Chemical nature of metabolic activation of natural products in traditional Chinese medicines possibly associated with toxicities

Yuyang Liu, Xu Wang, Minglu Liu, Xialing Hao, Ying Peng, Jiang Zheng

PDF(1130 KB)
PDF(1130 KB)
Acupuncture and Herbal Medicine ›› 2024, Vol. 4 ›› Issue (2) : 184-196. DOI: 10.1097/HM9.0000000000000118
Review Articles
research-article

Chemical nature of metabolic activation of natural products in traditional Chinese medicines possibly associated with toxicities

Author information +
History +

Abstract

Ensuring the safety of traditional Chinese medicines (TCM) has perennially presented a universal challenge in the healthcare realm. Meticulous investigations into the toxicological intricacies of natural products are of paramount significance, particularly regarding the metabolic transformation of these substances and the subsequent generation of reactive intermediates. This biochemical process underlies the genesis of diverse toxic manifestations, including hepatotoxicity, nephrotoxicity, pulmonary toxicity, and genotoxicity. Compounds sorted within TCM, including pyrrolizidine alkaloids, anthraquinones, furanoterpenoids, alkenylbenzenes, bisbenzylisoquinoline alkaloids, flavonoids, and methylenedioxyphenyl derivatives, evince a spectrum of deleterious mechanisms upon metabolic activation. This review provides a comprehensive delineation of the pathways through which these compounds induce toxicity via metabolic activation. This review emphasizes the chemical mechanisms involved in the metabolic activation of natural products that may trigger a toxic cascade, rather than a superficial phenomenon. Furthermore, this study enriches the extant literature by delving into advancements in elucidating the mechanisms of toxicity engendered by metabolic activation. In conclusion, this review highlights the importance of scrutinizing the mechanisms of toxicity and provides insights into the judicious and safe use of TCM.

Keywords

Covalent binding / Metabolic activation / Natural products / Toxicological mechanisms / Traditional Chinese medicines

Cite this article

Download citation ▾
Yuyang Liu, Xu Wang, Minglu Liu, Xialing Hao, Ying Peng, Jiang Zheng. Chemical nature of metabolic activation of natural products in traditional Chinese medicines possibly associated with toxicities. Acupuncture and Herbal Medicine, 2024, 4(2): 184‒196 https://doi.org/10.1097/HM9.0000000000000118

References

[[1]]
Cyranoski D. Why Chinese medicine is heading for clinics around the world. Nature. 2018;561(7724):448-450.
[[2]]
The world health organization’s decision about traditional Chinese medicine could backfire. Nature. 2019;570(7759):5.
[[3]]
Cyranoski D. China is promoting coronavirus treatments based on unproven traditional medicines. Nature. 2020. doi:10.1038/d41586-020-01284-x.
[[4]]
Xiao XH, Zhao X, Bai ZF, et al. New outlook on safety of traditional Chinese medicine: concept and practice. China J Chin Mater Med. 2023;48(10):2557.
[[5]]
Zhu H, Wang X, Wang X, et al. The toxicity and safety of Chinese medicine from the bench to the bedside. J Herb Med. 2021;28:100450.
[[6]]
Vanherweghem JL, Tielemans C, Abramowicz D, et al. Rapidly progressive interstitial renal fibrosis in young women: association with slimming regimen including Chinese herbs. Lancet. 1993;341(8842):387-391.
[[7]]
Balachandran P, Wei F, Lin R, et al. Structure activity relationships of aristolochic acid analogues: toxicity in cultured renal epithelial cells. Kidney Int. 2005;67:1797-1805.
[[8]]
Nortier JL, Martinez MC, Schmeiser HH, et al. Urothelial carcinoma associated with the use of a Chinese herb (Aristolochia fangchi). N Engl J Med. 2000;342:168.
[[9]]
Wang Y, Yang JB, Jin HY, Gu DL, Wang Q, Liu Y, Zan K, Fan J, Wang R, Wei F, Ma SC. Comparisons of physicochemical features and hepatoprotective potentials of unprocessed and processed polysaccharides from Polygonum multiflorum Thunb. Int J Biol Macromol. 2023;235:123901.
[[10]]
Xue XY, Quan YY, Gong LH, et al. A review of the processed Polygonum multiflorum (Thunb.) for hepatoprotection: clinial use, pharmacology and toxicology. J Ethnopharmacol. 2020;261:113121.
[[11]]
Wang YK, Li WQ, Xia S, et al. Metabolic activation of the toxic natural products from herbal and dietary supplements leading to toxicities. Front Pharmacol. 2021;12:758468.
[[12]]
Zhou SF, Xue CC, Yu XQ, et al. Metabolic activation of herbal and dietary constituents and its clinical and toxicological implications: an update. Curr Drug Metab. 2007;8(6):526.
[[13]]
Stravitz RT, Lee WM. Acute liver failure. Lancet. 2019;394(10201):869-881.
[[14]]
Jaeschke H, Ramachandran A. Acetaminophen hepatotoxicity: paradigm for understanding mechanisms of drug-induced liver injury. Annu Rev Pathol. 2024;19(1):453-478.
[[15]]
Attia SM. Deleterious effects of reactive metabolites. Oxid Med Cell Longev. 2010;3(4):238-253.
[[16]]
Pessayre D. Role of reactive metabolites in drug-induced hepatitis. J Hepatol. 1995;23:16-24.
[[17]]
Maddukuri VC, Bonkovsky HL. Herbal and dietary supplement hepatotoxicity. Clinical liver disease. 2014;4(1):1-3.
[[18]]
Smith LW, Culvenor CC. Plant sources of hepatotoxic pyrrolizidine alkaloids. J Nat Prod. 1981;44(2):129-152.
[[19]]
Stegelmeier BL, Edgar JA, Colegate SM, et al. Pyrrolizidine alkaloid plants, metabolism and toxicity. J Nat Toxins. 1999;8(1):95-116.
[[20]]
Cheng T, Li WW, Yang XJ, et al. Immunochemistry-based characterization of lysine residue modification by reactive metabolite of retrorsine in mice. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2020;38:315.
[[21]]
Li J, Li XM, Zhou MY, et al. Evaluating and predicting the correlations of hepatic concentration and pyrrole-protein adduction with hepatotoxicity induced by retrorsine based on pharmacokinetic/pharmacodynamic model. Toxicol Lett. 2023;373:152.
[[22]]
Guo Y, Ma ZG, Kou H, et al. Synergistic effects of pyrrolizidine alkaloids and lipopolysaccharide on preterm delivery and intrauterine fetal death in mice. Toxicol Lett. 2013;221(3): 212-218.
[[23]]
Lin G, Tang J, Liu X, et al. Deacetylclivorine: a gender-selective metabolite formed in female SD rat liver microsome. Drug Metab Dispos. 2007;35:607.
[[24]]
Ruan JQ, Gao H, Li N, et al. Blood pyrrole-protein adducts-a biomarker of pyrrolizidine alkaloid-induced liver injury in humans. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2015;33(4):404.
[[25]]
Li X, Yang XJ, Xiang E, et al. Maternal-fetal disposition and metabolism of retrorsine in pregnant rats. Drug Metab Dispos. 2018;46(4):422-428.
[[26]]
Yang XJ, Li WW, Li H, et al. A difference in internal exposure makes newly weaned mice more susceptible to the hepatotoxicity of retrorsine than adult mice. Chem Res Toxicol. 2018;31(12):1348.
[[27]]
Guo Y, Xiao D, Yang XJ, Zheng J, Hu SW, Wu PF, Li XX, Kou H, Wang H. Prenatal exposure to pyrrolizidine alkaloids induced hepatotoxicity and pulmonary injury in fetal rats. Reprod Toxicol. 2019;85:34.
[[28]]
Luo JY, Yang XJ, Qiu SK, et al. Sex difference in monocrotaline-induced developmental toxicity and fetal hepatotoxicity in rats. Toxicology. 2019;418:32-40.
[[29]]
Lin G, Wang JY, Li N, et al. Hepatic sinusoidal obstruction syndrome associated with consumption of Gynura segetum. J Hepatol. 2011;54(4):666.
[[30]]
Chen MX, Li L, Zhong DF, et al. 9-Glutathionyl-6,7-dihydro-1-hydroxymethyl-5H-pyrrolizine is the major pyrrolic glutathione conjugate of retronecine-type pyrrolizidine alkaloids in liver microsomes and in rats. Chem Res Toxicol. 2016;29(2):180-189.
[[31]]
Yang XJ, Li WW, Sun Y, et al. Comparative study of hepatotoxicity of pyrrolizidine alkaloids retrorsine and monocrotaline. Chem Res Toxicol. 2017;30(2):532.
[[32]]
Dai JY, Zhang F, Zheng J. Retrorsine, but not monocrotaline, is a mechanism-based inactivator of P450 3A4. Chem Biol Interact. 2010;183(1):49.
[[33]]
Li WW, Wang K, Lin G, et al. Lysine adduction by reactive metabolite(s) of monocrotaline. Chem Res Toxicol. 2016;29(3):333.
[[34]]
Yang M, Ma J, Ruan J, et al. Intestinal and hepatic biotransformation of pyrrolizidine alkaloid N-oxides to toxic pyrrolizidine alkaloids. Arch Toxicol. 2019;93(8):2197-2209.
[[35]]
Li WW, Cheng T, Jiang TT, et al. Hepatic RNA adduction derived from metabolic activation of retrorsine in vitro and in vivo. Chem Biol Interact. 2022;365:110047.
[[36]]
Matsuda H, Shimoda H, Morikawa T, et al. Phytoestrogens from the roots of Polygonum cuspidatum (Polygonaceae): structure-requirement of hydroxyanthraquinones for estrogenic activity. Bioorg Med Chem Lett. 2001;11(14):1839-1842.
[[37]]
Kuo CH, Sun SW. Analysis of nine rhubarb anthraquinones and bianthrones by micellar electrokinetic chromatography using experimental design. Anal Chim Acta. 2003;482(1):47-58.
[[38]]
Dong X, Fu J, Yin X, Cao S, Li X, Lin L, Huyiligeqi, Ni J. Emodin: a review of its pharmacology, toxicity and pharmacokinetics. Phytother Res. 2016;30(8):1207.
[[39]]
Wang X, Dong L, Zhao G, et al. 2,3,5,4’-Tetrahydroxy stilbene-2-O-β-D-glucoside, a mechanism-based inactivator of CYP2C19 and CYP3A4, potentiates hepatic protein adduction and hepatotoxicity induced by emodin in vivo. Chem Biol Interact. 2022;368:110234.
[[40]]
Lin L, Liu Y, Fu S, et al. Inhibition of mitochondrial complex function—the hepatotoxicity mechanism of emodin based on quantitative proteomic analyses. Cells. 2019;8(3):263.
[[41]]
Qin B, Xu Y, Chen J, et al. Chemical reactivity of emodin and its oxidative metabolites to thiols. Chem Res Toxicol. 2016;29(12):2114-2124.
[[42]]
Wang X, Ding Z, Ma K, et al. Cysteine-based protein covalent binding and hepatotoxicity induced by emodin. Chem Res Toxicol. 2022;35(2):293-302.
[[43]]
Wang M, Zhang Z, Ruan P, et al. Emodin-induced hepatotoxicity is enhanced by 3-methylcholanthrene through activating aryl hydrocarbon receptor and inducing CYP1A1 in vitro and in vivo. Chem Biol Interact. 2022;365:110089.
[[44]]
Wang X, Zhao G, Ju C, et al. Reduction of emodin-8-O-β-D-glucoside content participates in processing-based detoxification of polygoni multiflori radix. Phytomedicine. 2023;114:154750.
[[45]]
Li R, Li W, You Y, et al. Metabolic activation and cytotoxicity of aloe-emodin mediated by sulfotransferases. Chem Res Toxicol. 2019;32(6):1281-1288.
[[46]]
Wang X, Xin X, Sun Y, et al. Chemical reactivity of aloe-emodin and its hydroxylation metabolites to thiols. Chem Res Toxicol. 2019;32(2):234-244.
[[47]]
Qin X, Peng Y, Zheng J. In vitro and in vivo studies of the electrophilicity of physcion and its oxidative metabolites. Chem Res Toxicol. 2018;31:340-349.
[[48]]
Sun Y, Xin X, Zhang K, et al. Cytochrome P450 mediated metabolic activation of chrysophanol. Chem Biol Interact. 2018;289:57-67.
[[49]]
Xu Y, Mao X, Qin B, et al. In vitro and in vivo metabolic activation of rhein and characterization of glutathione conjugates derived from rhein. Chem Biol Interact. 2018;283:1-9.
[[50]]
Bunchorntavakul C, Reddy KR. Herbal and dietary supplement hepatotoxicity. Aliment Pharm Ther. 2013;37(1):3.
[[51]]
Tan R, Hu Z, Zhou M, et al. Diosbulbin B: an important component responsible for hepatotoxicity and protein covalent binding induced by Dioscorea bulbifera. Phytomed. 2022;102:154174.
[[52]]
Zhang Z, Lin D, Li W, et al. Sensitive bromine-based screening of potential toxic furanoids in Dioscorea Bulbifera. J Chromatog B Anal Technol Biomed Life Sci. 2017;1057:1.
[[53]]
Tian M, Peng Y, Zheng J. Metabolic activation and hepatotoxicity of furan-containing compounds. Drug Metab Dispos. 2022;50:655-670.
[[54]]
Lin D, Li C, Peng Y, et al. Cytochrome P450-mediated metabolic activation of diosbulbin B. Drug Metab Dispos. 2014;42(10):1727-1736.
[[55]]
Li W, Lin D, Gao H, et al. Metabolic activation of furan moiety makes Diosbulbin B hepatotoxic. Arch Toxicol. 2016;90:863-872.
[[56]]
Li H, Peng Y, Zheng J. Diosorea bulbifera L.-induced hepatotoxicity and involvement of metabolic activation of furanoterpenoids. Drug Metab Rev. 2020;52:568-584.
[[57]]
Zhang Z, Li H, Li W, et al. Evidence for polyamine, biogenic amine, and amino acid adduction resulting from metabolic activation of diosbulbin B. Chem Res Toxicol. 2020;33:1761-1769.
[[58]]
Wang K, Lin D, Guo X, et al. Chemical identity of interaction of protein with reactive metabolite of diosbulbin B in vitro and in vivo. Toxins. 2017;9:E249.
[[59]]
Lin D, Guo X, Gao H, et al. In vitro and in vivo studies of metabolic activation of 8-epidiosbulbin E acetate. Chem Res Toxicol. 2015;28:1737.
[[60]]
Lin D, Wang K, Gao H, et al. Lysine- and cysteine-based protein adductions derived from toxic metabolites of 8-epidiosbulbin E acetate. Toxicol Lett. 2016;264:20.
[[61]]
Lin D, Li W, Peng Y, et al. Role of metabolic activation in 8-epidiosbulbin E acetate-induced liver injury: mechanism of action of the hepatotoxic furanoid. Chem Res Toxicol. 2016;29(3):359-366.
[[62]]
Zhang N, Yang Y, Li W, et al. Asparagine and glutamine residues participate in protein covalent binding by epoxide metabolite of 8-epidiosbulbin E acetate in vitro and in vivo. Chem Res Toxicol. 2022;35:1821-1830.
[[63]]
Zhang N, Zhou S, Zhang Z, et al. Evidence for adduction of biologic amines with reactive metabolite of 8-epidiosbulbin E acetate in vitro and in vivo. Toxicol Lett. 2022;365:1-10.
[[64]]
Zhang S, Liu Y, Liu T, et al. DNA damage by reactive oxygen species resulting from metabolic activation of 8-epidiosbulbin E acetate in vitro and in vivo. Toxicol Appl Pharmacol. 2022;443:116007.
[[65]]
Lin D, Li W, Tian X, et al. In vitro DNA adduction resulting from metabolic activation of diosbulbin B and 8-epidiosbulbin E acetate. Chem Res Toxicol. 2018;32(1):38-48.
[[66]]
Pan J, Wu C, Tan R, et al. Difference in hepatotoxicity of furan-containing components in Cortex dictamni correlates the efficiency of their metabolic activation. Phytomedicine. 2023;114:154778.
[[67]]
Ran G, Liao Y, Wang X, et al. Mechanistic study of xanthotoxin-mediated inactivation of CYP1A2 and related drug-drug interaction with tacrine. Chem Res Toxicol. 2023;36(3):420-429.
[[68]]
Liu Y, Zhang S, Jiang T, et al. Mechanistic study of bergamottin-induced inactivation of CYP2C9. Food Chem Toxicol. 2021;153:112278.
[[69]]
Hu Z, Zhao Y, Li J, et al. Development of biomarker for Dioscorea bulbifera L. exposure and hepatotoxicity in rats. Phytomed. 2022;102:154172.
[[70]]
Wang K, Zheng L, Peng Y, et al. Selective and sensitive platform for function-based screening of potentially harmful furans. Anal Chem. 2014;86(21):10755-10762.
[[71]]
Li W, Hu Z, Jia C, et al. Mechanism-based cytotoxicity trend prediction of furan-containing pollutants present in a mixture. Environ Pollut. 2024;345:123511.
[[72]]
Li W, Hu Z, Sun C, et al. A metabolic activation-based chemoproteomic platform to profile adducted proteins derived from furan-containing compounds. ACS Chem Biol. 2022;17(4):873-882.
[[73]]
Hu Z, Li W, Lin D, et al. Development of polyclonal antibodies for detection of diosbulbin B derived cis-enedial protein adducts. Chem Res Toxicol. 2018;31:231.
[[74]]
Zhou S, Zhang N, Hu Z, et al. Immunochemical detection of protein modification derived from metabolic activation of 8-epidiosbulbin E acetate. Chem Res Toxicol. 2020;33:1752-1760.
[[75]]
Alhusainy W, Paini A, Punt A, et al. Inhibition of methyleugenol bioactivation by the herb-based constituent nevadensin and prediction of possible in vivo consequences using physiologically based kinetic modeling. Food Chem Toxicol. 2013;59:564.
[[76]]
Rietjens IM, Boersma MG, van der Woude H, et al. Flavonoids and alkenylbenzenes: mechanisms of mutagenic action and carcinogenic risk. Mutat Res. 2005;574(1-2):124-138.
[[77]]
Yang X, Feng Y, Zhang Z, et al. In vitro and in vivo evidence for RNA adduction resulting from metabolic activation of methyleugenol. J Agric Food Chem. 2020;68:15134-15141.
[[78]]
Wang X, Ran G, Liao Y, et al. RNA adduction resulting from metabolic activation of safrole mediated by P450 enzymes and sulfotransferases. Food Chem Toxicol. 2023;174:113688.
[[79]]
Feng Y, Wang S, Peng Y, et al. Urinary methyleugenol-deoxyadenosine adduct as potential biomarker of methyleugenol exposure. J Agricul Food Chem. 2018;66:1258.
[[80]]
Yao H, Peng Y, Zheng J. Identification of glutathione and cysteine conjugates derived from reactive metabolites of methyleugenol in rats. Chem Biol Interact. 2016;253:143-152.
[[81]]
Feng Y, Peng Y, Zheng J. Chemical interactions of cysteine residues with reactive metabolites of methyleugenol. Chem Res Toxicol. 2017;30:564.
[[82]]
Weber C, Opatz T. Bisbenzylisoquinoline alkaloids. Alkaloids Chem Biol. 2019;81:1-114.
[[83]]
Gao GY, Xiao PG. Review of studies of bisbenzylisoquinoline alkaloid (BBI) on distribution in higher plant and physiological activities. Nat Prod Res Dev. 1998;11(3):96.
[[84]]
Tian Y, Qian SN, Jiang Y, et al. The interaction between human breast cancer resistance protein (BCRP) and five bisbenzylisoquinoline alkaloids. Int J Pharm. 2013;453(2):371-379.
[[85]]
Yang LJ, Yang ZD, Li ZJ, et al. Stephtetrandrine A-D, bisbenzylisoquinoline alkaloids from Stephania tetrandra. Nat Prod Res. 2023;37(2):204-215.
[[86]]
Amin G, Farah A, Saba Z, et al. Berberine: pharmacological features in health, disease and aging. Curr Med Chem. 2024;31(10):1214-1234.
[[87]]
Jin H, Dai JY, Chen XY, et al. Pulmonary toxicity and metabolic activation of dauricine in CD-1 mice. J Pharmacol Exp Ther. 2010;332(3):738-746.
[[88]]
Jin H, Li L, Zhong DF, et al. Pulmonary toxicity and metabolic activation of tetrandrine in CD-1 mice. Chem Res Toxicol. 2011;24(12):2142-2152.
[[89]]
Jin H, Shen S, Chen XY, et al. CYP3A-mediated apoptosis of dauricine in cultured human bronchial epithelial cells and in lungs of CD-1 mice. Toxicol Appl Pharmacol. 2012;261(3):248-254.
[[90]]
Wang YY, Zhong DF, Chen XY, et al. Identification of quinone methide metabolites of dauricine in human liver microsomes and in rat bile. Chem Res Toxicol. 2009;22(5):824-834.
[[91]]
Tian Y, Shen S, Jiang Y, et al. CYP3A5 mediates bioactivation and cytotoxicity of tetrandrine. Arch Toxicol. 2016;90(7):1737-1748.
[[92]]
Xie H, Liu YY, Peng Y, et al. Detection of protein adduction derived from dauricine by alkaline permethylation. Anal Bioanal Chem. 2016;408(15):4111-4119.
[[93]]
Shen Q, Zuo MJ, Ma L, et al. Demethylation of neferine in human liver microsomes and formation of quinone methide metabolites mediated by CYP3A4 accentuates its cytotoxicity. Chem Biol Interact. 2014;224:89-99.
[[94]]
Sun Y, Yao T, Li H, et al. In vitro and in vivo metabolic activation of berbamine to quinone methide intermediate. J Biochem Mol Toxicol. 2017;31(4):1.
[[95]]
Seraffni M, Peluso I, Raguzzini A. Flavonoids as anti-inflammatory agents. Proc Nutr Soc. 2010;69(3):273.
[[96]]
Zhou YM, Dong XR, Xu D, et al. Therapeutic potential of traditional Chinese medicine for interstitial lung disease. J Ethnopharmacol. 2024;318(Pt A):116952.
[[97]]
Song L, Zhang W, Tang SY, et al. Natural products in traditional Chinese medicine: molecular mechanisms and therapeutic targets of renal fibrosis and state-of-the-art drug delivery systems. Biomed Pharmacother. 2024;170:116039.
[[98]]
Wei ZC, Chen J, Zuo F, et al. Traditional Chinese Medicine has great potential as candidate drugs for lung cancer: a review. J Ethnopharmacol. 2023;300:115748.
[[99]]
Rauf A, Imran M, Khan IA, et al. Anticancer potential of quercetin: a comprehensive review. Phytother Res. 2018;32(11):2109-2130.
[[100]]
Vrijsen R, Michotte Y, Boeyé A. Metabolic activation of quercetin mutagenicity. Mutat Res. 1990;232(2):243-248.
[[101]]
Poór M, Zrínyi Z, Kőszegi T. Structure related effects of flavonoid aglycones on cell cycle progression of HepG2 cells: Metabolic activation of fisetin and quercetin by catechol-O-methyltransferase (COMT). Biomed Pharmacother. 2016;83:998-1005.
[[102]]
Soares VCG, Varanda EA, Raddi MSG. In vitro basal and metabolism-mediated cytotoxicity of flavonoids. Food Chem Toxicol. 2006;44(6):835-838.
[[103]]
Chen Y, Guo XC, Ma YF, et al. Identification of quinone methide intermediate resulting from metabolic activation of icaritin in vitro and in vivo. Chem Res Toxicol. 2019;32(6):969-973.
[[104]]
Chen Y, Yu J, Wang X, et al. Characterization of glutathione conjugates derived from reactive metabolites of seven silymarin isomers. Xenobiotica. 2019;49(11):1269-1278.
[[105]]
Yoo HH, Lee SH, Jin C, et al. Mechanism-based inactivation of cytochrome P450 3A4 by methylenedioxyphenyl lignans from Acanthopanax chiisanensis. Planta Med. 2008;74(8):822-827.
[[106]]
Tu DZ, Mao X, Zhang F, et al. Reversible and irreversible inhibition of cytochrome p450 enzymes by methylophiopogonanone A. Drug Metab Dispos. 2020;49(6):459-469.
[[107]]
Cui TT, Wang Q, Tian XX, et al. Piperine is a mechanism-based inactivator of CYP3A. Drug Metab Dispos. 2020;48(2):123-134.
[[108]]
Nakajima M, Suzuki M, Yamaji R, et al. Isoform selective inhibition and inactivation of human cytochrome P450s by methylenedioxyphenyl compounds. Xenobiotica. 1999;29(12):1191-1202.
[[109]]
Mirzaei MS, Ivanov MV, Taherpour AA, et al. Mechanism-based inactivation of cytochrome P450 enzymes: computational insights. Chem Res Toxicol. 2021;34(4):959-987.
[[110]]
Mao X, Zhao HM, Wang Q, et al. Metabolism-based herb-drug interactions of Corydalis bugeana Herba with berberine in vitro and in vivo in rats. Biomed Chromatogr. 2019;33:e4514.
[[111]]
Mao X, Peng Y, Zheng J. In vitro and in vivo characterization of reactive intermediates of corynoline. Drug Metab Dispos. 2015;43(10):1491-1498.
[[112]]
Yang AH, Zhang L, Zhi DX, et al. Identification and analysis of the reactive metabolites related to the hepatotoxicity of safrole. Xenobiotica. 2018;48(11):1164-1172.
[[113]]
Hu LL, Wu F, He J, et al. Cytotoxicity of safrole in HepaRG cells: studies on the role of CYP1A2-mediated ortho-quinone metabolic activation. Xenobiotica. 2019;49(12):1504-1515.
[[114]]
Mao X, Hu ZX, Wang Q, et al. Nitidine chloride is a mechanism-based inactivator of CYP2D6. Drug Metab Dispos. 2018;46(8):1137-1145.
[[115]]
Liu YY, Cui TT, Peng Y, et al. Mechanism-based inactivation of cytochrome P450 2D6 by chelidonine. J Biochem Mol Toxicol. 2019;33(2):e22251.
[[116]]
Liu YY, Peng Y, Zhang ZY, et al. In vitro and in vivo studies of the metabolic activation of chelidonine. Chem Biol Interact. 2019;308:155.
[[117]]
Wang K, Rao JQ, Zhang TT, et al. Metabolic activation and covalent protein binding of berberrubine: insight into the underlying mechanism related to its hepatotoxicity. Drug Des Devel Ther. 2020;14:4423-4438.
[[118]]
Chi MN, Peng Y, Zheng J. Characterization of glutathione conjugates derived from reactive metabolites of bakuchiol. Chem Biol Interact. 2016;244:178.
[[119]]
Guo XC, Lin DJ, Li WW, et al. Electrophilicities and protein covalent binding of demethylation metabolites of colchicine. Chem Res Toxicol. 2016;29(3):296.
[[120]]
LoPachin RM, Geohagen BC, Nordstroem LU. Mechanisms of soft and hard electrophile toxicities. Toxicology. 2019;418:62-69.
[[121]]
Liu Z, He X, Wang LL, Zhang YH, Hai Y, Gao R. Chinese herbal medicine hepatotoxicity: the evaluation and recognization based on large-scale evidence database. Curr Drug Metab. 2019;20(2):138.
[[122]]
Gurley BJ, McGill MR, Koturbash I. Hepatotoxicity due to herbal dietary supplements: past, present and the future. Food Chem Toxicol. 2022;169:113445.
[[123]]
Liang ZJ, Lai YF, Li M, et al. Applying regulatory science in traditional Chinese medicines for improving public safety and facilitating innovation in China: a scoping review and regulatory implications. Chin Med. 2021;16(1):23.
[[124]]
Xiao XH, Zhao FB, Wang JB, et al. Traditional Chinese medicine (TCM) safety evaluation and pharmacovigilance. Chin Sci Bull. 2021;4:407.

RIGHTS & PERMISSIONS

2024 Acupuncture and Herbal Medicine
PDF(1130 KB)

Accesses

Citations

Detail

Sections
Recommended

/