Research progress on the hepatoprotective effect, pharmacokinetic properties, and hepatotoxicity of geniposide

Songyuan Tang , Guangli Yan , Ling Kong , Hui Sun , Chang Liu , Ying Han , Xijun Wang

Acupuncture and Herbal Medicine ›› 2025, Vol. 5 ›› Issue (2) : 136 -146.

PDF (685KB)
Acupuncture and Herbal Medicine ›› 2025, Vol. 5 ›› Issue (2) : 136 -146. DOI: 10.1097/HM9.0000000000000160
Review Articles
research-article

Research progress on the hepatoprotective effect, pharmacokinetic properties, and hepatotoxicity of geniposide

Author information +
History +
PDF (685KB)

Abstract

Liver disease (LD) is a global health problem caused by multiple factors. At present, there are still obvious problems with limited efficacy and strong side effects of drugs used in the clinical treatment of LD. Therefore, it is of great significance to search for effective hepatoprotective drugs from natural products. Geniposide (GS) is a cyclic ether terpenoid compound and a key component in the traditional Chinese medicine Gardenia jasminoides. It has a significant inhibitory effect on LD. However, there is currently no literature systematically analyzing its mechanism of action. To adapt to the environment of new drug research and the need for precision medication, this article summarizes the pathways and possible mechanisms of action discovered by GS in the treatment of LD, based on recent research literature: regulating bile stasis, antioxidant and anti-apoptosis, improving amino acid metabolism, improving energy metabolism, regulating lipid metabolism, anti-inflammatory and analgesic effects, etc. It also summarizes the pharmacokinetics of GS in vivo and discusses the liver toxicity of GS that is positively correlated with dosage. In addition, the existing problems in current research and possible future development directions were also discussed, to lay the foundation for the clinical development of natural product GS.

Keywords

Geniposide / Hepatotoxicity / Liver disease / Molecular mechanism / Pharmacokinetics

Cite this article

Download citation ▾
Songyuan Tang, Guangli Yan, Ling Kong, Hui Sun, Chang Liu, Ying Han, Xijun Wang. Research progress on the hepatoprotective effect, pharmacokinetic properties, and hepatotoxicity of geniposide. Acupuncture and Herbal Medicine, 2025, 5(2): 136-146 DOI:10.1097/HM9.0000000000000160

登录浏览全文

4963

注册一个新账户 忘记密码

Conflict of interest statement

Xijun Wang is an editorial board member of this journal.The other authors declare no conflict of interest.

Funding

This study was supported by the National Natural Science Foundation of China Key Project (81830110).

Author contributions

Songyuan Tang analyzed the data and wrote the manuscript. Guangli Yan, Ling Kong, Hui Sun, Chang Liu, Ying Han, and Xijun Wang revised the manuscript. All the authors read and approved the final manuscript.

Ethical approval of studies and informed consent

Not applicable.

Acknowledgments

None.

Data availability

All data generated or analyzed during this study are included in this published article.

References

[1]

Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med 2009; 360(26):2758-2769.

[2]

Avila MA, Dufour JF, Gerbes AL, et al. Recent advances in alcohol-related liver disease (ALD): summary of a Gut round table meeting. Gut 2019; 69(4):764-780.

[3]

Andrade RJ, Chalasani N, Björnsson ES, et al. Drug-induced liver injury. Nat Rev Dis Primers 2019; 5(1):58.

[4]

Björnsson HK, Bjöörnsson ES. Drug-induced liver injury: pathogenesis, epidemiology, clinical features, and practical management. Eur J Intern Med 2022;97:26-31.

[5]

Zeng DY, Li JM, Lin S, et al. Global burden of acute viral hepatitis and its association with socioeconomic development status, 1990-2019. J Hepatol 2021; 75(3):547-556.

[6]

Dubuisson J, Cosset FL. Virology and cell biology of the hepatitis C virus life cycle—an update. J Hepatol 2014; 61(1):S3-S13.

[7]

Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol 2015; 62(1):S47-S64.

[8]

Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016; 64(1):73-84.

[9]

Kawaguchi T, Tsutsumi T, Nakano D, et al. MAFLD: Renovation of clinical practice and disease awareness of fatty liver. Hepatol Res 2021; 52(5):422-432.

[10]

Muratori L, Lohse AW, Lenzi M. Diagnosis and management of autoimmune hepatitis. BMJ 2023;380:e070201.

[11]

Sharma R, Verna EC, Simon TG, et al. Cancer risk in patients with autoimmune hepatitis: a nationwide population-based cohort study with histopathology. Am J Epidemiol 2021; 191(2):298-319.

[12]

Devarbhavi H, Asrani SK, Arab JP, et al. Global burden of liver disease: 2023 update. J Hepatol 2023; 79(2):516-537.

[13]

Torbenson M, Washington K. Pathology of liver disease: advances in the last 50 years. Hum Pathol 2020;95:78-98.

[14]

Służały P, Paśko P, Galanty A. Natural products as hepatoprotective agents—a comprehensive review of clinical trials. Plants 2024; 13(14):1985.

[15]

GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the global burden of disease study 2017. Lancet 2018; 392(10159):1789-1858.

[16]

Oaklander AL, Lunn MPT, Hughes RC, et al. Treatments for chronic inflammatory demyelinating polyradiculoneuropathy (CIDP): an overview of systematic reviews. Cochrane Database Syst Rev 2017; 2017(1):CD010369.

[17]

Chen LP, Li MX, Yang ZQ, et al. Gardenia jasminoides Ellis: Ethnopharmacology, phytochemistry, and pharmacological and industrial applications of an important traditional Chinese medicine. J Ethnopharmacol 2020;257:112829.

[18]

Zhang AH, Sun H, Qiu S, et al. Advancing drug discovery and development from active constituents of yinchenhao tang, a famous traditional Chinese medicine formula. Evid Based Complement Alternat Med 2013;2013:1-6.

[19]

Ogasawara T, Morine Y, Ikemoto T, et al. Beneficial effects of Kampo medicine Inchin-ko-to on liver function and regeneration after hepatectomy in rats. Hepatol Res 2008; 38(8):818-824.

[20]

Li HS, Xi YF, Xin X, et al. Geniposide plus chlorogenic acid reverses non-alcoholic steatohepatitis via regulation of gut microbiota and bile acid signaling in a mouse model in vivo. Front Pharmacol 2023;14:1148737.

[21]

Wang XJ, Lv HT, Sun H, et al. Metabolic urinary profiling of alcohol hepatotoxicity and intervention effects of Yin Chen Hao Tang in rats using ultra-performance liquid chromatography/electrospray ionization quadruple time-of-flight mass spectrometry. J Pharm Biomed Anal 2008; 48(4):1161-1168.

[22]

Rinella ME, Neuschwander Tetri BA, Siddiqui MS, et al. AASLD practice guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023; 77(5):1797-1835.

[23]

Xian YX, Weng JP, Xu F. MAFLD vs. NAFLD: Shared features and potential changes in epidemiology, pathophysiology, diagnosis, and pharmacotherapy. Chin Med J (Engl) 2020; 134(1):8-19.

[24]

Fuchs CD, Trauner M. Role of bile acids and their receptors in gastrointestinal and hepatic pathophysiology. Nat Rev Gastroenterol Hepatol 2022; 19(7):432-450.

[25]

Huang WF, Kong DS. The intestinal microbiota as a therapeutic target in the treatment of NAFLD and ALD. Biomed Pharmacother 2021;135:111235.

[26]

Kitade H, Chen G, Ni Y, et al. Nonalcoholic fatty liver disease and insulin resistance: new insights and potential new treatments. Nutrients 2017; 9(4):387.

[27]

Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: an update. Metabolism 2016; 65(8):1109-1123.

[28]

Zhang TL, Zhang AH, Qiu S, et al. High-throughput metabolomics approach reveals new mechanistic insights for drug response of phenotypes of geniposide towards alcohol-induced liver injury by using liquid chromatography coupled to high resolution mass spectrometry. Mol Biosyst 2017; 13(1):73-82.

[29]

Qiu S, Zhang AH, Guan Y, et al. Functional metabolomics using UPLC-Q/TOF-MS combined with ingenuity pathway analysis as a promising strategy for evaluating the efficacy and discovering amino acid metabolism as a potential therapeutic mechanism-related target for geniposide against alcoholic liver disease. RSC Adv 2020; 10(5):2677-2690.

[30]

Liu LP, Wu Q, Chen YP, et al. Updated pharmacological effects, molecular mechanisms, and therapeutic potential of natural product geniposide. Molecules 2022; 27(10):3319.

[31]

Zhou YX, Zhang RQ, Rahman K, et al. Diverse pharmacological activities and potential medicinal benefits of geniposide. Evid Based Complement Alternat Med 2019;2019:4925682.

[32]

Song N, Xu H, Liu J, et al. Design of a highly potent GLP-1R and GCGR dual-agonist for recovering hepatic fibrosis. Acta Pharm Sin B 2022; 12(5):2443-2461.

[33]

Yin F, Liu JH, Zheng XX, et al. Geniposide induces the expression of heme Oxygenase-1 via PI3K/Nrf2-signaling to enhance the antioxidant capacity in primary hippocampal neurons. Biol Pharm Bull 2010; 33(11):1841-1846.

[34]

Wang JJ, Chen L, Liang ZB, et al. Genipin inhibits LPS-induced inflammatory response in BV2 microglial cells. Neurochem Res 2017; 42(10):2769-2776.

[35]

Tu YY, Li LL, Zhu LL, et al. Geniposide attenuates hyperglycemia-induced oxidative stress and inflammation by activating the Nrf2 signaling pathway in experimental diabetic retinopathy. Oxid Med Cell Longev 2021; 2021(1):9247947.

[36]

Seo MJ, Hong JM, Kim SJ, et al. Genipin protects d-galactosamine and lipopolysaccharide-induced hepatic injury through suppression of the necroptosis-mediated inflammasome signaling. Eur J Pharmacol 2017;812:128-137.

[37]

Liu JH, Yin F, Xiao H, et al. Glucagon-like peptide 1 receptor plays an essential role in geniposide attenuating lipotoxicity-induced β-cell apoptosis. Toxicol In Vitro 2012; 26(7):1093-1097.

[38]

Zhang ZH, Wang XJ, Zhang D, et al. Geniposide-mediated protection against amyloid deposition and behavioral impairment correlates with downregulation of mTOR signaling and enhanced autophagy in a mouse model of Alzheimer’s disease. Aging (Milano) 2019; 11(2):536-548.

[39]

Yamamoto M, Miura N, Ohtake N, et al. Genipin, a metabolite derived from the herbal medicine Inchin-ko-to, and suppression of Fas-induced lethal liver apoptosis in mice. Gastroenterology 2000; 118(2):380-389.

[40]

Kringen MK, Piehler AP, Grimholt RM, et al. Serum bilirubin concentration in healthy adult North-Europeans is strictly controlled by the UGT1A1 TA-repeat Variants. PLoS One 2014; 9(2):e90248.

[41]

Feldman AG, Sokol RJ. Neonatal cholestasis: emerging molecular diagnostics and potential novel therapeutics. Nat Rev Gastroenterol Hepatol 2019; 16(6):346-360.

[42]

Dong S, Chen QL, Song YN, et al. Mechanisms of CCl4-induced liver fibrosis with combined transcriptomic and proteomic analysis. J Toxicol Sci 2016; 41(4):561-572.

[43]

Hong X, Huang S, Jiang H, et al. Alcohol-related liver disease (ALD): Current perspectives on pathogenesis, therapeutic strategies, and animal models. Front Pharmacol 2024;15:1432480.

[44]

Farghali H, Kemelo MK, Canová NK. SIRT1 modulators in experimentally induced liver injury. Oxid Med Cell Longev 2019;2019:1-15.

[45]

Recena Aydos L, Aparecida Do Amaral L, Serafim De Souza R, et al. Nonalcoholic fatty liver disease induced by high-fat diet in C57bl/6 models. Nutrients 2019; 11(12):3067.

[46]

Pan PH, Wang YY, Lin SY, et al. Plumbagin ameliorates bile duct ligation-induced cholestatic liver injury in rats. Biomed Pharmacother 2022;151:113133.

[47]

Zhao YL, Ma X, Zhu Y, et al. Paeonia lactiflora Pall. protects against ANIT-induced cholestasis by activating Nrf2 via PI3K/Akt signaling pathway. Drug Des Devel Ther 2015;9:5061.

[48]

Li M, Cai SY, Boyer JL. Mechanisms of bile acid mediated inflammation in the liver. Mol Aspects Med 2017;56:45-53.

[49]

Chiang JYL, Ferrell JM. Bile acid metabolism in liver pathobiology. Gene Expr 2018; 18(2):71-87.

[50]

Cai JW, Rimal BP, Jiang CT, et al. Bile acid metabolism and signaling, the microbiota, and metabolic disease. Pharmacol Ther 2022;237:108238.

[51]

Liu ZM, Zhang ZF, Huang M, et al. Taurocholic acid is an active promoting factor, not just a biomarker of progression of liver cirrhosis: evidence from a human metabolomic study and in vitro experiments. BMC Gastroenterol 2018; 18(1):112.

[52]

Salhab A, Amer J, Lu YY, et al. Sodium+/taurocholate cotransporting polypeptide as target therapy for liver fibrosis. Gut 2021; 71(7):1373-1385.

[53]

Chiang JYL, Ferrell JM. Discovery of farnesoid X receptor and its role in bile acid metabolism. Mol Cell Endocrinol 2022;548:111618.

[54]

Tian SY, Chen SM, Pan CX, et al. FXR: structures, biology, and drug development for NASH and fibrosis diseases. Acta Pharmacol Sin 2022; 43(5):1120-1132.

[55]

Qin T, Hasnat M, Wang Z, et al. Geniposide alleviated bile acid-associated NLRP3 inflammasome activation by regulating SIRT1/FXR signaling in bile duct ligation-induced liver fibrosis. Phytomedicine 2023;118:154971.

[56]

Chen XL, Su SL, Liu R, et al. Chemical constituents and pharmacological action of bile acids from animal: a review. China J Chin Mater Med 2021; 46(19):4898-4906.

[57]

Ocvirk S, O’keefe SJD. Dietary fat, bile acid metabolism and colorectal cancer. Semin Cancer Biol 2021;73:347-355.

[58]

Simbrunner B, Trauner M, Reiberger T. Review article: therapeutic aspects of bile acid signalling in the gut-liver axis. Aliment Pharmacol Ther 2021; 54(10):1243-1262.

[59]

Rizzolo D, Kong B, Taylor RE, et al. Bile acid homeostasis in female mice deficient in Cyp7a1 and Cyp27a1. Acta Pharm Sin B 2021; 11(12):3847-3856.

[60]

Yan Y, Niu ZM, Sun C, et al. Hepatic thyroid hormone signalling modulates glucose homeostasis through the regulation of GLP-1 production via bile acid-mediated FXR antagonism. Nat Commun 2022; 13(1):6408.

[61]

Zaborska KE, Cummings BP.Rethinking bile acid metabolism and signaling for type 2 diabetes treatment. Curr Diab Rep 2018; 18(11):109.

[62]

Kong LN, Dong RC, Huang K, et al. Yangonin modulates lipid homeostasis, ameliorates cholestasis and cellular senescence in alcoholic liver disease via activating nuclear receptor FXR. Phytomedicine 2021;90:153629.

[63]

Clifford BL, Sedgeman LR, Williams KJ, et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab 2021; 33(8):1671-1684.e4.

[64]

Kosters A, Felix JC, Desai MS, et al. Impaired bile acid handling and aggravated liver injury in mice expressing a hepatocyte-specific RXRα variant lacking the DNA-binding domain. J Hepatol 2014; 60(2):362-369.

[65]

Pawlak M, Lefebvre P, Staels B. General molecular biology and architecture of nuclear receptors. Curr Top Med Chem 2012; 12(6):486-504.

[66]

Hasegawa Y, Kishimoto S, Takahashi H, et al. Altered expression of nuclear receptors affects the expression of metabolic enzymes and transporters in a rat model of cholestasis. Biol Pharm Bull 2009; 32(12):2046-2052.

[67]

Kong B, Wang L, Chiang JYL, et al. Mechanism of tissue-specific farnesoid X receptor in suppressing the expression of genes in bile-acid synthesis in mice. Hepatology 2012; 56(3):1034-1043.

[68]

Wang LL, Wu GX, Wu FH, et al. Geniposide attenuates ANIT-induced cholestasis through regulation of transporters and enzymes involved in bile acids homeostasis in rats. J Ethnopharmacol 2017;196:178-185.

[69]

Chen Z, Ma X, Zhao Y, et al. Yinchenhao decoction in the treatment of cholestasis: a systematic review and meta-analysis. J Ethnopharmacol 2015;168:208-216.

[70]

Qin SS, Tian JZ, Zhao Y, et al. Gardenia extract protects against intrahepatic cholestasis by regulating bile acid enterohepatic circulation. J Ethnopharmacol 2024;319:117083.

[71]

Liu JX, Li Y, Sun C, et al. Geniposide reduces cholesterol accumulation and increases its excretion by regulating the FXR-mediated liver-gut crosstalk of bile acids. Pharmacol Res 2020;152:104631.

[72]

Zorov DB, Juhaszova M, Sollott SJ.Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 2014; 94(3):909-950.

[73]

Yang SS, Lian GJ. ROS and diseases: role in metabolism and energy supply. Mol Cell Biochem 2020; 467(1):1-12.

[74]

Prieto I, Monsalve M. ROS homeostasis, a key determinant in liver ischemic-preconditioning. Redox Biol 2017;12:1020-1025.

[75]

Li S, Tan HY, Wang N, et al. The role of oxidative stress and antioxidants in liver diseases. Int J Mol Sci 2015; 16(11):26087-26124.

[76]

Seitz H, Mueller S.Die Bedeutung von Cytochrom P4502E1 bei der alkoholischen Lebererkrankung und bei der alkoholmediierten Karzinogenese. Z Gastroenterol 2019; 57(1):37-45.

[77]

Cichoż Lach H. Oxidative stress as a crucial factor in liver diseases. World J Gastroenterol 2014; 20(25):8082.

[78]

Liu T, Sun L, Zhang YB, et al. Imbalanced GSH/ROS and sequential cell death. J Biochem Mol Toxicol 2021; 36(1):e22942.

[79]

Wang JM, Zhang YY, Liu RX, et al. Geniposide protects against acute alcohol-induced liver injury in mice via up-regulating the expression of the main antioxidant enzymes. Can J Physiol Pharmacol 2015; 93(4):261-267.

[80]

Yang L, Bi LP, Jin LL, et al. Geniposide ameliorates liver fibrosis through reducing oxidative stress and inflammatory respose, inhibiting apoptosis and modulating overall metabolism. Front Pharmacol 2021;12:772635.

[81]

Roehlen N, Crouchet E, Baumert TF. Liver fibrosis: mechanistic concepts and therapeutic perspectives. Cells 2020; 9(4):875.

[82]

Miyata T, Nagy LE. Programmed cell death in alcohol-associated liver disease. Clin Mol Hepatol 2020; 26(4):618-625.

[83]

Schwabe RF, Luedde T. Apoptosis and necroptosis in the liver: a matter of life and death. Nat Rev Gastroenterol Hepatol 2018; 15(12):738-752.

[84]

Zhou Y, Wu RM, Wang XQ, et al. Roles of necroptosis in alcoholic liver disease and hepatic pathogenesis. Cell Prolif 2022; 55(3):e13193.

[85]

Eskandari E, Eaves CJ.Paradoxical roles of caspase-3 in regulating cell survival, proliferation, and tumorigenesis. J Cell Biol 2022; 221(6):e202201159.

[86]

Mirzayans R, Murray D. Do TUNEL and other apoptosis assays detect cell death in preclinical studies? Int J Mol Sci 2020; 21(23):9090.

[87]

Gopisetty G, Ramachandran K, Singal R. DNA methylation and apoptosis. Mol Immunol 2006; 43(11):1729-1740.

[88]

Ashkenazi A, Fairbrother WJ, Leverson JD, et al. From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov 2017; 16(4):273-284.

[89]

Peng X, Tan L, Song J, et al. Geniposide alleviated hydrogen peroxide-induced apoptosis of human hepatocytes via altering DNA methylation. Food Chem Toxicol 2023;182:114158.

[90]

Paulusma CC, Lamers WH, Broer S, et al. Amino acid metabolism, transport and signalling in the liver revisited. Biochem Pharmacol 2022;201:115074.

[91]

Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol 2017; 27(21):R1147-R1151.

[92]

Tedesco L, Corsetti G, Ruocco C, et al. A specific amino acid formula prevents alcoholic liver disease in rodents. Am J Physiol Gastrointest Liver Physiol 2018; 314(5):G566-G582.

[93]

Elhawary NA, Aljahdali IA, Abumansour IS, et al. Genetic etiology and clinical challenges of phenylketonuria. Hum Genomics 2022; 16(1):22.

[94]

Schlegel G, Scholz R, Ullrich K, et al. Phenylketonuria: direct and indirect effects of phenylalanine. Exp Neurol 2016;281:28-36.

[95]

Cruzat V, Macedo Rogero M, Noel Keane K, et al. Glutamine: metabolism and immune function, supplementation and clinical translation. Nutrients 2018; 10(11):1564.

[96]

Coqueiro AY, Rogero MM, Tirapegui J. Glutamine as an anti-fatigue amino acid in sports nutrition. Nutrients 2019; 11(4):863.

[97]

Achamrah N, Déchelotte P, Coëffier M. Glutamine and the regulation of intestinal permeability. Curr Opin Clin Nutr Metab Care 2017; 20(1):86-91.

[98]

Ho YY, Nakato J, Mizushige T, et al. l-Ornithine stimulates growth hormone release in a manner dependent on the ghrelin system. Food Funct 2017; 8(6):2110-2114.

[99]

Paßlack N, Kohn B, Zentek J. Effects of arginine and ornithine supplementation to a high-protein diet on selected cellular immune variables in adult cats. J Vet Intern Med 2020; 34(2):852-856.

[100]

Alvarez ME, Savouré A, Szabados L. Proline metabolism as regulatory hub. Trends Plant Sci 2022; 27(1):39-55.

[101]

Zhu JJ, Schwörer S, Berisa M, et al. Mitochondrial NADP(H) generation is essential for proline biosynthesis. Science 2021; 372(6545):968-972.

[102]

Zhou Y, Men LH, Pi ZF, et al. Fecal metabolomics of type 2 diabetic rats and treatment with gardenia jasminoides ellis based on mass spectrometry technique. J Agric Food Chem 2018; 66(6):1591-1599.

[103]

Siggins RW, Mcternan PM, Simon L, et al. Mitochondrial dysfunction: At the nexus between alcohol-associated immunometabolic dysregulation and tissue injury. Int J Mol Sci 2023; 24(10):8650.

[104]

Reinke H, Asher G. Circadian clock control of liver metabolic functions. Gastroenterology 2016; 150(3):574-580.

[105]

Rui L. Energy metabolism in the liver. Compr Physiol 2014; 4(1):177-197.

[106]

Xu FB, Li YF, Cao Z, et al. AFB1-induced mice liver injury involves mitochondrial dysfunction mediated by mitochondrial biogenesis inhibition. Ecotoxicol Environ Saf 2021;216:112213.

[107]

Fromenty B, Roden M. Mitochondrial alterations in fatty liver diseases. J Hepatol 2023; 78(2):415-429.

[108]

Mansouri A, Gattolliat CH, Asselah T. Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology 2018; 155(3):629-647.

[109]

Han D, Johnson HS, Rao MP, et al. Mitochondrial remodeling in the liver following chronic alcohol feeding to rats. Free Radic Biol Med 2017;102:100-110.

[110]

Qiu S, Zhang AH, Zhang TL, et al. Dissect new mechanistic insights for geniposide efficacy on the hepatoprotection using multiomics approach. Oncotarget 2017; 8(65):108760-108770.

[111]

Lv C, Liu XL, Liu HJ, et al. Geniposide attenuates mitochondrial dysfunction and memory deficits in APP/PS1 transgenic mice. Curr Alzheimer Res 2014; 11(6):580-587.

[112]

Martínez Reyes I, Chandel NS. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun 2020; 11(1):102.

[113]

Han S, Liu Y, Cai SJ, et al. IDH mutation in glioma: molecular mechanisms and potential therapeutic targets. Br J Cancer 2020; 122(11):1580-1589.

[114]

Dang L, Yen K, Attar EC. IDH mutations in cancer and progress toward development of targeted therapeutics. Ann Oncol 2016; 27(4):599-608.

[115]

Pirozzi CJ, Yan H. The implications of IDH mutations for cancer development and therapy. Nat Rev Clin Oncol 2021; 18(10):645-661.

[116]

Zarei M, Hue JJ, Hajihassani O, et al. Clinical development of IDH1 inhibitors for cancer therapy. Cancer Treat Rev 2022;103:102334.

[117]

Wang Y, Agarwal E, Bertolini I, et al. IDH2 reprograms mitochondrial dynamics in cancer through a HIF-1α-regulated pseudohypoxic state. FASEB J 2019; 33(12):13398-13411.

[118]

Wang XJ, Zhang AH, Yan GL, et al. Metabolomics and proteomics annotate therapeutic properties of geniposide: targeting and regulating multiple perturbed pathways. PLoS One 2013; 8(8):e71403.

[119]

Xie ZF, Zhang M, Song Q, et al. Development of the novel ACLY inhibitor 326E as a promising treatment for hypercholesterolemia. Acta Pharm Sin B 2023; 13(2):739-753.

[120]

Jeon S, Carr R. Alcohol effects on hepatic lipid metabolism. J Lipid Res 2020; 61(4):470-479.

[121]

You M, Arteel GE. Effect of ethanol on lipid metabolism. J Hepatol 2019; 70(2):237-248.

[122]

Pal SC, Méndez Sánchez N. Insulin resistance and adipose tissue interactions as the cornerstone of metabolic (dysfunction)- associated fatty liver disease pathogenesis. World J Gastroenterol 2023; 29(25):3999-4008.

[123]

Satapati S, Sunny NE, Kucejova B, et al. Elevated TCA cycle function in the pathology of diet-induced hepatic insulin resistance and fatty liver. J Lipid Res 2012; 53(6):1080-1092.

[124]

Todisco S, Santarsiero A, Convertini P, et al. PPAR Alpha as a metabolic modulator of the liver: role in the pathogenesis of nonalcoholic steatohepatitis (NASH). Biology 2022; 11(5):792.

[125]

Ma TT, Huang C, Zong GJ, et al. Hepatoprotective effects of geniposide in a rat model of nonalcoholic steatohepatitis. J Pharm Pharmacol 2011; 63(4):587-593.

[126]

Kojima K, Shimada T, Nagareda Y, et al. Preventive effect of geniposide on metabolic disease status in spontaneously obese type 2 diabetic mice and free fatty acid-treated HepG2 cells. Biol Pharm Bull 2011; 34(10):1613-1618.

[127]

Leng EN, Xiao Y, Mo ZT, et al. Synergistic effect of phytochemicals on cholesterol metabolism and lipid accumulation in HepG2 cells. BMC Complement Altern Med 2018; 18(1):122.

[128]

Shen D, Zhao D, Yang X, et al. Geniposide against atherosclerosis by inhibiting the formation of foam cell and lowering reverse lipid transport via p38/MAPK signaling pathways. Eur J Pharmacol 2019;864:172728.

[129]

Kano K, Aoki J, Hla T. Lysophospholipid mediators in health and disease. Annu Rev Pathol 2022; 17(1):459-483.

[130]

Jia JJ, Zhang HY, Liang XY, et al. Application of metabolomics to the discovery of biomarkers for ischemic stroke in the murine model: a comparison with the clinical results. Mol Neurobiol 2021; 58(12):6415-6426.

[131]

Chin CF, Galam DLA, Gao L, et al. Blood-derived lysophospholipid sustains hepatic phospholipids and fat storage necessary for hepatoprotection in overnutrition. J Clin Invest 2023; 133(17):e171267.

[132]

Meyer JJ, Dreyhaupt J, Schwerdel D, et al. Blood-based targeted metabolomics discriminate patients with alcoholic liver cirrhosis from those with non-cirrhotic liver damage: an explorative study. Dig Dis 2021; 40(2):223-231.

[133]

Innes JK, Calder PC.Omega-6 fatty acids and inflammation. Prostaglandins Leukot Essent Fatty Acids 2018;132:41-48.

[134]

Lucas GNC, Leitão ACC, Alencar RL, et al. Pathophysiological aspects of nephropathy caused by non-steroidal anti-inflammatory drugs. J Bras Nefrol 2019; 41(1):124-130.

[135]

Sonnweber T, Pizzini A, Nairz M, et al. Arachidonic acid metabolites in cardiovascular and metabolic diseases. Int J Mol Sci 2018; 19(11):3285.

[136]

Gai ZB, Visentin M, Gui T, et al. Effects of farnesoid X receptor activation on arachidonic acid metabolism, NF-kB signaling, and hepatic inflammation. Mol Pharmacol 2018; 94(2):802-811.

[137]

Melgar Lesmes P, Perramon M, Jiménez W. Roles of the hepatic endocannabinoid and apelin systems in the pathogenesis of liver fibrosis. Cells 2019; 8(11):1311.

[138]

Konishi T, Lentsch AB. Changes in arachidonic acid metabolism during liver ischemia triggers induction of inflammatory injury. Hepatology 2018; 68(4):1642-1643.

[139]

Kawahara K, Hohjoh H, Inazumi T, et al. Prostaglandin E2-induced inflammation: Relevance of prostaglandin E receptors. Biochim Biophys Acta 2015; 1851(4):414-421.

[140]

Hu XL, Yu DS, Zhuang LE, et al. Geniposide improves hepatic inflammation in diabetic db/db mice. Int Immunopharmacol 2018;59:141-147.

[141]

Wu H, Wu H, Yuan MY, et al. Pharmacokinetics and tissue distribution of jasminoidin in normal rats. J Anhui Tradit Chin Med Coll 2011; 30(2):57-60.

[142]

Yu D, Zhang Y, Guo LW, et al. Study on the absorption mechanism of geniposide in the Chinese formula Huang-Lian-Jie-Du- Tang in rats. AAPS PharmSciTech 2017; 18(4):1382-1392.

[143]

Wang YG, Li LL, Li HY, et al. Transdermal permeation of geniposide in the herbal complex liniment in vivo and in vitro. Int J Pharm 2010; 392(1-2):72-77.

[144]

Wang FG, Cao J, Hao JF, et al. Pharmacokinetics, bioavailability and tissue distribution of geniposide following intravenous and peroral administration to rats. Biopharm Drug Dispos 2014; 35(2):97-103.

[145]

Yu B, Ruan M, Cui XB, et al. Effects of borneol on the pharmacokinetics of geniposide in cortex, hippocampus, hypothalamus and striatum of conscious rat by simultaneous brain microdialysis coupled with UPLC-MS. J Pharm Biomed Anal 2013;77:128-132.

[146]

Wan JY, Long Y, Liu SY, et al. Geniposide-loaded liposomes for brain targeting: development, evaluation, and in vivo studies. AAPS PharmSciTech 2021; 22(7):222.

[147]

Chen CG, Han FX, Zhang Y, et al. Simultaneous determination of geniposide and its metabolites genipin and genipinine in culture of Aspergillus niger by HPLC. Biomed Chromatogr 2008; 22(7):753-757.

[148]

Han H, Yang L, Xu Y, et al. Identification of metabolites of geniposide in rat urine using ultra-performance liquid chromatography combined with electrospray ionization quadrupole time-of-flight tandem mass spectrometry. Rapid Commun Mass Spectrom 2011; 25(21):3339-3350.

[149]

Gong GH, Zheng ZM, Liu H, et al. Purification and characterization of a β-glucosidase from Aspergillus niger and its application in the hydrolysis of geniposide to genipin. J Microbiol Biotechnol 2014; 24(6):788-794.

[150]

Ding Y, Hou JW, Zhang Y, et al. Metabolism of genipin in rat and identification of metabolites by using ultraperformance liquid chromatography/quadrupole time-of-flight tandem mass spectrometry. Evid Based Complement Alternat Med 2013;2013:1-13.

[151]

Li Y, Cai W, Cai Q, et al. Comprehensive characterization of the in vitro and in vivo metabolites of geniposide in rats using ultra-high-performance liquid chromatography coupled with linear ion trap-Orbitrap mass spectrometer. Xenobiotica 2015; 46(4):357-368.

[152]

Li Y, Pan H, Li X, et al. Role of intestinal microbiota-mediated genipin dialdehyde intermediate formation in geniposide-induced hepatotoxicity in rats. Toxicol Appl Pharmacol 2019;377:114624.

[153]

Zhao YL, Huang HY, Lv N, et al. Glutathione s-transferases mediate in vitro and in vivo inactivation of genipin: Implications for an underlying detoxification mechanism. J Agric Food Chem 2023; 71(5):2399-2410.

[154]

Khanal T, Kim HG, Choi JH, et al. Biotransformation of geniposide by human intestinal microflora on cytotoxicity against HepG2 cells. Toxicol Lett 2012; 209(3):246-254.

[155]

Hobbs CA, Koyanagi M, Swartz C, et al. Genotoxicity evaluation of the naturally-derived food colorant, gardenia blue, and its precursor, genipin. Food Chem Toxicol 2018;118:695-708.

[156]

Qiu JN, Lin C, Ren GL, et al. Geniposide dosage and administration time: Balancing therapeutic benefits and adverse reactions in liver disease treatment. Phytomedicine 2024;132:155799.

[157]

Zeng XY, Jiang JJ, Liu SM, et al. Bidirectional effects of geniposide in liver injury: preclinical evidence construction based on meta-analysis. J Ethnopharmacol 2024;319:117061.

[158]

Tian JZ, Yi Y, Zhao Y, et al. Oral chronic toxicity study of geniposide in rats. J Ethnopharmacol 2018;213:166-175.

[159]

Ding Y, Zhang T, Tao JS, et al. Potential hepatotoxicity of geniposide, the major iridoid glycoside in dried ripe fruits of Gardenia jasminoides (Zhi-zi). Nat Prod Res 2013; 27(10):929-933.

AI Summary AI Mindmap
PDF (685KB)

441

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/