5,818
Views
23
CrossRef citations to date
0
Altmetric
Review

Medicinal plants with anti-mutagenic potential

, , , , , , , , & show all
Pages 309-318 | Received 09 Oct 2019, Accepted 05 Mar 2020, Published online: 10 Apr 2020

Abstract

This review presents an overview of published studies for a better understanding of the anti-mutagenic potential of medicinal plants and the precise indications for the utilization of natural compounds as chemo-preventive agents. Reports on the anti-mutagenic potential of medicinal plants published from 1997 to 2019 were searched through different scientific databases using the following keywords: medicinal plants and mutagens, carcinogens, the anti-mutagenic potential of medicinal plants. The data relevant to the anti-mutagenic potential of some common medicinal plants is summarized in this mini-review. These medicinal plants include Carum carvi, Withania somnifera, Panax ginseng, Mentha spicata, Curcuma zedoaria, Cassia angustifolia, Cymbopogon citrates, Ipomoea batatas, Glycyrrhiza glabra, Citrullus colocynthis, Capsicum annuum and Asparagus racemosus. An overview of the identified molecules or enzymes being targeted is also presented, with a focus on anti-carcinogenic and/or anti-mutagenic activity. The recent advancements in the research on medicinal plants pave the way for the better understanding and future prospects of the use of natural components as chemo-preventive and chemotherapeutic agents.

Introduction

Indigenous medicinal plants for the treatment of different diseases are receiving increasing attention. European countries, as well as Arabian countries, are taking a revived interest in herbal medicines due to their efficacy, safety and cost-effectiveness [Citation1]. Plants have been used for alleviating the sufferings from various kinds of conditions [Citation2, Citation3]. The interest in the study of such medicinal plants has developed with the aim to obtain therapeutically effective drugs [Citation4]. Many clinically documented herbal medicines have appeared in the official and unofficial compendia [Citation5, Citation6]. Many modern medicines have been developed because of research on plant extracts, including atropine, reserpine, ergot and digitalis, which are continuously used in modern medicines. Traditional western medicines have not been able to completely satisfy the population all over the world, so most of the population is returning to complementary and alternative medicines, which explains the greater acceptability of herbal medicines [Citation7, Citation8]. Medicinal plants have been used for the treatment of various diseases and their activities have been reported. Anti-mutagenic activities of extracts of various medicinal plants have been evaluated by using the microsomal system/Salmonella to investigate whether they contain direct or indirect anti-mutagens. Many plant extracts show anti-mutagenic activity, including Pteris multifida, Smilax china and Prunella vulgaris [Citation9–11]. For example, Sukumaran et al. [Citation12] reported the inhibition of tobacco-induced mutagenesis by eugenol and plant extracts. The anti-mutagenicity of turmeric oil has been reported [Citation13]. For many years it has been a continuous struggle to find natural substances having the ability to inhibit or reverse multi-stage carcinogenesis [Citation14]. A wide range of constituents present in medicinal plants or dietary herbs has been reported for their potential to inhibit carcinogenicity [Citation15]. Various dietary compounds or herbs have demonstrated inhibitory activities on the progression of cancer, and most often, their chemo-preventive effects are linked to anti-inflammatory or anti-oxidant activities that some of their components possess [Citation16–18]. Frequent consumption of vegetables, spices and fruit is linked to lower risks of different cancers as reported by epidemiological studies [Citation19, Citation20]. This protective activity is often attributed to the steroids and flavonoids that are found in medicinal plants and are a fundamental ingredient of human nutrition [Citation21, Citation22]. This review highlights some common medicinal plants possessing anti-mutagenic activities.

Methodology

For this literature review, published articles were searched through different databases, including PubMed, Medline, Scopus, Science Direct, Google Scholar and Web of Science. The literature search was performed using the following keywords: medicinal plants and mutagens, carcinogens, the anti-mutagenic potential of medicinal plants; and the relevant articles published in 1997–2019 were downloaded. The reported data on the anti-mutagenic role of studied medicinal plants were summarized to provide a brief overview of medicinal plants with potential therapeutic role against mutagens/carcinogens.

Molecular mechanisms underlying the anti-mutagenicity of natural products

The following mechanistic pathways of anti-mutagenicity have been proposed and reviewed [Citation23]:

  1. Inactivation of mutagens directly.

  2. Inhibition of chemical or metabolic activation of mutagens.

  3. DNA protection against the action of mutagens.

  4. Modification in DNA repair and DNA replication.

  5. Hepatic detoxification system modulation.

For example, Teel [Citation24] and Ahmed et al. [Citation25] described a mechanism of anti-mutagenesis by ellagic acid via making adducts with DNA and thus masking the binding sites to be occupied by the carcinogens or mutagens.

Plants having anti-mutagenic activity

The results from the performed literature search showed an array of plant extracts with anti-mutagenic properties (). In this mini-review, we discuss some of the medicinal plants with reported anti-mutagenic potential. These plants include Carum carvi, Withania somnifera, Panax ginseng, Mentha spicata, Curcuma zedoaria, Cassia angustifolia, Cymbopogon citrates, Ipomoea batatas, Glycyrrhiza glabra, Citrullus colocynthis, Capsicum annuum and Asparagus racemosus. Further, we summarized the molecules or enzymes being targeted and consequently results in anti-carcinogenic and/or anti-mutagenic activity. The plants that have potential against carcinogenesis and mutagenesis are also highlighted.

Table 1. Other medicinal plants possessing anti-mutagenic activity.

Carum carvi L

Carum carvi (caraway) belongs to family Apiaceae. Its seeds are widely used for medicinal purposes and they contain fatty acids and volatile oils [Citation47, Citation48]. The seeds of caraway are commonly used in cookies and loaves of bread as a seasoning [Citation49]. The data reported in earlier studies suggested that the use of spices reduced the risk of gastric cancer [Citation50, Citation51]. It was found that hot water extracts of C. carvi are anti-mutagenic against ICR-170, nitroso-dimethylamine and MNNG (N-methyl-N’-nitro-N-nitrosoguanidine) [Citation52]. D-Carvone, the main constituent of caraway oil, exhibited the potential to inhibit the development of stomach and pulmonary tumours induced by diethylnitrosamine [Citation53–57]. Mazaki et al. [Citation58] reported the inhibitory activities of C. carvi and its component on MNNG-induced mutagenicity and it was suggested that O6 methylguanine DNA methyltransferase may be responsible for the anti-mutagenic effect of Carum carvi. Using sequential extraction of caraway seeds with methanol, n-hexane and boiling water, it was reported that hot water extracts have more enhanced anti-mutagenic potential [Citation59].

Withania somnifera

Withania somnifera (family Solanaceae) is a well-known medicinal plant widely found in India and the Mediterranean region [Citation60, Citation61]. The parts used are the roots, bark, seeds, flowers, stem and leaves. It contains withaferin and is used as an inhibitor of human osteosarcoma, breast, colon, central nervous system (CNS), lung, bladder cell lines, and cancer [Citation62]. Withaferin-A is a purified steroidal lactone from the leaves of W. somnifera [Citation7] and has much bioactive potential, including anti-inflammatory, pro-apoptotic [Citation63] and anti-proliferative effects [Citation64]. Another study [Citation65] reported that combined treatment of withaferin-A with hyperthermia-induced significant apoptosis in HeLa cells by increasing the oxidative stress and reducing the GSH/GSSG ratio, JNK activation and mitochondria-caspase dependent apoptotic pathway involvement. Withaferin-A has been reported as a potential anticancer compound, as it inhibits tumour growth, metastasis and angiogenesis. More recently, it was evaluated that withaferin-A exhibited synergistic effects of chemotherapeutic agents like doxorubicin and cisplatin and induced cell death by reactive oxygen species generation leading to DNA damage resulting in apoptosis [Citation66, Citation67].

Khanam and Devi [Citation68] investigated methanolic extracts of W. somnifera roots for protective effect on mitomycin induced damage to the bone marrow and the anti-oxidant enzymes in the liver of mice. The protective effect was assessed using mouse bone marrow micronucleus test against micronuclei (MN) formation in normochromatic cells (NCEs) and polychromatic cells (PCEs) induced by 4 mg/kg b.w., i.p Mitomycin C. The study findings suggested that W. somnifera extract is useful for the prevention of DNA damage and one of the action mechanisms might involve scavenging the active oxygen radicals induced by the mutagens [Citation68].

Panax ginseng

Panax ginseng belongs to family Araliaceae; its roots are utilized as a general stimulant in traditional Oriental medicine for enhancing longevity, health and vitality, especially in the elderly [Citation69]. Its chemical constituents are theophylline, saponins, ginsenosides, caffeine, xanthines and theobromine [Citation70]. It has tonic, adaptogenic and anti-cancer activity [Citation71–73]. Rhee et al. [Citation74] reported the anti-mutagenic effect and transformation by P. ginseng root extracts. Raghavendran et al. [Citation75] observed that P. ginseng amends cytokines in bone marrow toxicity and myelopoiesis: ginsenoside Rg1 moderately supports myelopoiesis. White ginseng can reduce the mitotic division of cells, and prevent the metastasis of melanoma (B16) [Citation76]. It is reported to enhance the endogenous anti-oxidant system, which suppresses the production of harmful free radicals [Citation77]. Interestingly, the Korean red ginseng is chemically processed in such a way that its chemical composition is transformed and its biological properties are altered according to the need [Citation78]. Ginseng alkaloids and saponins prevent the metastasis and growth of lung cancer by preventing the encroachment and bonding of tumour cells with each other and with the neighbour cells [Citation79, Citation80]. Red ginseng is capable of preventing the metastasis of a tumour by restricting the activities of tumour cells and increasing their lysosomal vulnerability [Citation81, Citation82]. The extract of P. ginseng, when given orally to mice, showed a significant reduction in the number of tumour cells, shrinkage of tumour cells, and also increased the time duration of the appearance of tumour cells [Citation83]. Alpha-pyrrolidone, an alkaloid of ginseng, was tested for the first time in 1960 and was shown to have inhibitory effects on tumour cell lines [Citation84–89].

Mentha spicata

Mentha spicata belongs to the Lamiaceaefamily. The parts used are its bark and fruit. It contains nodifloretin, chrysoberyl, protocatechuic acid and protocatechuic aldehyde [Citation90, Citation91]. It has antimicrobial [Citation92] and anti-oxidant potential [Citation93]. Yu et al. [Citation94] reported the anti-mutagenic activity of M. spicata and evaluated its considerable inhibitory effects against mutagens. Some early studies reported its anti-mutagenic activities [Citation90, Citation95–99]. The volatile ingredients of spearmint like limonene, terpenoids, menthone, carvone, dihydrocarvinone, menthol and other compounds belonging to the same class have therapeutic and cancer chemo-preventive activities [Citation100]. Aqueous extract of M. spicata acted as an inhibiting agent or anti-mutagen via inhibitory activities on DNA adduct formation or carcinogen metabolism in vitro [Citation101].

Curcuma zedoaria

It belongs to family Zingiberaceae. Its common name is Narkachur/White turmeric and is found in India and Indonesia. It contains curcumenol, procurcumenol, zederone and comosone II [Citation102, Citation103]. The parts used are the leaves. It has anti-oxidant [Citation103] and anti-cancer activity [Citation104]. Curcumin has been reported as an anti-mutagenic agent [Citation105]. In another study, two extracts of C. zedoaria (aqueous and methanol) exhibited no mutagenicity when investigated in several Salmonella typhimurium strains with or without the S9 mix [Citation106]. Furthermore, the methanolic extract exhibited better anti-mutagenic activity than the aqueous extract against 2-amino-3-methylimidazo (4,5-f) quinoline and 4-nitroquinoline-N-oxide. The inhibitory activities of the two extracts on lipid peroxidation were comparable [Citation107]. Extracts of C. zedoaria had a moderate anti-mutagenic effect against mutation induced by benzo [a] pyrene [Citation9–11].

Cassia angustifolia

It belongs to family Poaceae. Its common name is senna and is found in Sri Lanka. It contains flavonoids, apigenin, phenolic compounds, luteolin, kaempferol and quercetin [Citation108]. The parts used are its leaves [Citation109]. It has anti-mutagenic activity [Citation110]. Senna is widely used as a laxative [Citation111], however, potential side effects like genotoxicity have also been reported [Citation112]. Its water extract showed anti-mutagenic activity by four experimental assays. Furthermore, the aqueous extract was not mutagenic or cytotoxic to Escherichia coli strains [Citation113].

Cymbopogon citratus

It belongs to family Leguminosae. Its common name is lemongrass and is found in India. It contains tinnevellin glycoside, D-3-O-methyl inositol and kaempferol [Citation114]. The parts used are its leaves. It has anti-cancer activity [Citation115]. The alcoholic extract of Lemongrass is reportedly anti-mutagenic [Citation116, Citation117].

Ipomoea batatas L

Ipomoea batatas L. (common name sweet potato) belongs to family Convolvulaceae. It is found in America and contains phenolic acid, tannins, saponins, coumarin, anthraquinone and triterpenes [Citation118, Citation119]. The parts commonly used are the roots. Yoshimoto et al. [Citation120] demonstrated that I. batatas possess anti-mutagenic effect due to its mono-, di-, and tri-caffeoylquinic acid derivatives. The aqueous extracts of I. batatas had anti-mutagenic activities against Trp-P-1 in Salmonella typhimurium TA 98. Among four varieties of different root flesh colours, the Ayamurasaki variety (purple colored flesh) significantly decreased the mutation rate induced by tryptophan pyrolysis products and by the grilled meat of beef (dimethyl sulfoxide extract), and it was hypothesized that like-wise the heterocyclic amines, the pigment in anthocyanin reduced the mutation activities [Citation121]. Two pigments of anthocyanin obtained from the purple-coloured variety of I. batatas effectively inhibited the potential of tryptophan pyrolysis products to induce mutations in rat liver homogenate [Citation122].

Glycyrrhiza glabra L

It belongs to family Leguminosae. Its common name is liquorice and is found in Southern Europe and different parts of Asia, including India. It contains glycyrrhizic acid, glycyrrhizin and Licor-phenone [Citation123]. The parts used are its rhizome [Citation124]. It has anti-mutagenic activity. Certain phytoconstituents can be employed as dietary supplements, functional foods and even as drugs due to their chemo-preventive effects [Citation125]. The natural compounds may serve as potential chemo-preventive agents due to their anti-genotoxic and free-radical scavenging activity [Citation126]. Moreover, they act to prevent the modulatory functions of some mutagenic agents in the cytoplasmic environment of Salmonella typhimurium and play a significant role to restrict the microbial infection [Citation127]. Extracts of G. glabra were fractionated and isolated to show that its biochemical compounds are effective against the mutagenicity caused by oxidative stress [Citation128, Citation129]. Methanolic extracts of G. glabra are effective in preventing the mutations caused by oxidative mutagens; especially glycyrrhizic acid was more effective in reducing the genotoxicity of the oxidative mutagens [Citation130]. Glycyrrhizic acid (120 μmol/L) shows high potential to reduce free radicals in the DPPH radical scavenging assay, suggesting that it potentially modulates the activities of genotoxins via free-radical scavenging [Citation130]. In addition, Karahan et al. [Citation131] confirmed the anti-oxidant potential of the methanolic extracts of G. glabra var. glandulifera roots and reported the antimicrobial activities that were less potent against gram-negative than against gram-positive ones. Their study results indicated that the biological activities and the contents of chemical compounds in the natural liquorice populations might be affected under different environmental conditions in each habitat [Citation131].

Citrullus colocynthis

It belongs to family Cucurbitaceae. Its common name is bitter apple [Citation132] and is found in Africa and India. It contains isoorientin 3′-Ο-methyl ether, isovitexin and iso-saponarin [Citation14]. The parts used are mainly their seeds. It has anti-cancer activity [Citation133]. In vitro analysis of the methanolic extracts of the seeds of C. colocynthis was carried out in the bacterial system to observe its effects on mutagenesis. The methanolic extract of seeds of C. colocynthis was not effective in inducing resistance to serve as a mutagen for the antibiotic treatments (streptomycin & rifampicin). Therefore, these findings suggested that plant extract had no genotoxic effects on the cell’s system [Citation134, Citation135]. C. colocynthis fruit extract mitigated the genotoxic potential of cyclophosphamide in mice bone marrow cells [Citation134, Citation135].

Capsicum annuum

It belongs to the family Solanaceae. Its common name is pepper and is found in the Americas and South Asia. Although it is not native to Europe, it is grown there too. It contains capsaicin [Citation136]. The parts used are fruits. It has anti-mutagenic activity [Citation137]. Capsaicin (CPS) is the principal constituent of peppers [Citation138] and shows antitumor activity by targeting several molecular pathways [Citation139].

Asparagus racemosus

Asparagus racemosus (family Liliaceae) is an important medicinal plant shown to increase cellular vitality and resistance and promote the general wellbeing. It is widely distributed in India, the Southern part of China, Sri Lanka, Australia, Java and tropical Africa. The pharmacological activities of this plant include its use as a brain tonic, antiepileptic, antihypertensive agent, a regulator of cardiac disorders, in the treatment of spermatogenic irregularities, oligospermia and male genital dysfunction [Citation140–142]. Methanolic and aqueous-methanolic extracts of A. racemosus roots have shown anti-mutagenic effect against sodium azide and 4-nitro-ophenylenediamine (NPD) induced mutations through the Ames mutagenicity assay using Salmonella typhimurium strains TA98 and TA100 [Citation143]. Hence, A. racemosus extracts have been indicated as a new source of anti-mutagenic agents [Citation143].

Conclusions

This mini-review briefly documents some reports on the anti-mutagenic potential of different medicinal plants and alternative components in prokaryotic and eukaryotic model systems. The large body of evidence demonstrating the anti-mutagenic activities of different plant extracts has raised hopes that they may also have chemo-preventive and therapeutic potential in different cancer models. Proofs from both in vitro and in vivo studies advocate that they can definitely restrain multiple molecular targets that play a significant role in both chronic inflammation and cancer, but in some cases, there may be a difference in efficacy due to the dosage, the origin of the extract or the cell line used. However, in future, more targeted investigations are needed for better understanding of the molecular mechanisms of action against different mutagens. Further, clinical studies, as well as more research efforts targeting appropriate drug delivery systems are necessary to exploit completely their reported efficacy for the prevention and management of various cancers. Finally, clinical studies in humans could considerably improve our understanding of the macroscopic activities of the active compounds in these common medicinal plants.

Author contributions

MA, MR, RZ and MD conceived the review, drafted the manuscript, analyzed the literature and critically reviewed the manuscript. AWC, AH and MM performed the literature search and took part in analyzing the literature. SAZ, MAS and FSK involved in literature search and critically read the manuscript. All the authors approved the final draft for publication.

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • Gurib-Fakim A. Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol Aspects Med. 2006;27(1):1–93.
  • Zamora-Martínez MC, de Pascual Pola CN. Medicinal plants used in some rural populations of Oaxaca, Puebla and Veracruz, Mexico. J Ethnopharmacol. 1992;35(3):229–257.
  • Dar RA, Shahnawaz M, Qazi PH. General overview of medicinal plants: a review. J Phytopharmacol. 2017;6(6):349–351.
  • Taylor J, Rabe T, McGaw LJ, et al. Towards the scientific validation of traditional medicinal plants. Plant Growth Regul. 2001;34(1):23–37.
  • White SD. Deciphering integrated Chinese and ‘Western medicine’ in the rural Lijiang basin: state policy and local practice (s) in socialist China. Social Sci Med (1982). 1999;49(10):1333–1347.
  • Reid A-M, Oosthuizen CB, Fibrich BD, et al. Traditional medicine: the ancient roots of modern practice. Chap 1. In: Lall M, editor. Medicinal plants for holistic health and well-being. London (UK): Elsevier; 2018. p. 1–11.
  • Raskin I, Ribnicky DM, Komarnytsky S, et al. Plants and human health in the twenty-first century. TRENDS Biotechnol. 2002;20(12):522–531.
  • Veiga M, Costa EM, Silva S, et al. Impact of plant extracts upon human health: a review. Crit Rev Food Sci Nutr. 2020;60(5):873–814.
  • Lee H, Lin J-Y. Antimutagenic activity of extracts from anticancer drugs in Chinese medicine. Mutat Res/Genet Toxicol. 1988;204(2):229–234.
  • Guo W, Tai H-Y, Wang N, et al. Chinese medicines for cancer treatment from the metabolomics perspective. In: Metabolomics-new insights into biology and medicine. London, UK: IntechOpen; 2019. p. 1–27. Available from: https://cdn.intechopen.com/pdfs/68857.pdf.
  • Liu MM, Huang KM, Qian L, et al. Effects of bioactive constituents in the Traditional Chinese medicinal formula Si–Wu–Tang on Nrf2 signaling and neoplastic cellular transformation. Phytomedicine. 2018;40:1–9.
  • Sukumaran K, Kuttan R. Inhibition of tobacco-induced mutagenesis by eugenol and plant extracts. Mutat Res/Genet Toxicol. 1995;343(1):25–30.
  • Jayaprakasha GK, Jena BS, Negi PS, et al. Evaluation of antioxidant activities and antimutagenicity of turmeric oil: a byproduct from curcumin production. Zeitsch Naturforsch C. 2002;57(9–10):828–835.
  • Benariba N, Djaziri R, Bellakhdar W, et al. Phytochemical screening and free radical scavenging activity of Citrullus colocynthis seeds extracts. Asian Pacific J Tropic Biomed. 2013;3(1):35–40.
  • Huang W-Y, Cai Y-Z, Zhang Y. Natural phenolic compounds from medicinal herbs and dietary plants: potential use for cancer prevention. Nutrit Cancer. 2009;62(1):1–20.
  • Kapinova A, Kubatka P, Golubnitschaja O, et al. Dietary phytochemicals in breast cancer research: anticancer effects and potential utility for effective chemoprevention. Environ Health Prev Med. 2018;23(1):1–18.
  • Surh Y-J. Molecular mechanisms of chemopreventive effects of selected dietary and medicinal phenolic substances. Mutat Res/Fundament Mol Mech Mutagen. 1999;428(1–2):305–327.
  • Surh Y-J, Lee J-Y, Choi K-J, et al. Effects of selected ginsenosides on phorbol ester-induced expression of cyclooxygenase-2 and activation of NF-κB and ERK1/2 in mouse skin. Ann NY Acad Sci. 2002;973(1):396–401.
  • Block G, Patterson B, Subar A. Fruit, vegetables, and cancer prevention: a review of the epidemiological evidence. Nutrit Cancer. 1992;18(1):1–29.
  • Jochems SHJ, Van Osch FHM, Bryan RT, et al. Impact of dietary patterns and the main food groups on mortality and recurrence in cancer survivors: a systematic review of current epidemiological literature. BMJ Open. 2018;8(2):e014530.
  • Tsyrlov IB, Mikhailenko VM, Gelboin HV. Isozyme-and species-specific susceptibility of cDNA-expressed CYP1A P-450s to different flavonoids. Biochim Biophys Acta (BBA) Protein Struct Mol Enzymol. 1994;1205(2):325–335.
  • Quradha MM, Khan R, Rehman M-u, et al. Chemical composition and in vitro anticancer, antimicrobial and antioxidant activities of essential oil and methanol extract from Rumex nervosus. Nat Prod Res. 2019;33(17):2554–2559.
  • Słoczyńska K, Powroźnik B, Pękala E, et al. Antimutagenic compounds and their possible mechanisms of action. J Appl Genet. 2014;55(2):273–285.
  • Teel RW. Ellagic acid binding to DNA as a possible mechanism for its antimutagenic and anticarcinogenic action. Cancer Lett. 1986;30(3):329–336.
  • Ahmed T, N. Setzer W, Fazel Nabavi S, et al. Insights into effects of ellagic acid on the nervous system: a mini review. CPD. 2016;22(10):1350–1360.
  • Miyazawa M, Sakano K, Nakamura S-i, et al. Antimutagenic activity of isoflavone from Pueraria lobata. J Agric Food Chem. 2001;49(1):336–341.
  • Miyazawa M, Hisama M. Antimutagenic activity of phenylpropanoids from clove (Syzygium aromaticum). J Agric Food Chem. 2003;51(22):6413–6422.
  • Ga̧Siorowski K, Szyba K, Brokos B, et al. Antimutagenic activity of anthocyanins isolated from Aronia melanocarpa fruits. Cancer Letters. 1997;119(1):37–46.
  • Lee K-T, Sohn I-C, Park H-J, et al. Essential moiety for antimutagenic and cytotoxic activity of hederagenin monodesmosides and bisdesmosides isolated from the stem bark of Kalopanax pictus. Planta Med. 2000;66(04):329–332.
  • Abdelwahed A, Bouhlel I, Skandrani I, et al. Study of antimutagenic and antioxidant activities of Gallic acid and 1, 2, 3, 4, 6-pentagalloylglucose from Pistacia lentiscus: confirmation by microarray expression profiling. Chem Biol Interact. 2007;165(1):1–13.
  • Saraç N, Şen B. Antioxidant, mutagenic, antimutagenic activities, and phenolic compounds of Liquidambar orientalis Mill. var. orientalis. Ind Crops Prod. 2014;53:60–64.
  • Snijman PW, Swanevelder S, Joubert E, et al. The antimutagenic activity of the major flavonoids of rooibos (Aspalathus linearis): some dose-response effects on mutagen activation-flavonoid interactions. Mutat Res/Genet Toxicol Environ Mutagen. 2007;631(2):111–123.
  • Elias R, Méo MD, Vidal-Ollivier E, et al. Antimutagenic activity of some saponins isolated from Calendula officinalis L., C. arvensis L. and Hedera helix L. Mutagenesis. 1990;5(4):327–332.
  • Verma PK, Raina R, Agarwal S, et al. Phytochemical ingredients and pharmacological potential of Calendula officinalis Linn. PBR. 2018;4(2):1–17.
  • Zahin M, Ahmad I, Aqil F. Antioxidant and antimutagenic activity of Carum copticum fruit extracts. Toxicol in Vitro. 2010;24(4):1243–1249.
  • Martínez CJ, Loarca-Piña G, Ortíz GD. Antimutagenic activity of phenolic compounds, oligosaccharides and quinolizidinic alkaloids from Lupinus campestris seeds. Food Addit Contam. 2003;20(10):940–948.
  • Lim J-C, Park JH, Budesinsky M, et al. Antimutagenic constituents from the thorns of Gleditsia sinensis. Chem Pharm Bull. 2005;53(5):561–564.
  • Wożniak D, E, Lamer‐Zarawska A. Matkowski Antimutagenic and antiradical properties of flavones from the roots of Scutellaria baicalensis Georgi. Nahrung. 2004;48(1):9–12.
  • Križková L, Chovanová Z, Ďuračková Z, et al. Antimutagenic i90n vitro activity of plant polyphenols: Pycnogenol® and Ginkgo biloba extract (EGb 761). Phytother Res. 2008;22(3):384–388.
  • Wozniak D, Janda B, Kapusta I, et al. Antimutagenic and anti-oxidant activities of isoflavonoids from Belamcanda chinensis (L.) DC. Mutat Res/Genet Toxicol Environ Mutagen. 2010;696(2):148–153.
  • Dauer A, Metzner P, Schimmer O. Proanthocyanidins from the bark of Hamamelis virginiana exhibit antimutagenic properties against nitroaromatic compounds. Planta Med. 1998;64(04):324–327.
  • Choi J, Lee H, Park K-Y, et al. In vitro antimutagenic effects of anthraquinone aglycones and naphthopyrone glycosides from Cassia tora. Planta Med. 1997;63(01):11–14.
  • Vuković-Gačić B, Nikčević S, Berić-Bjedov T, et al. Antimutagenic effect of essential oil of sage (Salvia officinalis L.) and its monoterpenes against UV-induced mutations in Escherichia coli and Saccharomyces cerevisiae. Food Chem Toxicol. 2006;44(10):1730–1738.
  • Hayder N, Ben Ammar R, Abdelwahed A, et al. Antibacterial and antimutagenic activitiy of extracts and essential oil from (Tunisian) Pistacia lentiscus. Toxicol Environ Chem. 2005;87(4):567–573.
  • Fragoso V, Nascimento N. C d, Moura DJ, et al. Antioxidant and antimutagenic properties of the monoterpene indole alkaloid psychollatine and the crude foliar extract of Psychotria umbellata Vell. Toxicol in Vitro. 2008;22(3):559–566.
  • Gowri S, Chinnaswamy P. Evaluation of in vitro antimutagenic activity of Caralluma adscendens roxb. in bacterial reverse mutation assay. J Nat Prod Plant Resour. 2011;1(4):27–34.
  • Seidler-Lozykowska K, Baranska M, Baranski R, et al. Raman analysis of caraway (Carum carvi L.) single fruits. Evaluation of essential oil content and its composition. J Agric Food Chem. 2010;58(9):5271–5275.
  • Goyal M, Gupta VK, Singh N. Carum Carvi: an updated review. Ind J Pharmaceut Biol Res. 2018;6(04):14–24.
  • Yu LL, Zhou KK, Parry J. Antioxidant properties of cold-pressed black caraway, carrot, cranberry, and hemp seed oils. Food Chem. 2005;91(4):723–729.
  • Buiatti E, Palli D, Decarli A, et al. A case-control study of gastric cancer and diet in Italy. Int J Cancer. 1989;44(4):611–616.
  • Morrison MEW, Joseph JM, McCann SE, et al. Cruciferous vegetable consumption and stomach cancer: a case-control study. Nutrit Cancer. 2020;72(1):52–10.
  • Higashimoto M, Purintrapiban J, Kataoka K, et al. Mutagenicity and antimutagenicity of extracts of three spices and a medicinal plant in Thailand. Mutat Res Lett. 1993;303(3):135–142.
  • Abbas MM, Kandil YI, Abbas MA. R-(-)-carvone attenuated doxorubicin induced cardiotoxicity in vivo and potentiated its anticancer toxicity in vitro. Balkan Med J. 2019;37(2):98–103.
  • Wattenberg LW. Inhibition of carcinogenesis by minor dietary constituents. Cancer Res. 1992;52(7 Suppl):2085s–22091.
  • Wattenberg LW, Sparnins VL, Barany G. Inhibition of N-nitrosodiethylamine carcinogenesis in mice by naturally occurring organosulfur compounds and monoterpenes. Cancer Res. 1989;49(10):2689–2692.
  • Islam MT. Literature and scientifically carvone is an anticancer agent. Pharmacol Online. 2019;2:24–30.
  • Sharma DR, Kumar S, Kumar V, et al. Comprehensive review on nutraceutical significance of phytochemicals as functional food ingredients for human health management. J Pharmacogn Phytochem. 2019;8(5):385–395.
  • Mazaki M, Kataoka K, Kinouchi T, et al. Inhibitory effects of caraway (Carum carvi L.) and its component on N-methyl-N’-nitro-N-nitrosoguanidine-induced mutagenicity. J Med Invest. 2006;53(1,2):123–133.
  • Naderi-Kalali B, Allameh A, Rasaee MJ, et al. Suppressive effects of caraway (Carum carvi) extracts on 2, 3, 7, 8-tetrachloro-dibenzo-p-dioxin-dependent gene expression of cytochrome P450 1A1 in the rat H4IIE cells. Toxicol in Vitro. 2005;19(3):373–377.
  • Singh N, Bhalla M, De Jager P, et al. An overview on ashwagandha: a rasayana (rejuvenator) of ayurveda. Afr J Trad Compl Alt Med. 2011;8(5 Suppl):208–213.
  • Achar GPK, et al. Scientific validation of the usefulness of Withania somnifera Dunal in the prevention and treatment of cancer. In: Aktar M, Swamy M, editor. Anticancer plants: properties and application. Singapore: Springer;2018. p. 285–301.
  • Rai M, Jogee PS, Agarkar G, et al. Anticancer activities of Withania somnifera: Current research, formulations, and future perspectives. Pharm Biol. 2016;54(2):189–197.
  • Mayola E, Gallerne C, Esposti DD, et al. Withaferin-A induces apoptosis in human melanoma cells through generation of reactive oxygen species and down-regulation of Bcl-2. Apoptosis. 2011;16(10):1014–1027.
  • Berghe WV, et al. Molecular insight in the multifunctional activities of Withaferin-A. Biochem Pharmacol. 2012;84(10):1282–1291.
  • Cui Z-G, Piao J-L, Rehman MUR, et al. Molecular mechanisms of hyperthermia-induced apoptosis enhanced by withaferin-A. Eur J Pharmacol. 2014;723:99–107.
  • Devi PU. Withania somnifera Dunal (Ashwagandha): potential plant source of a promising drug for cancer chemotherapy and radiosensitization. Ind J Experiment Biol. 1996;34(10):927–932.
  • Fong MY, Jin S, Rane M, et al. Withaferin-A synergizes the therapeutic effect of doxorubicin through ROS-mediated autophagy in ovarian cancer. PloS One. 2012;7(7):e42265. doi: 10.1371/journal.pone.0042265.
  • Khanam S, Devi K. Antimutagenic activity of ashwagandha. J Natural Remed. 2005;5(2):126–131.
  • Choi H-J, et al. Antioxidantive, phospholipase A2 inhibiting, and anticancer effect of polyphenol rich fractions from Panax ginseng CA Meyer. Appl Biol Chem. 2003;46(3):251–256.
  • Shin B-K, Kwon SW, Park JH. Chemical diversity of ginseng saponins from Panax ginseng. J Ginseng Res. 2015;39(4):287–298.
  • Clark AM. Natural products as a resource for new drugs. Pharm Res. 1996;13(8):1133–1141.
  • Kim J-H. Pharmacological and medical applications of Panax ginseng and ginsenosides: a review for use in cardiovascular diseases. J Ginseng Res. 2018;42(3):264–269.
  • Li J, Yu H, Wang S, et al. Natural products, an important resource for discovery of multitarget drugs and functional food for regulation of hepatic glucose metabolism. DDDT. 2018;12:121–135.
  • Rhee Y, Ahn J, Choe J, et al. Inhibition of mutagenesis and transformation by root extracts of Panax ginseng in vitro. Planta Med. 1991;57(02):125–128.
  • Raghavendran HRB, Sathyanath R, Shin J, et al. Panax ginseng modulates cytokines in bone marrow toxicity and myelopoiesis: ginsenoside Rg1 partially supports myelopoiesis. PloS One. 2012;7(4):e33733.
  • Wang ZY, Agarwal R, Zhou ZC, et al. Inhibition of mutagenicity in Salmonella typhimurium and skin tumor initiating and tumor promoting activities in SENCAR mice by glycyrrhetinic acid: comparison of 18α-and 18β-stereoisomers. Carcinogenesis. 1991;12(2):187–192.
  • Abdel-Wahhab MA, Ahmed HH. Protective effect of Korean Panax ginseng against chromium VI toxicity and free radicals generation in rats. J Ginseng Res. 2004;28(1):11–17.
  • Metwaly AM, Lianlian Z, Luqi H, et al. Black ginseng and its saponins: preparation, phytochemistry and pharmacological effects. Molecules. 2019;24(10):1856.
  • Mochizuki M, Yoo Y, Matsuzawa K, et al. Inhibitory effect of tumor metastasis in mice by saponins, ginsenoside-Rb2, 20 (R)-and 20 (S)-ginsenoside-Rg3, of red ginseng. Biol Pharm Bull. 1995;18(9):1197–1202.
  • Yang H, Oh K-H, Kim HJ, et al. Ginsenoside-Rb2 and 20 (S)-ginsenoside-Rg3 from Korean red ginseng prevent rotavirus infection in newborn mice. J Microbiol Biotechnol. 2018;28(3):391–396.
  • Kubo M, Chun-Ning T, Matsuda H. Influence of the 70% methanolic extract from red ginseng on the lysosome of tumor cells and on the cytocidal effect of mitomycin c1. Planta Med. 1992;58(5):424–428.
  • Luo S, Yang M, Lv D, et al. TMEPAI increases lysosome stability and promotes autophagy. Int J Biochem Cell Biol. 2016;76:98–106.
  • Panwar M, Kumar M, Samarth R, et al. Evaluation of chemopreventive action and antimutagenic effect of the standardized Panax ginseng extract, EFLA400, in Swiss albino mice. Phytother Res. 2005;19(1):65–71.
  • Yun T-K. Panax ginseng: a non-organ-specific cancer preventive? Lancet Oncol. 2001;2(1):49–55.
  • Yun T-K. and Epidemiological evidence of the cancer-preventive effects of Panax ginseng C. a. Meyer. Nutrition Reviews. 2009;54(11):S71–S81.
  • Kim SI, Park JH, Ryu J-H, et al. Ginsenoside Rg 5, a genuine dammarane glycoside from Korean red ginseng. Arch Pharm Res. 1996;19(6):551–553.
  • Kumar A. Immunomodulatory response induced by ginseng. J Ginseng Res. 2003;27(3):115–119.
  • Wakabayashi C, et al. In vivo antimetastatic action of ginseng protopanaxadiol saponins is based on their intestinal bacterial metabolites after oral administration. Oncol Res Featur Preclin Clin Cancer Therap. 1997;9(8):411–417.
  • Kim EH, Kim W. An insight into ginsenoside metabolite compound K as a potential tool for skin disorder. Evid-Based Complement Alternat Med. 2018;2018:1–8..
  • Anwar F, Abbas A, Mehmood T, et al. Mentha: a genus rich in vital nutra-pharmaceuticals: a review. Phytother Res. 2019;33(10):2548–2570.
  • Buleandra M, Oprea E, Popa DE, et al. Comparative chemical analysis of Mentha piperita and M. spicata and a fast assessment of commercial peppermint teas. Nat Prod Commun. 2016;11(4):1934578X1601100.
  • Aggarwal KK, Khanuja SPS, Ahmad A, et al. Antimicrobial activity profiles of the two enantiomers of limonene and carvone isolated from the oils of Mentha spicata and Anethum sowa. Flavour Fragr J. 2002;17(1):59–63.
  • Kanatt SR, Chander R, Sharma A. Antioxidant potential of mint (Mentha spicata L.) in radiation-processed lamb meat. Food Chem. 2007;100(2):451–458.
  • Yu TW, Xu M, Dashwood RH. Antimutagenic activity of spearmint. Environ Mol Mutagen. 2004;44(5):387–393.
  • Morita K, Hara M, Kada T. Studies on natural desmutagens: screening for vegetable and fruit factors active in inactivation of mutagenic pyrolysis products from amino acids. Agric Biol Chem. 1978;42(6):1235–1238.
  • Natake M, Kanazawa K, Mizuno M, et al. Herb water-extracts markedly suppress the mutagenicity of Trp-P-2. Agric Biol Chem. 1989;53(5):1423–1425.
  • Samman MA. Mint prevents shamma-induced carcinogenesis in hamster cheek pouch. Carcinogenesis. 1998;19(10):1795–1801.
  • Kanazawa K. Research and development for bioavailable functional foods in food science. FSTR. 2018;24(2):183–191.
  • Eren Y. Mutagenic and cytotoxic activities of Limonium globuliferum methanol extracts. Cytotechnology. 2016;68(5):2115–2124.
  • Satomi Y, Miyamoto S, Gould MN. Induction of AP-1 activity by perillyl alcohol in breast cancer cells. Carcinogenesis. 1999;20(10):1957–1961.
  • Dashwood RH. Modulation of heterocyclic amine-induced mutagenicity and carcinogenicity: an ‘A-to-Z’guide to chemopreventive agents, promoters, and transgenic models. Mutat Res/Rev Mutat Res. 2002;511(2):89–112.
  • Ayati Z, Ramezani M, Amiri MS, et al. Ethnobotany, phytochemistry and traditional uses of Curcuma spp. and pharmacological profile of two important species (C. longa and C. zedoaria): a review. CPD. 2019;25(8):871–935.
  • de Fátima Navarro D, de Souza MM, Neto RA, et al. Phytochemical analysis and analgesic properties of Curcuma zedoaria grown in Brazil. Phytomedicine. 2002;9(5):427–432.
  • Kim KI, et al. Antitumor, genotoxicity and anticlastogenic activities of polysaccharide from Curcuma zedoaria. Mol Cells. 2000;10(4):392–398.
  • van den Berghe PVE, Stapelbroek JM, Krieger E, et al. Reduced expression of ATP7B affected by Wilson disease–causing mutations is rescued by pharmacological folding chaperones 4‐phenylbutyrate and curcumin. Hepatology. 2009;50(6):1783–1795.
  • Lim TK. Edible medicinal and non-medicinal plants. Vol. 1. Cham: Springer; 2012.
  • Peng C-H, Chiu W-T, Juan C-W, et al. Pivotal role of curcuminoids on the antimutagenic activity of Curcuma zedoaria extracts. Drug Chem Toxicol. 2010;33(1):64–76.
  • Al-Marzoqi AH, Hadi MY, Hameed IH. Determination of metabolites products by Cassia angustifolia and evaluate antimicrobial activity. J Pharmacogn Phytother. 2016;8(2):25–48.
  • Müller B, Kraus J, Franz G. Chemical structure and biological activity of water-soluble polysaccharides from Cassia angustifolia leaves. Planta Med. 1989;55(6):536–539.
  • Zhao Y, Zhao K, Jiang K, et al. A review of flavonoids from cassia species and their biological activity. CPB. 2016;17(13):1134–1146.
  • Raimondi F, et al. Reactive nitrogen species modulate the effects of rhein, an active component of senna laxatives, on human epithelium in vitro. J Pediatr Gastroenterol Nutrit. 2002;34(5):529–534.
  • Mukhopadhyay MJ, Saha A, Dutta A, et al. Genotoxicity of sennosides on the bone marrow cells of mice. Food Chem Toxicol. 1998;36(11):937–940.
  • Silva CR, Monteiro MR, Rocha HM, et al. Assessment of antimutagenic and genotoxic potential of senna (Cassia angustifolia Vahl.) aqueous extract using in vitro assays. Toxicol in Vitro. 2008;22(1):212–218.
  • Asaolu M, Oyeyemi O, Olanlokun J. Chemical compositions, phytochemical constituents and in vitro biological activity of various extracts of Cymbopogon citratus. Pakistan J Nutrit. 2009;8(12):1920–1922.
  • Thangam R, Sathuvan M, Poongodi A, et al. Activation of intrinsic apoptotic signaling pathway in cancer cells by Cymbopogon citratus polysaccharide fractions. Carbohydr Polym. 2014;107:138–150.
  • Manvitha K, Bidya B. Review on pharmacological activity of Cymbopogon citratus. Int J Herb Med. 2014;6:7.
  • Vinitketkumnuen U, et al. Antimutagenicity of lemon grass (Cymbopogon citratus Stapf) to various known mutagens in salmonella mutation assay. Mutat Res/Genet Toxicol. 1994;341(1):71–75.
  • Odake K, Terahara N, Saito N, et al. Chemical structures of two anthocyanins from purple sweet potato, Ipomoea batatas. Phytochemistry. 1992;31(6):2127–2130.
  • Gras CC, Nemetz N, Carle R, et al. Anthocyanins from purple sweet potato (Ipomoea batatas (L.) Lam.) and their color modulation by the addition of phenolic acids and food-grade phenolic plant extracts. Food Chem. 2017;235:265–274.
  • Yoshimoto M, Yahara S, Okuno S, et al. Antimutagenicity of mono-, di-, and tri-caffeoylquinic acid derivatives isolated from sweetpotato (Ipomoea batatas L.) leaf. Biosci Biotechnol Biochem. 2002;66(11):2336–2341.
  • Saigusa N, Terahara N, Ohba R. Evaluation of DPPH-radical-scavenging activity and antimutagenicity and analysis of anthocyanins in an alcoholic fermented beverage produced from cooked or raw purple-fleshed sweet potato (Ipomoea batatas cv. Ayamurasaki) roots. FSTR. 2005;11(4):390–394.
  • Yoshimoto M, Okuno S, Yoshinaga M, et al. Antimutagenicity of sweetpotato (Ipomoea batatas) roots. Biosci Biotechnol Biochem. 1999;63(3):537–541.
  • Varsha S, Agrawal R, Sonam P. Phytochemical screening and determination of anti-bacterial and anti-oxidant potential of Glycyrrhiza glabra root extracts. J Environ Res Develop. 2013;7(4A):1552.
  • Dhingra D, Parle M, Kulkarni S. Memory enhancing activity of Glycyrrhiza glabra in mice. J Ethnopharmacol. 2004;91(2–3):361–365.
  • Jo E, Kim S, Ra J, et al. Chemopreventive properties of the ethanol extract of Chinese licorice (Glycyrrhiza uralensis) root: induction of apoptosis and G1 cell cycle arrest in MCF-7 human breast cancer cells. Cancer Lett. 2005;230(2):239–247.
  • Khattak KF, Simpson TJ. Effect of gamma irradiation on the antimicrobial and free radical scavenging activities of Glycyrrhiza glabra root. Radiat Phys Chem. 2010;79(4):507–512.
  • Zani F, Cuzzoni M, Daglia M, et al. Inhibition of mutagenicity in Salmonella typhimurium by glycyrrhiza glabra extract, glycyrrhizinic acid and 18α-and 18β-glycyrrhetinic acids. Planta Med. 1993;59(06):502–507.
  • Mitscher LA, Telikepalli H, McGhee E, et al. Natural antimutagenic agents. Mutat Res/Fundament Mol Mech Mutagen. 1996;350(1):143–152.
  • Khan MS, Ahmad I. Diversity of antimutagenic phytocompounds from Indian medicinal plants. In: Herbal medicine in India. Singapore: Springer; 2020. p. 401–412.
  • Kaur S, Kaur P, Sharma N, et al. Modulation of genotoxicity of oxidative mutagens by glycyrrhizic acid from Glycyrrhiza glabra L. Phcogn Res. 2012;4(4):189.
  • Karahan F, Avsar C, Ozyigit II, et al. Antimicrobial and antioxidant activities of medicinal plant Glycyrrhiza glabra var. glandulifera from different habitats. Biotechnol Biotechnol Equip. 2016;30(4):797–804.
  • Heydari M, Homayouni K, Hashempur MH, et al. Topical Citrullus colocynthis (bitter apple) extract oil in painful diabetic neuropathy: a double-blind randomized placebo-controlled clinical trial [外用西瓜属药西瓜瓤 (苦苹果) 提取油治疗痛性糖尿病神经病变: 项双盲随机安慰剂对照的临床试验]. J Diabetes. 2016;8(2):246–252.
  • Tannin-Spitz T, Grossman S, Dovrat S, et al. Growth inhibitory activity of cucurbitacin glucosides isolated from Citrullus colocynthis on human breast cancer cells. Biochem Pharmacol. 2007;73(1):56–67.
  • Abbas AH, Obaid HH, Sagban LH, et al. Mutagenic and anti-mutagenic effect of alcoholic extract of Citrullius colocynthis seeds using bacterial system (G-system). J Kerbala Univ. 2012;10(4):229–236.
  • Shokrzadeh M, Chabra A, Naghshvar F, et al. The mitigating effect of Citrullus colocynthis (L.) fruit extract against genotoxicity induced by cyclophosphamide in mice bone marrow cells. Scientific World J. 2013;2013:1–8.
  • Conforti F, Statti GA, Menichini F. and biological variability of hot pepper fruits (Capsicum annuum var. acuminatum L.) in relation to maturity stage. Food Chem. 2007;102(4):1096–1104.
  • Sim K-H, Han Y-S. The antimutagenic and antioxidant effects of red pepper seed and red pepper pericarp (Capsicum annuum L.). JFN. 2007;12(4):273–278.
  • Mori A, Lehmann S, O’Kelly J, et al. Capsaicin, a component of red peppers, inhibits the growth of androgen-independent, p53 mutant prostate cancer cells. Cancer Res. 2006;66(6):3222–3229.
  • Thoennissen NH, O’Kelly J, Lu D, et al. Capsaicin causes cell-cycle arrest and apoptosis in ER-positive and-negative breast cancer cells by modulating the EGFR/HER-2 pathway. Oncogene. 2010;29(2):285–296.
  • Lynch M, Ackerman MS, Gout J-F, et al. Genetic drift, selection and the evolution of the mutation rate. Nat Rev Genet. 2016;17(11):704–714.
  • Gomase V, Sherkhane A. Isolation, structure elucidation and biotransformation studies on secondary metabolites from Asparagus racemosus. Int J Micr Res. 2010;2(1):7–9.
  • Dartsch PC. The potential of asparagus‐P® to inactivate reactive oxygen radicals. Phytother Res. 2008;22(2):217–222.
  • Singla R, et al. In-vitro antimutagenic activity of Asparagus racemosus: an ayurvedic medicinal plant. Am J Drug Discovery Dev. 2013;3(4):1–7.