Open access peer-reviewed chapter - ONLINE FIRST

Dietary Plant Flavone Cynaroside and Its Biological Significance

Written By

Sabina Gayibova, Eva Ivanisova and Ulugbek Gayibov

Submitted: 22 January 2024 Reviewed: 26 April 2024 Published: 03 June 2024

DOI: 10.5772/intechopen.1005623

Herbs and Spices - New Perspectives in Human Health and Food Industry IntechOpen
Herbs and Spices - New Perspectives in Human Health and Food Indu... Edited by Eva Ivanišová

From the Edited Volume

Herbs and Spices - New Perspectives in Human Health and Food Industry [Working Title]

Ph.D. Eva Ivanišová

Chapter metrics overview

17 Chapter Downloads

View Full Metrics

Abstract

Flavonoids, the most diverse group of natural polyphenolics, are secondary plant metabolites that play a crucial role in human health protection. Two main classes—flavonols and flavones—comprise the main body of flavonoids with antioxidant properties and high biological activity, proven both in vitro and in vivo. Purified samples of flavones represent special interest. One of them, luteolin-7-glucoside (cynaroside), has attracted increasing attention as a potential agent possessing a number of biological activities. The current understanding of cynaroside bioactivities is outlined in this chapter, along with research gaps and potential future directions for this flavonoid’s study.

Keywords

  • antioxidant
  • anticancer
  • cardio-protection
  • cynaroside
  • flavone
  • hepatoprotection

1. Introduction

Flavonoids assigned as low molecular weight secondary metabolism phytochemicals perform various biological properties both manifested in vitro and in vivo [1]. Extensive range of studies strongly suggest that long-term consumption of diets rich in plant flavonoids offers protection against development of cancers [2], cardiovascular diseases [3], diabetes [4], neurodegenerative diseases [5], and so on. By now, the mechanisms of flavonoids action have been proposed that include the influence of signaling processes, flowing in living systems, due to the specific interaction with regulatory proteins [6], the modification of eicosanoid biosynthesis [7], the prevention of platelet aggregation [8], the promotion of relaxation of cardiovascular smooth muscle cells [9], and so forth.

Flavones and flavone-derived compounds are a large group of flavonoids with a great number of properties and presented in vegetables, fruits, and aromatic plants [10]. The term “flavone” was used for the first time in 1895 by von Kostanecki and Tambor who were pioneers in the structural work of this particular class of flavonoids [11].

Cynaroside—which is 7-O-glucoside of luteolin (other names are luteolin-7-O-β-D-glucopyranoside or luteolin-7-O-β- Dglucopyranoside-5,3′,4′-trihydroxyflavone, Luteoloside [12], luteolin-7-glucoside, nephrocizin [13])—is a natural flavone, one of the bioactive compounds purified from various genuses of plant families: Apiaceae [14], Lamiaceae [15], Asteráceae [16], and so on (Figure 1 and Table 1).

Figure 1.

Chemical structure of cynaroside.

FamilySpecies
ApiaceaePimpinella anisum L. [10], Daucus carota [17], Angelica keiskei [18], Anthriscus sylvestris (L.) Hoffm. [19], Dystaenia takeshimana [20], Ferula varia [21]
AsteraceaeCynara scolymus [22], Ixeris chinensis [22], Chrysanthemum morfolium Ramat [23], Glossogyne tenuifolia [24], Tanacetum parthenium [25], Taraxacum officinale [25], Scorzonera cana var. jacquiniana [26], Scorzonera cinerea [26], Scorzonera eriophora [26], Scorzonera incisa [26], Scorzonera parviflora [26], Dendranthema morifolium [27], Chrysanthemum indicum Linnén. [28], Chrysanthemum morifolium [28], Achillea millefolium L. [29], Artemisia rupestris [30], Carduus crispus [31], Bidens tripartite [32], Cirsium canum (L.) [32], Smallanthus sonchifolius [33]
BalsaminaceaeImpatiens textori [33]
CaprifoliaceaeLonicera japonica [34], Lonicera maackii [35]
FabaceaeLeucaena leucocephala [36], Vicia pannonica var. purpurascens [37]
JuncaceaeJuncus jerardi Lois., Luzula silvatica, Juncus ariculatus, Juncus conglameratus, Juncus filiformis, Juncus tenuis [38]
LamiaceaeSalvia officinalis [39], Scutellaria immaculata and Scutellaria ramosissima [39], Lycopus europaeus L. [40], Elsholtzia blanda (Benth.) Benth [41], Phlomis younghusbandii [42], Melittis melissophyllum L. [43]
MyrtaceaePimenta racemosa [44]
OleaceaeLigustrum lucidum Ait [45], Phillyrea latifolia [46], Ligustrum delavayanum and L. vulgare [47]
PolygonaceaePolygonum orientale [48], Rumex hastatus [15]
PteridaceaePteris multifida [49]
RosaceaeCrataegus monogyna [50], Agrimonia pilosa Ledeb [51], Prunus mume [52]
RubiaceaeOphiorrhiza mungos Linn [52]
SalicaceaeSalix matsudana [53]
ScrophulariaceaeVerbascum densiflorum and V. Phlomoides [54], Verbascum nubicum [55]

Table 1.

Natural sources of cynaroside.

The name “cynaroside” originates from the latin term Cynara denoting the name of the genus of artichokes where this flavone was firstly isolated in 1955 and defined to be glycoside in 1958 by Michaud (J.) and Masquelier (J.) correspondingly [56, 57]. Since that, interest to cynaroside has been growing considerably. For instance, due to electronic database PubMed, search for the term “cynaroside, luteolin-7-O-glucoside, nephrocizin” shows growing trend in publications (Figure 2).

Figure 2.

Trend of research in the field of cynaroside represented by PubMed database.

Several lines of evidence have emerged in recent years, suggesting that cynaroside, despite its nontoxic nature, can minimize hepatotoxicity [58]; repair the antioxidant system [59]; normalize energy, lipid, and carbohydrate metabolism; and enhance bile flow [60]. Therefore, scientific interest in flavones as therapeutic agents is rapidly increasing. The most interesting biological activities of cynaroside are summarized in Figure 3.

Figure 3.

Some biological activities of cynaroside.

The purpose of this chapter was to provide an overview of the distribution and biological properties of cynaroside. In this paper, we concentrate our attention on the most common knowledge about the biological effects of plant flavone cynaroside in the context of relevance to influencing human physiology and therefore interesting from a general perspective of improving human well-being.

The electronic databases included PubMed, and Google Scholar was initially used to find articles (published as early as 1955 up to manuscript submission). More than 100 articles were found in applied databases and manually screened sources. Reported publications focus on the cynaroside identification in plants, isolation, its biological and pharmacological properties in vivo and in vitro, but any articles summarizing or analyzing all this information were not observed. As was mentioned above, the search terms included “cynaroside” as well as “luteolin-7-glucoside” and “nephrocizin” (the mostly used alternative terms for “cynaroside”). Firstly, the search was limited to English-Language articles. Citation lists of retrieved articles were screened manually to ensure sensitivity of the search strategy. References of the included papers were then hand searched to identify in potential relevant studies in other languages (only abstract was used in Russian, French, and Chinese articles). Only published articles were included. No protocol was developed for this review.

Advertisement

2. Potential health benefits

Even though there is some data on LD50 of the plant extracts abundant on cynaroside [55], acute toxicity data on pure cynaroside are not available. In general, in vivo experiments carried on rats showed that the administration of cynaroside to six-week-old male Wistar rats for 1 week did not change animals’ food and beverage consumption, as well as animal body weights when compared to the control [1]. Thus, the rats treated with pure cynaroside up to a dose of 30 mg/kg did not demonstrate any signs of hepatotoxicity [61].

Advertisement

3. Hepatoprotective effect

The proposed hepatoprotective effect of cynaroside was due to the great importance of Cynara (artischocke) abundant in the flavonoid in folk medicine against liver complaints.

In vivo carbon tetrachloride (CCl4)-induced toxicity model revealed that pretreatment of experimental rats with cynaroside suppressed the elevation of alanine aminotransferase (ALT, GPT, and glutamic-pyruvic transaminase), aspartate transaminase (AST, GOT, and glutamic oxaloacetic transaminase), malondialdehyde (MDA), and 8-Oxo-2′-deoxyguanosine (8-OHdG) and inhibited the reduction of glutathione (GSH) in a dose-dependent manner [61]. In vitro experiments with fresh hepatocytes, primary hepatocyte cultures, and immortalized cell lines also showed the protective effect of cynaroside from cell damage induced by carbon tetrachloride (CCl4) [61], as well as brombenzene [62] and t-BHP [63]. Interestingly, Adzet et al. [64] observed specific GOT-recovering effect on isolated hepatocytes preincubated with cynaroside.

However, Quisheng et al. [60] in vivo demonstrated dose-dependent decline of both AST and ALT levels in blood serum. It is well accepted that increased enzyme levels are indicative of a tissue injury: the attenuated increase of serum AST and ALT indicates a direct protective role in hepatocytes; while AST is not specific to the liver, any elevation of the ALT is a direct indication of a liver injury. In the meanwhile, some authors mark that ALT and AST have relatively long half-lives (T1/2) (literature estimates approximately 17 and 47 hours, respectively) [65, 66] and thus do not reflect immediate changes in liver injury or recovery [67]. Therefore it could be worth considering that in the work of Adzet et al., the period of incubation of cynaroside with toxicated cells did not last more than 1 hour, while in the work of Quisheng et al. [60], all treated animals were sacrificed 24 hours after receiving the administration of the LUTG or hepatotoxin. Thus, it might be worth considering the preliminary incubation time as one important factor in cynaroside hepatoprotection.

Cynaroide in vitro also recovered significant (up to 81%) GSH level in tBHP-induced HepG2 cells after 5 hours of incubation [63], which was even higher than by luteolin (53%) and twice higher than quercetin (40%), and in vivo cynaroside was able to attenuate the CCl4-induced reduction of GSH in a dose-dependent manner [61, 63]. On the other hand, cynaroside did not have any effect on the recovery level of GST either on cell lines in vitro or on cells isolated from rats that were subjected to brombenzene in vivo [62]; the effect of cynaroside on its own in normal conditions was not shown by the authors.

It is known that GSH conjugates can be formed directly or be catalyzed by GST [68]. All the toxins mentioned above (CCl4, brombenzene, t-BHP) are known to initiate the formation of free reactive species. Thus, CCl4 intoxication metabolized by CYP2E1 is forced by the prevention of lipid peroxidation by decreasing the level of LPO and reducing the level of MDA. Wang et al. [69] found that cynaroside affected some isoforms of P450, namely, isoforms CYP2C9, 3A4, and 1A2; however, no effect was found on 2E1 isoform. That lets to propose that in the case of CCl4 intoxication, cynaroside does not have an effect of toxin metabolization, but protects the lipids through its antiradical properties against CCl3 metabolite.

The results of Park et al. [62] indicate a protective effect of cynaroside in reducing the degree of liver lipid peroxidation caused by the hepatotoxin bromobenzene in vivo in rats. Thus, cynaroside at a concentration of 10 mg/kg almost completely neutralized the process of lipid peroxidation caused by hepatoxin and restored the level of the enzyme epoxide hydrolase (an enzyme that deactivates bromobenzene). Bromobenzene is converted by P450 enzymes to its reactive, toxic metabolite bromobenzene 3,4-epoxide that is metabolized to a nontoxic bromobenzene 3,4-dihydrodiol by either epoxide hydrolase or glutathione S-transferase. Cynaroside even not effecting GST was nevertheless shown to recover the level of epoxide hydrolase, suggesting that cynaroside might improve the level of hydrolase and also act as a radical scavenger.

There are two pathways by which tBHP is metabolized; both of them induce oxidative stress. The first, provided by cytochrome P450, leads to the production of peroxyl and alkoxyl radicals [70]. These radicals initiate lipoperoxidation of membrane phospholipids with subsequent alterations to membrane fluidity and permeability. The other pathway employs glutathione peroxidase. tBHP is detoxified to tert-butanol, and GSH is depleted by oxidation to its disulfide form (GSSG) [71].

Results showed that cynaroside protects from AST and ALT leakage and also strongly suggest the ability of cynaroside in protecting hepatocyte against membrane fragility. Recent findings have demonstrated that the carbon ring B is deeply immersed into lipid bilayer. A ring is adjusted toward the aqueous environment; the glycosylated form (in position 7) is dispersed over the lipid bilayer, thus providing the defense for double bonds from oxidation [72].

Advertisement

4. Cardio-protective effect

It has been shown that cynaroside in pharmacological concentrations has a positive inotropic and vasodilator effect. On the models of isolated hearts of rabbits and guinea pigs, perfused using the Langendorff technique, it was shown that cynaroside increases pressure in the left ventricle, accelerates the process of blood ejection into the arterial system, and enhances the total and relative coronary blood flow and, consequently, the supply oxygen for nonischemic myocardial damage [73, 74].

Also, in the last decade, the processes of apoptosis in cardiovascular pathologies have been intensively studied, and more and more researchers are assigning a leading role to apoptosis in the pathogenesis of various cardiovascular diseases. Xiao Sun et al. [70] demonstrated that cynaroside exhibits cytoprotective properties in relation to the H9C2 cardiac myocyte cell line against H2O2-induced apoptosis. Cardiomyocyte cells exposed to H2O2 suffered severe damage accompanied by apoptosis. Preincubation of cells with cynaroside reduced the degree of ROS generation and inhibition of caspase activation both in the mitochondrial pathway and through the so-called death receptors by enhancing the endogenous antioxidant activity of superoxide dismutase, glutathione peroxidase, and catalase, thereby inhibiting the formation of intracellular ROS. In addition, cynaroside supported normal mitochondrial function by regulating the expression of Bcl-2 protein (B-cell lymphoma protein 2, apoptosis regulator protein Bcl-2), as well as the expression of protein kinases JNK (c-Jun N-terminal kinase, mitogen-activated protein kinase) and p53 (cell cycle regulating transcription factor). The results of microscopic studies showed that after incubation with cynaroside, there was a dose-dependent restoration of the morphological structures of cardiomyocytes damaged by H2O2, such as a dense membrane packing compared to a loose membrane layer, normal physiological spindle shape versus cell swelling, round shape of the nucleus in comparison with vacuolar degeneration, and so forth. An analysis of generalized experimental data also indicates some possible mechanisms of the protective modulating action of cynaroside in certain pathological conditions of the heart, which indicates an urgent need for further in-depth study of the mechanisms of the action of cynaroside and also to consider it as the basis for the creation of potentially effective and safe cardioprotective agents that can make a significant contribution to the treatment and prevention of a number of cardiovascular diseases.

Advertisement

5. Anticancer effect

Evidences are accumulating that pure cynaroside possesses antiproliferative and cytotoxic effects on the tested cancer cell lines with minimal toxicity toward the normal (VERO) cells, indicating cancer cell-specific cytotoxic (apoptotic) effect. Various researchers mentioned that cynaroside does not cause cytotoxicity in relation to normal cells (human neonatal epidermal keratinocytes (HEKn) [75], VERO [76], non-tumorigenic BEAS-2B [77], and Huh7 [78].

Luteolin-7-glucoside in more or less extent has been shown to inhibit a proliferation of a series of human cancer cell lines: breast (MCF-7 [76, 77, 79, 80], MDA-MB-231 [80], A549 [76, 80], H292 [81], renal [77], colorectal (COLO 320 DM [76], gastric (AGS [76], liver (HepG-2 [78, 79, 80], Hep 3B [80, 82], LO2 [83], SNU-449 [82], Huh-7 [82], SMMC-7721 [82], MHCCLM3 [82] and MHCC97-H [82], cervical (HeLa [79], melanoma (UACC-62 [77], and some others (mouse fibroblast cell line L929 [84]. No significant effect was observed on macrophage RAW264.7 cells, Ehrlich ascites carcinoma [36], and Rhabdomyosarcoma [12].

Advertisement

6. Anti-inflammatory activity

Palombo et al. [75] in their study on a model of psoriasis showed the ability of luteolin-7-glucoside to regulate proliferative responses, as well as differentiation of cultured human keratinocytes treated with interleukin 22 (IL-22). It was found that local administration of cynaroside leads to a noticeable reduction in acanthosis. Thus, treatment with cynaroside led to a decrease in the expression of proliferation markers, an increase in the production of differentiation markers, and, in general, to a phenotypic improvement.

Other in vivo tests have shown that luteolin-7-O-glucoside is responsible for wound healing activity. It has also been found to have significant anti-inflammatory, antioxidant, anti-hyaluronidase and anti-collagenase activities. Phases in the healing processes (inflammation, proliferation, and remodeling) were observed and recorded in the experimental groups to varying degrees. All tissues treated with cynaroside ointments showed good healing processes with the potential for faster reepithelialization and high collagen concentrations compared to other groups tested. The anti-inflammatory activity of cynaroside was mainly demonstrated by the inhibition of nuclear factor-kappa B (NF-kappa B), cyclooxygenase-2 (COX-2), 5-lipoxygenase (5-LOX), and inducible NO synthase (iNOS) [85, 86].

Advertisement

7. Radical scavenging activity

An analysis of the scientific literature has shown that cynaroside has significant antiradical (ARA) and antioxidant (AOA) properties in relation to free radicals and ROS molecules (superoxide anion (O2•−) [80], hydroxyl radical (HO) [87], peroxyl radicals (ROO) [23], and nitric oxide (NO) [85]. ARA of cynaroside using the DPPH method is quite widely presented in various works [23, 88]. It has been shown that cynaroside has a fairly high efficiency in the reduction of DPPH molecules (IC50 value varies in different studies: from 13.90 ± 1.46 μM to 277.3 ± 14.9 mmol/mol DPPH. In addition, it was found that cynaroside dose-dependently inhibits the formation of superoxide anion induced by various stimuli, such as N-formylmethionyl-leucyl-phenylalanine, human polymorphonuclear neutrophils and peripheral blood mononuclear cells, phorbol-12-myristate-13-acetate (PMA) and arachidonic acid (AA) in human neutrophils, superoxide anion produced by the xanthine/xanthine oxidase system, and so on [89]. Hsu et al. [80] showed that cynaroside prevents DNA dissociation caused by hydroxyl radical. Cynaroside at a maximum concentration of 1.8 μg/ml provided 64.0 (3.9%) and 83.1 (2.9%) protection of supercoiled DNA against nonspecific and site-specific oxidation caused by hydroxyl radical, respectively. In comparison, 4 μg/ml Trolox (a water-soluble vitamin E analog) provided 83.5 (0.5%) and 42.6 (2.5%) protection. Cynaroside also effectively slows down lipid peroxidation caused by peroxyl radical as well as reduces the concentration of nitric oxide (NO) free radicals produced by endothelial nitric oxide synthase (eNOS) under pathological conditions [85].

Advertisement

8. Antibacterial activity

Isolated cynaroside also exhibits antimicrobial activity, especially against gram-negative bacteria (Escherichia coli, Streptococcus pyogenes, and Micrococcus luteus with minimal inhibition zone of 0.5 mm), suppresses the formation of biofilm of gram-positive Pseudomonas aeruginosa and Staphylococcus aureus, and increases the frequency of mutations that reduce the resistance of Salmonella typhimurium to ciprofloxacin. Cynaroside also exhibits high activity against yeast fungi Candida albicans, Candida lusitaniae, Saccharomyces cerevisiae, and Saccharomyces carlsbergensis and molds Aspergillus niger, Penicillium oxalicum, Mucor mucedo, and Cladosporium cucumerinum [19].

Advertisement

9. Anti-coronavirus activity

The World Health Organization (WHO) welcomes the use of traditional medicines, including against COVID-19, assuming they must undergo the same serious testing for effectiveness and safety as any other new medicines. Moezzi [90] proposed cynaroside as a promising agent against SARS-nCoV2 by docking the flavonoid on the active site of the main peptidase.

Advertisement

10. Conclusion

The current chapter efforts to coordinate with current studies to identify more accurate biomarkers of the risks for nutrition-related diseases and should lead to dietary recommendations and the formulation of new food products. The food and nutritional supplement industry is very interested in the development and promotion of flavone-rich products as a result of the current evidence for flavones’ protective effects against diseases. An integration of the results of past and future experiments in various disciplines, including biochemistry, cell biology, physiology, pathophysiology, epidemiology, and food chemistry, will be needed to identify the most effective properties of cynaroside and to determine the optimal levels of intake for better health.

Acknowledgments

This study was supported by fund by the EU Erasmus Mundus Partnership (Action 2) CASIA project.

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Sá C, Oliveira AR, Machado C, Azevedo M, Pereira-Wilson C. Effects on liver lipid metabolism of the naturally occurring dietary flavone Luteolin-7-glucoside. Evidence-Based Complementary and Alternative Medicine. 2015;2015:e647832. Available from: https://www.hindawi.com/journals/ecam/2015/647832/
  2. 2. Batra P, Sharma AK. Anti-cancer potential of flavonoids: Recent trends and future perspectives. 3 Biotech. 2013;3(6):439-459
  3. 3. McCullough ML, Peterson JJ, Patel R, Jacques PF, Shah R, Dwyer JT. Flavonoid intake and cardiovascular disease mortality in a prospective cohort of US adults. The American Journal of Clinical Nutrition. 2012;95(2):454-464. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3260072/pdf/ajcn9520454.pdf
  4. 4. Testa R, Bonfigli AR, Genovese S, De Nigris V, Ceriello A. The possible role of flavonoids in the prevention of diabetic complications. Nutrients. 2016;8(5):1-13. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4882722/
  5. 5. Costa SL, Silva VDA, dos Santos SC, Santos CC, Paris I, Muñoz P, et al. Impact of plant-derived flavonoids on neurodegenerative diseases. Neurotoxicity Research. 2016;30(1):41-52
  6. 6. Jana K, Yin X, Schiffer RB, Chen JJ, Pandey A, Stocco DM, et al. Chrysin, a natural flavonoid enhances steroidogenesis and steroidogenic acute regulatory protein gene expression in mouse Leydig cells. Journal of Endocrinology. 2008;197(2):315-323
  7. 7. Kim HP, Son KH, Chang HW, Kang SS. Anti-inflammatory plant flavonoids and cellular action mechanisms. Journal of Pharmacological Sciences. 2004;96(3):229-245
  8. 8. Ja G, Ml L, Castillo J, Benavente-Garcia O, Vicente V, Rivera J. Flavonoids inhibit platelet function through binding to the thromboxane A2 receptor. Journal of Thrombosis and Haemostasis. 2005;3(2):369-376
  9. 9. Horáková Ľ. Flavonoids in prevention of diseases with respect to modulation of Ca-pump function. Interdisciplinary Toxicology. 2011;4(3):45-60
  10. 10. Kumar S, Pandey AK. Chemistry and biological activities of flavonoids: An overview. The Scientific World Journal. 2013;2013(162750):1-16
  11. 11. von Kostanecki S, Tambor J. Ueber die Constitution des Fisetins. Berichte der Deutschen Chemischen Gesellschaft. 1895;28:2302-2309
  12. 12. Cao Z, Ding Y, Ke Z, Cao L, Li N, Ding G, et al. Luteoloside acts as 3C protease inhibitor of enterovirus 71 in vitro. PLoS One. 2016;11(2):e0148693
  13. 13. Lin Y, Chen TY, Tseng HW, Lee MH, Chen ST. Chemical and biological evaluation of nephrocizin in protecting nerve growth factor-differentiated PC12 cells by 6-hydroxydopamine-induced neurotoxicity. Phytochemistry. 2012;84:102-115
  14. 14. Choudhary M, Kumar V, Malhotra H, Singh S. Medicinal plants with potential antiarthritic activity. Journal of Intercultural Ethnopharmacology. 2015;4(2):147-179. DOI: 10.5455/jice.20150313021918
  15. 15. Sahreen S, Khan MR, Khan RA. Comprehensive assessment of phenolics and antiradical potential of Rumex hastatus D. Don. Roots. BMC Complementary and Alternative Medicine. 2014;14:47. DOI: 10.1186/1472-6882-14-47
  16. 16. Ahmed SA, Kamel EM. Cytotoxic activities of flavonoids from Centaurea scoparia. The Scientific World Journal. 2014;2014:274207. DOI: 10.1155/2014/274207
  17. 17. Shojaii A, Abdollahi FM. Review of pharmacological properties and chemical constituents of Pimpinella anisum. ISRN Pharmaceutics. 2012;2012:1-8. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3405664/
  18. 18. Brooks JS, Williams EH, Feeny P. Quantification of contact oviposition stimulants for black swallowtail butterfly, Papilio polyxenes, on the leaf surfaces of wild carrot, Daucus carota. Journal of Chemical Ecology. 1996;22(12):2341-2357
  19. 19. Žemlička L, Fodran P, Lukeš V, Vagánek A, Slováková M, Staško A, et al. Physicochemical and biological properties of luteolin-7-O-β-d-glucoside (cynaroside) isolated from Anthriscus sylvestris (L.) Hoffm. Monatshefte für Chemie—Chemical Monthly. 2014;145(8):1307-1318
  20. 20. Kim JS, Kim JC, Shim SH, Lee EJ, Jin W, Bae K, et al. Chemical constituents of the root of Dystaenia takeshimana and their anti-inflammatory activity. Archives of Pharmacal Research. 2006;29(8):617-623. DOI: 10.1007/BF02968244
  21. 21. Mamatkhanova M, Khalilov R, Syrov V, Mamatkhanov A, Kotenko L, Satimov G, et al. Technology for cinaroside production from the aerial part of Ferula varia and evaluation of its hypoazotemic activity. Pharmaceutical Chemistry Journal. 2009;43(3):160-162
  22. 22. Wang J, Lv D, Liang X, Zhao M, Zhang S. Study on chemical constituents of the Ixeris chinensis. Journal of Chinese Medicinal Material. 2011;34(11):1706-1708
  23. 23. Hu B, Jiang H, Yang J, Zeng S. Determination of luteolin and luteolin-7-beta-D-glucoside in chrysanthemum morfolium Ramat. From different collection time by RP-HPLC. Journal of Zhejiang University Medical Sciences. 2004;31(1):29-32
  24. 24. Wu MJ, Wang L, Ding HY, Weng CY, Yen JH. Glossogyne tenuifolia acts to inhibit inflammatory mediator production in a macrophage cell line by downregulating LPS-induced NF-κB. Journal of Biomedical Science. 2004;11(2):186-199
  25. 25. Williams CA, Hoult JRS, Harborne JB, Greenham J, Eagles J. A biologically active lipophilic flavonol from Tanacetum parthenium. Phytochemistry. 1995;38(1):267-270
  26. 26. Williams CA, Goldstone F, Greenham J. Flavonoids, cinnamic acids and coumarins from the different tissues and medicinal preparations of Taraxacum officinale. Phytochemistry. 1996;42(1):121-127
  27. 27. Küpeli Akkol E, Bahadır Acıkara Ö, Süntar I, Ergene B, Saltan ÇG. Ethnopharmacological evaluation of some Scorzonera species: In vivo anti-inflammatory and antinociceptive effects. Journal of Ethnopharmacology. 2012;140(2):261-270
  28. 28. Wu XL, Li CW, Chen HM, Su ZQ , Zhao XN, Chen JN, et al. Anti-inflammatory effect of supercritical-carbon dioxide fluid extract from flowers and buds of Chrysanthemum indicum Linnén. Evidence-Based Complementary and Alternative Medicine. 2013;2013:1-13
  29. 29. Vitalini S, Beretta G, Iriti M, Orsenigo S, Basilico N, Dall Acqua S, et al. Phenolic compounds from Achillea millefolium L. and their bioactivity. Acta Biochimica Polonica. 2011;58(2):203-209
  30. 30. Shuai S. Flavonoids of Erigeron canadensis. China Journal of Chinese Materia Medica. 2012;37(19):2902-2905
  31. 31. Jeong DM, Jung HA, Choi JS. Comparative antioxidant activity and HPLC profiles of some selected Korean thistles. Archives of Pharmacal Research. 2008;31(1):28-33
  32. 32. Wolniak M, Tomczykowa M, Tomczyk M, Gudej J, Wawer I. Antioxidant activity of extracts and flavonoids from Bidens tripartita. Acta Poloniae Pharmaceutica. 2008;64(5):441-447
  33. 33. Russo D, Malafronte N, Frescura D, Imbrenda G, Faraone I, Milella L, et al. Antioxidant activities and quali-quantitative analysis of different Smallanthus sonchifolius [(Poepp. and Endl.) H. Robinson] landrace extracts. Natural Product Research. 2014;29(17):1673-1677
  34. 34. Iwaoka E, Oku H, Iinuma M, Ishiguro K. Allergy-preventive effects of the flowers of Impatiens textori. Biological & Pharmaceutical Bulletin. 2010;33(4):714-716
  35. 35. Cipollini D, Stevenson R, Enright S, Eyles A, Bonello P. Phenolic metabolites in leaves of the invasive shrub, Lonicera maackii, and their potential phytotoxic and anti-herbivore effects. Journal of Chemical Ecology. 2008;34(2):144-152
  36. 36. Hassan RA, Tawfik WA, Abou-Setta LM. The flavonoid constitunts of Leucaena leucocephala. Growing in Egypt, and their biological activity. African Journal of Traditional Complementary and Alternative Medicine. 2014;11(1)67-72
  37. 37. Kozan E, Anul SA, Tatlı Iİ. In vitro anthelmintic effect of Vicia pannonica var. purpurascens on trichostrongylosis in sheep. Experimental Parasitology. 2013;134(3):299-303
  38. 38. Stabursvik A. Luteolin-7-glucoside as a characteristic component of the plant genera Juncus and Luzula (family Juncaceae). Acta Chemica Scandinavica. 1968;22:2371-2373
  39. 39. Lima CF, Valentao PCR, Andrade PB, Seabra RM, Fernandes-Ferreira M, Pereira-Wilson C. Water and methanolic extracts of Salvia officinalis protect HepG2 cells from t-BHP induced oxidative damage. Chemico-Biological Interactions. 2007;167(2):107-115
  40. 40. Wojciechowski H, Gumbinger HG, Vahlensieck U, Winterhoff H, Nahrstedt A, Kemper FH. Analysis of the components of Lycopus europaeus L. in body fluids during metabolism studies comparison of capillary electrophoresis and high-performance liquid chromatography. Journal of Chromatography A. 1995;717(1-2):261-270
  41. 41. Zhang W. Separation and identification of flavonoid glucosides from Elsholtzia blanda (Benth.) Benth. China Journal of Chinese Materia Medica. 1999;24(2):96-96
  42. 42. Gao Y, Rui-chao L, Wang GL, Zhao H, Gao Y, Ciren B. Studies on the chemical constituents of Phlomis younghusbandii. Journal of Chinese Medicinal Materials. 2007;30(10):1239
  43. 43. Skrzypczak-Pietraszek E, Pietraszek J. Seasonal changes of flavonoid content in Melittis melissophyllum L. (Lamiaceae). Chemistry & Biodiversity. 2014;11(4):562-570
  44. 44. Moharram FA, Al-Gendy AA, El-Shenawy SM, Ibrahim BM, Zarka MA. Phenolic profile, anti-inflammatory, antinociceptive, anti-ulcerogenic and hepatoprotective activities of Pimenta racemosa leaves. BMC Complementary and Alternative Medicine. 2018;18(1):1-15
  45. 45. Wang J, Hou G. Chemical constituents of the flower of Ligustrum lucidum Ait. Journal of Chinese Medicinal Material. 1990;15(3):168-170, 191
  46. 46. Yazıcı-Tütüniş S, Gürel-Gurevin E, Üstünova S, Demirci-Tansel C, Meriçli F. Possible effects of Phillyrea latifolia on weight loss in rats fed a high-energy diet. Pharmaceutical Biology. 2016;54(10):1991-1997
  47. 47. Nagy M, Križková L, Mučaji P, Kontšeková Z, Šeršeň F, Krajčovič J. Antimutagenic activity and radical scavenging activity of water infusions and phenolics from Ligustrum plants leaves. Molecules. 2009;14(1):509-518
  48. 48. Huang Y, Zhang P, He F, Zheng L, Wang Y, Wu J. Simultaneous determination of four bioactive flavonoids from Polygonum orientale L. in dog plasma by UPLC–ESI-MS/MS and application of the technique to pharmacokinetic studies. Journal of Chromatography B. 2014;957:96-104
  49. 49. Wang W, Wang Z, Zhou Y. Studies on chemical constituents from Pteris multifida. Journal of Chinese Medicinal Materials. 2008;31(8):1165-1167
  50. 50. Nabavi S, Habtemariam S, Ahmed T, Sureda A, Daglia M, Sobarzo-Sánchez E, et al. Polyphenolic composition of Crataegus monogyna Jacq.: From chemistry to medical applications. Nutrients. 2015;7(9):7708-7728. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4586556/
  51. 51. Zhu L, Tan J, Wang B, He R, Liu Y, Zheng C. Antioxidant activities of aqueous extract from Agrimonia pilosa Ledeb and its fractions. Chemistry & Biodiversity. 2009;6(10):1716-1726
  52. 52. Shin EJ, Hur HJ, Sung MJ, Park JH, Yang HJ, Kim MS, et al. Ethanol extract of the Prunus mume fruits stimulates glucose uptake by regulating PPAR-γ in C2C12 myotubes and ameliorates glucose intolerance and fat accumulation in mice fed a high-fat diet. Food Chemistry. 2013;141(4):4115-4121
  53. 53. Zhang J, Zhang YN, Han L. Studies on chemical constituents of leaves of Salix matsudana Koidz and their influence on lipolysis. China Journal of Chinese Materia Medica. 2000;25(9):538-541
  54. 54. Klimek B, Olszewska MA, Tokar M. Simultaneous determination of flavonoids and phenylethanoids in the flowers of Verbascum densiflorum and V. phlomoides by high-performance liquid chromatography. Phytochemical Analysis. 2009;21(2):150-156
  55. 55. El Gizawy H, Hussein MA, Abdel-Sattar E. Biological activities, isolated compounds and HPLC profile of Verbascum nubicum. Pharmaceutical Biology. 2019;57(1):485-497
  56. 56. Michaud J, Masquelier J. Essai d’identification du cynaroside. Bulletin-Societe de Pharmacie de Bordeaux. 1958;97(2):77-81
  57. 57. Masquelier J, Michaud J. Note sur les constituants polyphénoliques du Cynara scolymus L. Bulletin-Societe de Pharmacie de Bordeaux. 1955;95(2):65-67
  58. 58. Tien YH, Chen B, Hsu G-SW, Lin W, Huang JH, Lu Y. Hepatoprotective and anti-oxidant activities of Glossogyne tenuifolia against acetaminophen-induced hepatotoxicity in mice. The American Journal of Chinese Medicine. 2014;42(06):1385-1398
  59. 59. Song YS, Park CM. Luteolin and luteolin-7-O-glucoside strengthen antioxidative potential through the modulation of Nrf2/MAPK mediated HO-1 signaling cascade in RAW 264.7 cells. Food and Chemical Toxicology. 2014;65:70-75
  60. 60. Gebhardt R. Inhibition of cholesterol biosynthesis in HepG2 cells by artichoke extracts is reinforced by glucosidase pretreatment. Phytotherapy Research. 2002;16(4):368-372. DOI: 10.1002/ptr.960
  61. 61. Qiusheng Z, Xiling S, Xubo MS, Changhai W. Protective effects of luteolin-7-glucoside against liver injury caused by carbon tetrachloride in rats. Die Pharmazie. 2004;59(4):286-289
  62. 62. Park JC, Park JG, Kim HJ, Hur JM, Lee JH, Sung NJ, et al. Effects of extract from Angelica keiskei and its component, cynaroside, on the hepatic bromobenzene-metabolizing enzyme system in rats. Phytotherapy Research. 2002;16(S1):24-27
  63. 63. Lima CF, Fernandes-Ferreira M, Pereira-Wilson C. Phenolic compounds protect HepG2 cells from oxidative damage: Relevance of glutathione levels. Life Sciences. 2006;79(21):2056-2068
  64. 64. Adzet T, Camarasa J, Laguna JC. Hepatoprotective activity of polyphenolic compounds from Cynara scolymus against CCl4 toxicity in isolated rat hepatocytes. Journal of Natural Products. 1987;50(4):612-617
  65. 65. Hayes PC, Bouchier IA, Beckett GJ. Glutathione S-transferase in humans in health and disease. Gut. 1991;32:813-818
  66. 66. Knapen MF, Peters WH, Mulder TP, Steegers EA. A marker for hepatocellular damage. The Lancet. 2000;355(9213):1463-1464. Available from: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(05)74662-6/fulltext
  67. 67. Maina I, Rule JA, Wians FH, Poirier M, Grant L, Lee WM. α-Glutathione S-transferase: A new biomarker for liver injury? Journal of Applied Laboratory Medicine. 2016;1(2):119-128. DOI: 10.1373/jalm.2016.020412
  68. 68. Ishikawa T, Ali-Osman F. Glutathione-associated cis-diamminedichloroplatinum (II) metabolism and ATP-dependent efflux from leukemia cells. Molecular characterization of glutathione-platinum complex and its biological significance. Journal of Biological Chemistry. 1993;268(27):20116-20125
  69. 69. Wang L, Ma X, Wang J, Li C. In vitro inhibitory effects of Cynaroside on human liver cytochrome P450 enzymes. Pharmacology. 2019;104(5-6):296-302
  70. 70. Sun X, Sun G, Wang M, Xiao J, Sun X. Protective effects of cynaroside against H2O2-induced apoptosis in H9c2 cardiomyoblasts. Journal of Cellular Biochemistry. 2011;112(8):2019-2029
  71. 71. Martín-Cordero C, López-Lazaro M, Piñero J, Ortiz T, Cortés F, Ayuso MJ. Glucosylated isoflavones as DNA topoisomerase II poisons. Journal of Enzyme Inhibition. 2000;15(5):455-460
  72. 72. Kapral-Piotrowska J, Strawa JW, Jakimiuk K, Wiater A, Tomczyk M, Gruszecki WI, et al. Investigation of the membrane localization and interaction of selected flavonoids by NMR and FTIR spectroscopy. International Journal of Molecular Sciences. 2023;24(20):15275. DOI: 10.3390/ijms242015275
  73. 73. Rump AFE, Schüßler M, Açar D, Cordes A, Theisohn M, Rösen R, et al. Functional and antiischemic effects of luteolin-7-glucoside in isolated rabbit hearts. General Pharmacology-the Vascular System. 1994;25(6):1137-1142
  74. 74. Rump AFE, Schüssler M, Acar D, Cordes A, Ratke R, Theisohn M, et al. Effects of different inotropes with antioxidant properties on acute regional myocardial ischemia in isolated rabbit hearts. General Pharmacology: The Vascular System. 1995;26(3):603-611
  75. 75. Palombo R, Savini I, Avigliano L, Madonna S, Cavani A, Albanesi C, et al. Luteolin-7-glucoside inhibits IL-22/STAT3 pathway, reducing proliferation, acanthosis, and inflammation in keratinocytes and in mouse psoriatic model. Cell Death & Disease. 2016;7(8):e2344. Available from: https://pubmed.ncbi.nlm.nih.gov/27537526/
  76. 76. Baskar AA, Ignacimuthu S, Michael GP, Al NK. Cancer chemopreventive potential of Luteolin-7-O-glucoside isolated from Ophiorrhiza mungos Linn. Nutrition and Cancer. 2010;1-9:130-138
  77. 77. Gálvez M, Martín-Cordero C, López-Lázaro M, Cortés F, Ayuso MJ. Cytotoxic effect of Plantago spp. on cancer cell lines. Journal of Ethnopharmacology. 2003;88(2-3):125-130. DOI: 10.1016/s0378-8741(03)00192-2
  78. 78. Hwang YJ, Lee E, Kim HR, Hwang K. Molecular mechanisms of luteolin-7-O-glucoside-induced growth inhibition on human liver cancer cells: G2/M cell cycle arrest and caspase-independent apoptotic signaling pathways. Journal of Biochemistry and Molecular Biology. 2013;46(12):611-616
  79. 79. Mamadalieva NZ, Herrmann F, El-Readi MZ, Tahrani A, Hamoud R, Egamberdieva D, et al. Flavonoids in Scutellaria immaculata and S. ramosissima (Lamiaceae) and their biological activity. Journal of Pharmacy and Pharmacology. 2011;63(10):1346-1357
  80. 80. Hsu HF, Houng JY, Chang CL, Wu CC, Chang FR, Wu YC. Antioxidant activity, cytotoxicity, and DNA information of Glossogyne tenuifolia. Journal of Agricultural and Food Chemistry. 2005;53(15):6117-6125
  81. 81. Zhou M, Shen S, Zhao X, Gong X. Luteoloside induces G0/G1 arrest and pro-death autophagy through the ROS-mediated AKT/mTOR/p70S6K signalling pathway in human non-small cell lung cancer cell lines. Biochemical and Biophysical Research Communications. 2017;494(1-2):263-269
  82. 82. Fan SH, Wang Y, Lu J, Zheng YL, Wu DM, Li MQ , et al. Luteoloside suppresses proliferation and metastasis of hepatocellular carcinoma cells by inhibition of NLRP3 inflammasome. PLoS One. 2014;9(2):e89961-e89961
  83. 83. Shao J, Wang C, Li L, Liang H, Dai J, Ling X, et al. Luteoloside inhibits proliferation and promotes intrinsic and extrinsic pathway-mediated apoptosis involving MAPK and mTOR signaling pathways in human cervical cancer cells. International Journal of Molecular Sciences. 2018;19(6):1664
  84. 84. Emad F, Khalafalah AK, El Sayed MA, Mohamed AEH, Stadler M, Helaly SE. Three new polyacetylene glycosides (PAGs) from the aerial part of Launaea capitata (Asteraceae) with anti-biofilm activity against Staphylococcus aureus. Fitoterapia. 2020;143:104548
  85. 85. Lee SA, Moon S, Han SH, Hwang EJ, Park BR, Kim JS, et al. Chondroprotective effects of aqueous extract of Anthriscus sylvestris leaves on osteoarthritis in vitro and in vivo through MAPKs and NF-κB signaling inhibition. Biomedicine and Pharmacotherapy. 2018;103:1202-1211
  86. 86. Szymański M, Dudek-Makuch M, Witkowska-Banaszczak E, et al. Comparison of the chemical composition and antioxidant activity of essential oils from the leaves and flowers of Sambucus nigra. Pharmaceutical Chemistry Journal. 2020;54:496-503. DOI: 10.1007/s11094-020-02228-5
  87. 87. Grzegorczyk-Karolak I, Wysokińska H, Olas B. Studies on the antioxidant properties of extracts from the roots and shoots of two Scutellaria species in human blood plasma. Acta Biochimica Polonica. 2015;62(2):253-258
  88. 88. Zou Y, Zhang M, Zhang T, Wu J, Wang J, Liu K, et al. Antioxidant and anti-inflammatory activities of Cynaroside from Elsholtiza bodinieri. Natural Product Communications. 2018;13(11):1934578X1801301-1
  89. 89. Lu J, Feng X, Sun Q , Lu H, Manabe M, Sugahara K, et al. Effect of six flavonoid compounds from Ixeris sonchifolia on stimulus-induced superoxide generation and tyrosyl phosphorylation in human neutrophils. Clinica Chimica Acta. 2002;316(1-2):95-99
  90. 90. Moezzi MS. Comprehensive in silico screening of flavonoids against SARS-CoV-2 main protease. Journal of Biomolecular Structure and Dynamics. 2022;1-14:9448-9461

Written By

Sabina Gayibova, Eva Ivanisova and Ulugbek Gayibov

Submitted: 22 January 2024 Reviewed: 26 April 2024 Published: 03 June 2024