLeaves[15]
14xanthanol AcetateLeaves[15]
15isoxanthanolLeaves[15]
16xanthumanolLeaves[16]
17deacetoxylxanthuminLeaves[16]
18xanthatinLeaves[16]
19xanthinosinLeaves[16]
20tomentosinLeaves[16]
218-epi-tomentosinLeaves[17]
2211α,13-dihydroxanthuminolLeaves[18]
23desacetylxanthanolLeaves[18]
24(2E,4E,1’S,2’R,4’S,6’R)-dihydrophaseic acidFruits[19]
258-epi-xanthatin Aerial parts[20]
262-hydroxy xanthinosinAerial parts[21]
27lasidiol p-methoxybenzoateLeaves[18]
281β, 4β, 4α,5α-diepoxyxanth-11(13)-en-12-oic acidAerial parts[22]
2911α,13-dihydroxanthatinAerial parts[22]
304β,5β-epoxyxanthatin-1α,4α-endoperoxideAerial parts[22]
314-epi-xanthanolAerial parts[22]
324-epi-isoxanthanolAerial parts[22]
334-oxo-bedfordia acidAerial parts[22]
342-hydroxytomentosinAerial parts[20]
352-hydroxytomentosin-1β,5β-epoxide Aerial parts[20]
36xanthnonAerial parts[21]
376β,9β-dihydroxy-8-epi-xanthatinLeaves[23]
38inusoniolideAerial parts[21]
39(3S,5R,6S,7E)-5,6-epoxy-3-hydroxy-7-megastigmene-9-one Fruits[24]
40pungiolide EAerial parts[25]
41pungiolide AAerial parts[25]
42pungiolide DAerial parts[25]
435-azuleneacetic acidAerial parts[21]
44dihydrophaseic acid sodium salt 4’-O-d-glucopyranosideFruits[26]
45(3S,5R,6R,7E,9S)-megastigman-7ene-3,5,6,9-tetrol-3-O-d-glucopyranosideAerial parts[27]
Triterpenoids46betulinic acidRoots[28]
47betulinRoots[28]
48erythrodiolRoots[28]
49lup-20(29)-en-3β-olAerial parts[27]
Triterpenoids50lupenyl acetate Aerial parts[29]
51lupeol acetateWhole plants[30]
52β-amyrinAerial parts[31]
53oleanolic acidAerial parts[31]
54α-amyrinLeaves[32]
Phenylpropenoids551,3,5-tri-O-caffeoylquinic acidFruits[33]
563,5-di-O-caffeoylquinic acidFruits[33]
57neochlorogenic acid methyl esterFruits[34]
581,3-di-O-caffeoylquinic acidFruits[34]
59methyl-3,5-di-O-caffeoylquinic acidFruits[34]
60chlorogenic acid Fruits[35]
611,4-di-O-caffeoylquinic acid Fruits[35]
624,5-di-O-caffeoylquinic acid Fruits[35]
635-O-caffeoylquinic acid Fruits[35]
641,5-di-O-caffeoylquinic acidFruits[36]
653,4-di-caffeoylquinic acid methyl esterFruits[37]
663,5-di-caffeoylquinic acid methyl esterFruits[37]
674-O-caffeoyl quinic acid methyl esterFruits[38]
68N-trans-feruloyl tyramineRoots[39]
699,9’-O-di-(E)-feruloyl-(-)-secoisolariciresinolRoots[39]
70xanthiumnolic AFruits[40]
71xanthiumnolic CFruits[40]
722,3-dihydroxy-1-(4-hydroxy-3-methoxyphenyl)-propan-1-oneFruits[41]
73threo-guaiacylglycerol-8-O-4’- (coniferyl alcohol) etherFruits[42]
74erythro-guaiacylglycerol-8-O-4’-(coniferyl alcohol) ether Fruits[42]
75threo-1-phenyl-(4-hydroxy-3-methoxy)-2-phenyl-(4’’-hydroxy-3’’-methoxy)-1,3-propanediolFruits[42]
76(1S,2R)-1,2-bis(4-hydroxy-3-methoxyphenyl)-1,3-propanediolFruits[42]
77threo-guaiacylglycerol-β-coniferyl aldehyde etherFruits[42]
78erythro-guaiacylglycerol-β-coniferyl aldehyde etherFruits[42]
79xanthiumnolic DFruits[40]
80xanthiumnolic EFruits[40]
81ferulic acid Fruits[43]
82caffeic acidFruits[36]
83protocatechuic acidFruits[19]
84isovanillic acidWhole plants[30]
857-(4-hydroxy-3-methoxyphenyl)-1-phenylhept-4-en-3-oneRoots[28]
Phenylpropenoids86xanthiazone-(2-O-caffeoyl)-β-d-glucopyranosideWhole plants[44]
87rel-(2α,3β)-7-O-methylcedrusinFruits[42]
88caffeic acid choline esterFruits[38]
89icariside D1 Fruits[45]
903-methoxy-4-hydroxy-transcinnamaldehyde Fruits[24]
91methylchlorogenateFruits[46]
92icariside F2 Fruits[45]
93arbutin Fruits[45]
94coniferineFruits[45]
953-hydoxy-1-(4-hydroxy-phenyl)-propan-1-oneFruits[47]
96ω-hydroxypropioguaiacone Fruits[45]
97caffeic acid ethyl esterFruits[19]
984-hydroxy-3-methoxycinnamaldehydeFruits[37]
99p-hydroxybenzaldehyde Fruits[24]
Lignanoids100xanthiumnolic BFruits[40]
101(-)-1-O-d-glucopyranosyl-2-{2-methoxy-4-[1-(E)-propen-3-ol]phenoxyl}-propane-3-olFruits[48]
102leptolepisol DFruits[48]
103dihydrodehydrodiconiferyl alcoholFruits[48]
104chushizisin EFruits[48]
105(-)-(2R)-1-O-d-glucopyranosyl-2-{2-methoxy-4-[(E)formylviny1]phenoxyl}propane-3-olFruits[48]
106(-)-7R,8S-dehydrodiconiferyl alcoholFruits[48]
107(-)-simulanolFruits[48]
1082-(4-hydroxy-3-methoxyphenyl)-3-(2-hydroxy-5-methoxyphenyl)-3-oxo-1-propanolFruits[48]
109diospyrosinFruits[48]
110dehydrodiconiferyl alcoholFruits[48]
111balanophonin AFruits[48]
112threo-dihydroxydehydrodiconiferyl alcoholFruits[48]
Lignanoids1131-(4-hydroxy-3-methoxy)-phenyl-2-[4-(1,2,3-trihydroxypropyl)-2-methoxy]-phenoxy-1,3-propandiolFruits[48]
1147R,8S-dihydrodehydrodiconiferyl alcohol 4-O-d-glucopyranosideFruits[48]
115syringaresinolRoots[39]
116fructusol AFruits[42]
117balanophonin Fruits[24]
1184-oxopinoresinolRoots[28]
119pinoresinol Fruits[24]
Coumarins120jatrocin B Roots[39]
Coumarins121cleomiscosin A Roots[39]
122cleomiscosin C Roots[39]
123scopoletin Roots[39]
Steroids124stigmast-4-en-β-ol-3-one Roots[39]
125β-sitostenone Roots[39]
126β-sitosterol Fruits, Leaves[39]
127daucosterolFruits[39]
1285α,8α-epidioxy-22E-ergosta-6,22-dien-3β-ol Roots[39]
1296β-hydroxy-stigmast-4,22-dien-3-oneRoots[28]
1306β-hydroxy-stigmast-4-en-3-oneRoots[28]
1313-oxo-△(4,5)-sitostenoneRoots[28]
132β-daucosterolRoots[28]
133β-stigmasterolRoots[28]
1347-ketositosterolRoots[28]
135stigmasterolAerial parts[31]
136β-sitosterol-3-O-d-glucopyranosideAerial parts[31]
137ergosterolWhole plants[30]
138taraxasteryl acetateWhole plants[30]
1397α-hydroxy-β-sitosterol (stigmast-5-ene-3β,7α-diol) Fruits[24]
140stigmast-4-ene-3β,6α-diolFruits[24]
14114-methyl-12,13-dehydro-sitosterol-heptadeconateLeaves[32]
Glycosides142atractylosideFruits[49]
143carboxyatractylosideBurrs[50]
1443β-norpinan-2-one 3-O-d-apiofuranosyl-(1→6)-β-d-glucopyranoside Fruits[41]
145(6Z)-3-hydroxymethyl-7-methylocta-1,6-dien-3-ol 8-O-d-glucopyranoside Fruits[41]
146(6E)-3-hydroxymethyl-7-methylocta-1,6-dien-3-ol 8-O-d-glucopyranoside Fruits[41]
1477-[(β-d-apiofuranosyl-(1→6)-β-d-glucopyranosyl)oxymethy]-8,8-dimethyl-4,8-dihydrobenzo[1,4]thiazine-3,5-dioneFruits[41]
1483’,4’-dedisulphated-atractylosideFruits[46]
1492-methyl-3-buten-2-ol-β-d-ap-iofuranosyl-(1→6)-β-d-glucopyranosideFruits[51]
150everlastoside C Fruits[51]
Flavonoids151ononinFruits[43]
152quercetinFruits[37]
153allopatuletinFruits[37]
154patuletin-3-glucuronideFruits[34]
Flavonoids155quercetin-3-O-glucuronideFruits[34]
156formononetinFruits[43]
Tihiazdes157xanthiazoneFruits[36]
1582-hydroxy-xanthiazoneFruits[42]
1597-hydroxymethyl-8,8-dimethyl-4,8-dihydrobenzol[1,4]thiazine-3,5-dione-11-O-d-glucopyranosideFruits[43]
1602-hydroxy-7-hydroxymethyl-8,8-dimethyl-4,8-dihydrobenzol[1,4]thiazine-3,5-dione-11-O-d-glucopyranosideFruits[43]
1617-Hydroxymethyl-8,8-dimethyl-4,8-dihydrobenzol[1,4]thiazine-3,5-dione-(2-O-caffeoyl)-β-d-glucopyranosideFruits[52]
Anthraquinones & naphthoquinones162xanthialdehydeFruits[53]
163chrysophanic acidFruits[54]
164emodinFruits[54]
165aloe emodinFruits[54]
1665-hydroxy-3,6-dimethoxy-7-methyl-1,4-naphthalenedioneRoots[28]
Other compounds1675-methyluracilRoots[39]
168uracil Roots[39]
169sibiricumthionolFruits[19]
170indole-3-carbaldehyde Fruits[45]
171N-(1’-d-deoxyxylitolyl)-6,7-dimethyl-1,4-dihydro-2,3-quinoxalinedioneFruits[38]
172nonadecanoic acidRoots[39]
173hexadecanoic acidLeaves[32]
Table 3. Pharmacological effects of X. strumarium.
Table 3. Pharmacological effects of X. strumarium.
EffectsDetailExtracts/CompoundsConcentration/DoseIn Vivo/In vitroReference
Anti-AR effectsInhibiting C 48/80-induced systemic anaphylaxisWEXMice, 0.01–1 g/kg (p.o.)in vivo[61,62]
Inhibiting histamine and TNF-α released from RPMCWEXRPMC, 0.01–1 mg/mLin vitro[63]
Modulating the HMC-1- and PBMNC-mediated inflammatory and immunological reactionsWEXHMC-1, PBMNC, 0.25–1 mg/mLin vitro[63]
Inhibiting histamine and cAMP released from RPMCMEXRPMC, 20–500 μg/mLin vitro[64]
Ameliorate the nasal symptoms of OVA induced AR rats via anti-allergic; down-regulating IgE; anti-inflammatory and analgesic propertiesCXTRats, 5, 10, 20 mg/kg (p.o.)in vivo[65]
Anti-tumor effectsLung cancer
Growth inhibition by suppression of STAT3, GSK3β and β-cateninxanthatinCell lines of A549, H1975, H1299, H1650 & HCC827, 1–40 μMin vitro[66,67,68]
Triggering Chk1-mediated DNA damage and destabilization of Cdc25C via lysosomal degradationxanthatin
Cytotoxic effects on A549 cell8-epi-xanthatinIC50 = 1.1 μg/mLin vitro[17]
8-epi-xanthatin epoxideIC50 = 3.0 μMin vitro[69]
xanthatinIC50 = 1.3 μg/mLin vitro[17]
8-epi-xanthatin-1α,
5α-epoxide
IC50 = 9.5 μMin vitro[25]
1β-hydroxyl-5α-chloro-8-epi-xanthatinIC50 = 20.7 μMin vitro[25]
EEXAIC50 = 52.2 μg/mLin vitro[70]
Breast cancer
Cytotoxic effects on MDA-MB-231 cellsxanthatinIC50 = 13.9 μg/mLin vitro[71]
Cytotoxic effects on MDA-MB-231 cellsxanthinosinIC50 = 4.8 μg/mLin vitro[71]
Inhibiting cell growth via inducing caspase independent cell deathxanthatinMDA-MB-231 cells, 5–25 μMin vitro[72]
Anti-tumor effectsUp-regulating GADD45 γ tumor suppressor gene; inducing the prolonged expression of c-Fos via N-acetyl-l-cysteine-sensitive mechanismxanthatinMDA-MB-231 cells, 2.5–10 μMin vitro[73,74]
Cytotoxic effects on MFC7 cellsEEXAIC50 = 70.6 μg/mLin vitro[70]
Cervical cancer
Altering the antioxidant levelsWEXHela cells, 12.5–50 μg/mLin vitro[75]
Promoting apoptosis via inhibiting thioredoxin reductase and eliciting oxidative stressxanthatinHela cells, 5–20 µMin vitro[76]
Colon cancer
Cytotoxic effects on HCT-15 cellsxanthatinED50 = 1.1 μg/mLin vitro[17]
8-epi-xanthatinED50 = 0.1 μg/mLin vitro[17]
Cytotoxic effects on WiDr cellsxanthatinIC50 = 6.15 μg/mLin vitro[71]
xanthinosinIC50 = 2.65 μg/mLin vitro[71]
Cytotoxic effects on BGC-823 cellseremophil-1(10),11(13)-dien-12,8β-olideIC50 = 13.22 µMin vitro[77]
8-epi-xanthatin-1β,5β-epoxideIC50 = 2.43 µMin vitro[77]
tomentosinIC50 = 4.54 µMin vitro[77]
Cytotoxic effects on KE-97 cellseremophil-1(10),11(13)-dien-12,8β-olideIC50 = 4.41 µMin vitro[77]
8-epi-xanthatin-1β,5β-epoxideIC50 = 1.44 µMin vitro[77]
tomentosinIC50 = 3.47 µMin vitro[77]
Inducing G2/M cell cycle arrest and apoptosisxanthatinMKN-45 Cells, 3.9–18.6 µMin vitro[75]
Potentiating both extrinsic and intrinsic TRAIL-mediated apoptosis pathways and also
decreased the level of cell survival protein Bcl-2
xanthinosinAGS cells, 8 µMin vitro[18]
lasidiol p-methoxybenzoateAGS cells, 16 µMin vitro[18]
Cytotoxic effects on CT26 cellsEEXAIC50 = 58.9 μg/mLin vitro[70]
CFEEXAIC50 = 25.3 μg/mLin vitro[70]
Cytotoxic effects on AGS cellsfructusnoid CIC50 = 7.6 µMin vitro[79]
Liver cancer
Cytotoxic effects on SNU387 cells1β-hydroxyl-5α-chloro-8-epi-xanthatinIC50 =5.1 µMin vitro[25]
Cytotoxic effects on HepG2 cellsMEXLC50 = 112.9 μg/mLin vitro[80]
EAFMEXLC50 = 68.739 μg/mLin vitro[80]
Induction of apoptosis via inhibiting thioredoxin reductase and eliciting oxidative stressxanthatinHepG2 cells, 5–40 μMin vitro[76]
Meningioma
Cytotoxic effects on SK-MEL-2 cellsxanthatinED50 = 0.5 μg/mLin vitro[17]
8-epi-xanthatinED50 = 0.2 μg/mLin vitro[17]
Inhibiting melanin synthesis through downregulation of tyrosinase via GSK3β phosphorylationEEXSMel-Ab cells, 1–50 µg/mLin vitro[81]
Inhibiting cell proliferation associated with
activation of Wnt/β-catenin pathway and inhibition of angiogenesis
xanthatinB16-F10 cells, 2.5–40μMin vitro[82]
Mice, 0.1–0.4 mg/10 g(i.p.)in vivo[82]
Anti-tumor effectsLeukemia
Cytotoxic effects on P-388 cellsDFEEXAIC50 = 1.64 μg/mLin vitro[83]
Cytotoxic effects on HL-60 cellsxanthatinIC50 = 52.50 µg/mLin vitro[84]
Cytotoxic effects on Jurkat cellsMEXLC50 = 50.18 µg/mLin vitro[80]
EAFMEXLC50 = 48.73 µg/mLin vitro[80]
Other tumors
Cytotoxic effects on XF-498 cellsxanthatinED50 = 1.7 μg/mLin vitro[17]
8-epi-xanthatinED50 = 1.3 μg/mLin vitro[17]
Cytotoxic effects on S180 cellsWEXMice, 5–20 g/kgin vivo[85]
Cytotoxic effects on HEP-2 cellsCEXR12.5–100 µg/mLin vitro[86]
MEXR12.5–100 µg/mLin vitro[86]
Anti-inflammatory and analgesic effectsAnti-inflammatory
Inhibitting LPS-stimulated inflammatory WEX10, 100 and 1000 µg/mLin vitro[87]
Inhibitting LPS-stimulated inflammatoryMEX30, 60 and 90 mg/mLin vitro[88]
xanthatin and xanthinosinIC50 = 0.47 and 11.2 μMin vitro[89]
MEXLIC50 = 87 μg/mLin vitro[90]
MEXR50–400 μg/mLin vitro[91]
WEX0.5, 1 and 2 mg/mLin vitro[92]
MEX0–300 μg/mLin vitro[93]
MEXA0–300 μg/mLin vitro[94]
xanthiumnolic EIC50 = 8.73 μM.in vitro[26]
Inhibiting carrageenan induced hind paw edemaMEX100, 200 mg/kg/d (p.o.)in vivo[88]
WEX0.1, 0.5 and 1.0 g/kg, (p.o.)in vitro[95]
MEXL100, 200 and 400 mg/kg body weight.in vivo[90]
Inhibiting croton-oil-induced ear edema NFEEXMice, 0.5, 0.75 and 1.0 mg/earin vivo[96]
Inhibiting both PGE 2 synthesis and 5-lipoxygenase activityxanthatin100 and 97 mg/mL, respectivelyin vitro[84]
Inhibiting production of TARC/CCL17 and MDC/CCL22 induced by TNF-α/IFN-γEEX10 μg/mLin vitro[97]
Analgesic effect
Ameliorating HCl/EtOH-induced gastritis lesionsMEXA50 and 200 mg/kg (p.o.)in vivo[94]
Analgesic effect on acetic acid-induced abdominal constriction test and a hot plate testMEX100, 200 mg/kg/d (p.o.)in vivo[88]
Reducing the number of writhings induced by acetic acidNFEEXMice, 100,200 and 400 mg/kg body wt.in vivo[96]
Analgesic effect on writhing and formalin testsWXF0.1, 0.5 and 1.0 g/kg, (p.o.)in vivo[95]
Analgesic effect on hot plate test, acetic acid induced writhing test and formalin testEEX250 and 500 mg/kg body weightin vivo[98]
Insecticide and antiparasitic effectsAntiplasmodial activity against T. evansiEEXL5, 50, 500 and 1000 µg/mLin vitro[99]
100, 300 and 1000 mg/kg (i.p.)in vivo[99]
Insecticidal effects against T. b. brucei xanthatinIC50 = 2.63 µg/mLin vitro[84]
Anti-insect effects towards P. viteanaMEXLC50 = 11.02 (w/w)in vitro[100]
Insecticide and antiparasitic effectsAntiplasmodial activity against P. bergheiEEXLIC50 = 4 µg/mLin vitro[101]
Insecticidal properties against C. chinensisWEXL1%, 2% and 4% concentrationin vitro[102]
Anti-nematode activity against Meloidogyne javanicaEEX3%, 6% and 12% concentrationin vitro[103]
Insecticidal effects against A. caspius, C. pipiensMEXLC50 = 531.07 and 502.32 μg/mL, respectivelyin vitro[80]
Antioxidant effectsScavenging DPPHCEXR and MEXRLC50 = 10.28 and 40.40 µg/mLin vitro[86]
WEX0.05–0.2 mg/mLin vitro[95]
EEXR and CEXRIC50 = 29.81 and 24.85 µg/mLin vitro[106]
EEXLIC50 = 85 µg/mLin vitro[107]
Scavenging DPPHhexadecanoic acid;
α- amyrin; 14-methyl-12, 13-dehydro-sitosterol-heptadeconate
IC50 = 106.4, 64.16 and 76.18 µg/mLin vitro[32]
Scavenging DPPHEOX138.87 μg/mLin vitro[108]
MEXNot mentionedin vitro[28]
Scavenging nitric oxideEEXR and CEXRIC50 = 395.20 and 415.80 µg/mLin vitro[106]
EEXLIC50 = 72 µg/mLin vitro[107]
Scavenging hydrogen peroxideEEXR and CEXRIC50 = 10.18 and 9.23 µg/mLin vitro[106]
EEXLIC50 = 62 µg/mLin vitro[107]
Increasing of superoxide dismutase, glutathione peroxidase, glutathione reductase and catalase contentsPEEXW250 and 500 mg/kg body weight (p.o for 20 days)in vivo[104]
Liposome protectionWEX0.05–0.2 mg/mLin vitro[95]
Scavenging ABTSWEX0.05–0.2 mg/mLin vitro[95]
Reducing activityWEX0.05–0.2 mg/mLin vitro[95]
Increasing of SOD, CAT, GSH and GPx contentsMEXS100 and 200 mg/kg (p.o., for 10 days)in vivo[105]
Superoxide anionEEXR and CEXRIC50 = 495.30 and 418.30 µg/mLin vitro[106]
Scavenging hydroxyl radicalshexadecanoic acid;
α- amyrin; 14-methyl-12, 13-dehydro-sitosterol-heptadeconate
IC50 = 127.4, 83.96 and 84.4 µg/mLin vitro[32]
FRAP antioxidant activityMEXNot mentionedin vitro[28]
Antibacterial and antifungal effectsAntibacterial
Inhibitory effects against V. choleraeWEXFTNot mentionedin vitro[109]
Inhibitory effects against S. epidermidis, B. cereus, K. pneumoniae, P. aeruginosa and S. fyphixanthatinMIC = 31.3, 62.5, 31.3, 125 and 125 µg/mLin vitro[110]
Inhibitory effects against K. pneumoniae, P. vulgaris, P. Aeruginosa, P. putida, S. typhimurium, B. cereus, B. subtilis, S. epidermidisMEXL500 and 100 mg/mLin vitro[111]
Inhibitory effects against E. coliβ-sitosterol and β-daucosterolMIC = 0.17 and 0.35 µg/mLin vitro[112]
Inhibitory effects towards K. pneumonia, P. mirabilis, E. coli, B. subtilis, E. faecalis, S. aureusMEXL50, 100, 150, 200 and 250 mg/mL, respectivelyin vitro[113]
WEXL
Antibacterial and antifungal effectsInhibitory effects against S. aureus, B. subtilis, K. pneumoniae and P. aeruginosaEOXLMIC = 0.5, 1.3, 4.8 and 20.5 µg/mL, respectivelyin vitro[114]
Inhibitory effects against Shiga toxin-producing E. coliEOXL30, 60 and 120 mg/mLin vitro[115]
Inhibitory effects against S. aureus and E. coliWEXMIC = 31.25 and 7.81 mg/mL, respectivelyin vitro[116]
Inhibitory effects against R. toxicus, S. aureus and P. S. syringaeEOXMIC = 25, 50 and 50 µg/mL, respectivelyin vitro[108]
Antifungal
Inhibitory effects against P. drechslerideacetylxanthuminMIC = 12.5 µg/mLin vitro[117]
Inhibitory effects against P. infestansMEXMIC = 2.0% w/vin vitro[118]
Inhibitory effects against C. albicans and A. nigerEOXLMIC = 55.2 and 34.3 µg/mL, respectivelyin vitro[114]
Inhibitory effects against P. oryzae and F. oxysporumEOXMIC = 12.5 and 50 µg/mL, respectivelyin vitro[108]
Inhibitory effects against A. niger, A. flavus, F. oxysporum, F. solani, A. alternata and P. digitatumEOXLMIC = 8 µg/mL and MFC = 8 µg/mLin vitro[119]
Antidiabetic effectsExhibiting potent hypoglycemic activityWEX15 and 30 mg/kg (i.p.)in vivo[120]
Decreasing the plasma glucose in diabetic ratscaffeic acid0.5–3 mg/kg (i.v.)in vivo[121]
Decreasing the blood glucose and HbA1C level and increase the level of insulinMEXS100 and 200 mg/kg (p.o., for 30 days)in vivo[105]
Inhibitory effect against rAR and rhARmethyl-3,5-di-O-caffeoylquinateIC50 = 0.30 and 0.67 µM, respectivelyin vivo[47]
Inhibitory effect against α-glucosidaseCFMEXLIC50 = 72 µg/mLin vitro[122]
Inhibitory effect against α-glucosidase MEXIC50 = 15.25 µg/mLin vivo[28]
Antilipidemic effectsDecreasing plasma cholesterol, triglyceride, LDL, and VLDL and increasing plasma HDL levelsCEXR and EEXR200 and 400 mg/kg (p.o.)in vivo[106]
Improving lipid homeostasisWEX570 and 1140 mg/kg (p.o., for 6 weeks)in vivo[123]
Decreasing blood glucose, TC, TG, LDLC levels and increasing HDLC levels.WEX3.7 and 11.11 g/kg (p.o., for 4 weeks)in vivo[124]
Antiviral activityAntiviral activity against duck hepatitis B virusWEX0.01, 0.1 and 1 g/kg (i.g., for 10 days)in vivo[125]
Antiviral activity against Influenza A virusnorxanthantolide FIC50 = 6.4 µMin vitro[13]
2-desoxy-6-epi-parthemollinIC50 = 8.6 µMin vitro[13]
xanthatinIC50 = 8.4 µMin vitro[13]
threo-guaiacylglycerol-8′-vanillic acid etherIC50 = 8.4 µMin vitro[13]
caffeic acid ethyl esterIC50 = 3.7 µMin vitro[13]
Other pharmacological effectsAnti-septic activityCXT10, 20 and 40 mg/kg(i.p.)in vivo[126]
Attenuating hepatic steatosisWEX570 and 1140 mg/kg (p.o., for 6 weeks)in vivo[127]
Anti-arthritic effectEEX75 and 300 mg/kg (p.o.)in vivo[128]
Other pharmacological effectsAnti-pyretic activityMEXW200 and 400 mg/kg (p.o.)in vivo[129]
Anti-epileptic activityPEEXW250 and 500 mg/kg (p.o., for 20 days)in vivo[130]
Antiurolithiatic effectHEEXB500 mg/kg (p.o.)in vivo[131]
Antiulcer effectEEXL200 and 400 mg/kgin vivo[132]
Cardioprotective effectCXT10, 20 and 40 mg/kg (p.o.)in vivo[133]
Table 4. Toxicities and side effects of X. strumarium.
Table 4. Toxicities and side effects of X. strumarium.
Extracts/CompoundsAnimal/SubjectsLD50/Toxic Dose RangeToxic ReactionsReference
WEXmiceLD50 = 201.14 g/kg (i.g., crude herb mass equivalent)Death[139]
WEXmiceLD50 = 167.60 g/kg (i.g., crude herb mass equivalent)Death[140]
EEXmiceLD50 = 275.41 g/kg (i.g., crude herb mass equivalent)Death[140]
WEXmiceLD50 = 194.15g/kg (i.g., crude herb mass equivalent)Death[141]
carboxyatractylosideswine10–100 mg (i.v.)Death[142]
atractylosidemice50–200 mg/kg (i.p.)Increasing contents of ALT, AST, ALP, MDA in mice serum[143]
carbxyatractylosidemice50–150 mg/kg (i.p.)Increasing contents of ALT, AST, ALP, MDA in mice serum[143]
NFEEXmice0.06, 0.3, 0.7 g/kg (i.g., for 28 days)Weight loss, enlarged hepatic cell space, karyolysis and inflammatory cell infiltration[145]
WFEEXmice0.06, 0.3, 0.7 g/kg (i.g., for 28 days)Weight loss, enlarged hepatic cell space, karyolysis, and inflammatory cell infiltration[145]
WEXmice21.0 g/kg (i.g., for 28 days)Weight loss and increase of ALT, AST in mice serum[146]
WEXmice7.5, 15.0 and 30.0 g/kg (i.g., for 5 days)Increasing contents of VLDL/LDL, β-HB, glutamate, choline, acetate, glucose in serum[147]
WEXmice16.7 g/kg (i.g., for 7 days)Increasing contents of GLDH, α-GST and decreasing miRNA-122 [148]
MEXAmice100, 200, 300 mg/kgDepressing the action of central nervous system[149]
atractylosiderat hepatocytes0.01–0.05 g/LReducing cell viability and intracellular GSH content[150]
atractyloside, carbxyatractylosideL-02 cells, BRL cells 100 μmol/L for 48 hInhibiting cell proliferation, improving LDH activity [147]
WEXHK-2 cells100 μg/mLInhibiting cell proliferation[151]
HEEXACHO cells25–100 μg/mLInducing DNA damage[152]
EFEEXMIHA cells IC50 = 231.1 μg/mlDecreasing viability of cell[153]
WEXzebrafish15 μg/mLDecreasing hatch rate[154]

Share and Cite

MDPI and ACS Style

Fan, W.; Fan, L.; Peng, C.; Zhang, Q.; Wang, L.; Li, L.; Wang, J.; Zhang, D.; Peng, W.; Wu, C. Traditional Uses, Botany, Phytochemistry, Pharmacology, Pharmacokinetics and Toxicology of Xanthium strumarium L.: A Review. Molecules 2019, 24, 359. https://doi.org/10.3390/molecules24020359

AMA Style

Fan W, Fan L, Peng C, Zhang Q, Wang L, Li L, Wang J, Zhang D, Peng W, Wu C. Traditional Uses, Botany, Phytochemistry, Pharmacology, Pharmacokinetics and Toxicology of Xanthium strumarium L.: A Review. Molecules. 2019; 24(2):359. https://doi.org/10.3390/molecules24020359

Chicago/Turabian Style

Fan, Wenxiang, Linhong Fan, Chengyi Peng, Qing Zhang, Li Wang, Lin Li, Jiaolong Wang, Dayong Zhang, Wei Peng, and Chunjie Wu. 2019. "Traditional Uses, Botany, Phytochemistry, Pharmacology, Pharmacokinetics and Toxicology of Xanthium strumarium L.: A Review" Molecules 24, no. 2: 359. https://doi.org/10.3390/molecules24020359

Article Metrics

Back to TopTop