2,135
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Protective effect of Qingluotongbi formula against Tripterygium wilfordii induced liver injury in mice by improving fatty acid β-oxidation and mitochondrial biosynthesis

, , , , &
Pages 80-88 | Received 09 Aug 2022, Accepted 07 Dec 2022, Published online: 21 Dec 2022

References

  • Begriche K, Massart J, Robin M-A, Borgne-Sanchez A, Fromenty B. 2011. Drug-induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and deleterious consequences for the liver. J Hepatol. 54(4):773–794.
  • Brunt EM. 2000. Grading and staging the histopathological lesions of chronic hepatitis: the Knodell histology activity index and beyond. Hepatology. 31(1):241–246.
  • Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ, Mattson MP. 2010. Cellular stress responses, the Hormesis Paradigm, and Vitagenes: novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid Redox Signal. 13(11):1763–1811.
  • Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield DA, Giuffrida Stella AM. 2007. Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat Rev Neurosci. 8(10):766–775.
  • Cao L, Yan M, Li H, Zhang B, Fang P. 2015. Progress on mechanism of Tripterygium wilfordii-induced liver injury and detoxification mechanism of licorice. Zhongguo Zhong Yao Za Zhi. 40(13):2537–2541.
  • Chen X, Zhao Y, Luo W, Chen S, Lin F, Zhang X, Fan S, Shen X, Wang Y, Liang G. 2020. Celastrol induces ROS-mediated apoptosis via directly targeting peroxiredoxin-2 in gastric cancer cells. Theranostics. 10(22):10290–10308.
  • Chen Y, Yang Y, Miller ML, Shen D, Shertzer HG, Stringer KF, Wang B, Schneider SN, Nebert DW, Dalton TP. 2007. Hepatocyte-specific Gclc deletion leads to rapid onset of steatosis with mitochondrial injury and liver failure. Hepatology. 45(5):1118–1128.
  • Cheng C-F, Ku H-C, Lin H. 2018. PGC-1α as a pivotal factor in lipid and metabolic regulation. Int J Mol Sci. 19(11):3447.
  • Dai M, Peng W, Zhang T, Zhao Q, Ma X, Cheng Y, Wang C, Li F. 2022. Metabolomics reveals the role of PPARα in Tripterygium wilfordii-induced liver injury. J Ethnopharmacol. 289:115090.
  • Desvergne B, Michalik L, Wahli W. 2006. Transcriptional regulation of metabolism. Physiol Rev. 86(2):465–514.
  • Drake J, Sultana R, Aksenova M, Calabrese V, Butterfield DA. 2003. Elevation of mitochondrial glutathione by gamma-glutamylcysteine ethyl ester protects mitochondria against peroxynitrite-induced oxidative stress. J Neurosci Res. 74(6):917–927.
  • Duan J, Wang Z, Duan R, Yang C, Zhao R, Feng Q, Qin Y, Jiang J, Gu S, Lv K, et al. 2022. Therapeutic targeting of hepatic ACSL4 ameliorates NASH in mice. Hepatology. 75(1):140–153.
  • Feng Z, Zhou C, Dong S, Liu Z, Liu T, Zhou L, Zhou X. 2019. Catalpol and Panax notoginseng saponins synergistically alleviate triptolide-induced hepatotoxicity through Nrf2/ARE pathway. Toxicol in Vitro. 56:141–149.
  • Flameng W, Borgers M, Daenen W, Stalpaert G. 1980. Ultrastructural and cytochemical correlates of myocardial protection by cardiac hypothermia in man. J Thorac Cardiovasc Surg. 79(3):413–424.
  • Fromenty B, Pessayre D. 1995. Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity. Pharmacol Ther. 67(1):101–154.
  • Fu Q, Jiang Z, Zhang L. 2013. Impairment of triptolide on liver mitochondria in isolated liver mitochondria and HL7702 cell line. Chin J Integr Med. 19(9):683–688.
  • Hirsova P, Ibrabim SH, Gores GJ, Malhi H. 2016. Lipotoxic lethal and sublethal stress signaling in hepatocytes: relevance to NASH pathogenesis. J Lipid Res. 57(10):1758–1770.
  • Hu D-D, Zhao Q, Cheng Y, Xiao X-R, Huang J-F, Qu Y, Li X, Tang Y-M, Bao W-M, Yang J-H, et al. 2019. The protective roles of PPARα activation in triptolide-induced liver injury. Toxicol Sci. 171(1):1–12.
  • Kelly DP, Scarpulla RC. 2004. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev. 18(4):357–368.
  • Kersten S, Stienstra R. 2017. The role and regulation of the peroxisome proliferator activated receptor alpha in human liver. Biochimie. 136:75–84.
  • Kim Y, Lim JH, Kim EN, Hong YA, Park H-J, Chung S, Choi BS, Kim Y-S, Park JY, Kim HW, et al. 2022. Adiponectin receptor agonist ameliorates cardiac lipotoxicity via enhancing ceramide metabolism in type 2 diabetic mice. Cell Death Dis. 13(3):282.
  • Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, Ferrell LD, Liu Y-C, Torbenson MS, Unalp-Arida A, et al.; Nonalcoholic Steatohepatitis Clinical Research Network. 2005. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology. 41(6):1313–1321.
  • Li X, Xu Z, Jiang Z, Sun L, Ji J, Miao J, Zhang X, Li X, Huang S, Wang T, et al. 2014. Hypoglycemic effect of catalpol on high-fat diet/streptozotocin-induced diabetic mice by increasing skeletal muscle mitochondrial biogenesis. Acta Biochim Biophys Sin. 46(9):738–748.
  • Liu X, Hu C, Li H, Wu L, Xiong Y, Tang X, Deng S. 2021. Metabolic profiling of fatty acids in Tripterygium wilfordii multiglucoside- and triptolide-induced liver-injured rats. Open Life Sci. 16(1):184–197.
  • Liu X, Liu Y, Cheng M, Xiao H. 2015. Metabolomic responses of human hepatocytes to emodin, aristolochic acid, and triptolide: chemicals purified from traditional Chinese medicines. J Biochem Mol Toxicol. 29(11):533–543.
  • Machado MV, Diehl AM. 2016. Pathogenesis of nonalcoholic steatohepatitis. Gastroenterology. 150(8):1769–1777.
  • Malaguarnera L, Di Rosa M, Zambito AM, dell’Ombra N, Nicoletti F, Malaguarnera M. 2006. Chitotriosidase gene expression in Kupffer cells from patients with non‐alcoholic fatty liver disease. Gut. 55(9):1313–1320.
  • Morio B, Panthu B, Bassot A, Rieusset J. 2021. Role of mitochondria in liver metabolic health and diseases. Cell Calcium. 94:102336.
  • Natarajan SK, Eapen CE, Pullimood AB, Balasubramanian KA. 2006. Oxidative stress in experimental liver microvesicular steatosis: role of mitochondria and peroxisomes. J Gastroenterol Hepatol. 21(8):1240–1249.
  • Perluigi M, Di Domenico F, Giorgi A, Schininà ME, Coccia R, Cini C, Bellia F, Cambria MT, Cornelius C, Butterfield DA, et al. 2010. Redox proteomics in aging rat brain: involvement of mitochondrial reduced glutathione status and mitochondrial protein oxidation in the aging process. J Neurosci Res. 88(16):3498–3507.
  • Selivanov VA, Votyakova TV, Pivtoraiko VN, Zeak J, Sukhomlin T, Trucco M, Roca J, Cascante M. 2011. Reactive oxygen species production by forward and reverse electron fluxes in the mitochondrial respiratory chain. PLoS Comput Biol. 7(3):e1001115.
  • Shen T, Liu Y, Shang J, Xie Q, Li J, Yan M, Xu J, Niu J, Liu J, Watkins PB, et al. 2019. Incidence and etiology of drug-induced liver injury in mainland China. Gastroenterology. 156(8):2230–2241.e11.
  • Song C, Xu Y, Lu Y. 2020. Use of Tripterygium wilfordii Hook F for immune-mediated inflammatory diseases: progress and future prospects. J Zhejiang Univ Sci B. 21(4):280–290.
  • Sun X, Johnson J, St. John JC. 2018. Global DNA methylation synergistically regulates the nuclear and mitochondrial genomes in glioblastoma cells. Nucleic Acids Res. 46(12):5977–5995.
  • VanderVeen BN, Fix DK, Carson JA. 2017. Disrupted skeletal muscle mitochondrial synamics, mitophagy, and biogenesis during cancer cachexia: a role for inflammation. Oxid Med Cell Longev. 2017:3292087.
  • Vega RB, Huss JM, Kelly DP. 2000. The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor alpha in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes. Mol Cell Biol. 20(5):1868–1876.
  • Wang Y, Palmfeldt J, Gregersen N, Makhov AM, Conway JF, Wang M, McCalley SP, Basu S, Alharbi H, St Croix C, et al. 2019. Mitochondrial fatty acid oxidation and the electron transport chain comprise a multifunctional mitochondrial protein complex. J Biol Chem. 294(33):12380–12391.
  • Weaver RJ, Blomme EA, Chadwick AE, Copple IM, Gerets HHJ, Goldring CE, Guillouzo A, Hewitt PG, Ingelman-Sundberg M, Jensen KG, et al. 2020. Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat Rev Drug Discov. 19(2):131–148.
  • Xie T, Zhou X, Wang S, Lu Y, Zhu H, Kang A, Deng H, Xu J, Shen C, Di L, et al. 2016. Development and application of a comprehensive lipidomic analysis to investigate Tripterygium wilfordii-induced liver injury. Anal Bioanal Chem. 408(16):4341–4355.
  • Yang Z-P, Bao Y-M. 2017. Research progress on regulation effect of Panax notoginseng saponins on mitochondria. Zhongguo Zhong Yao Za Zhi. 42(5):870–874.
  • Yao J, Jiang Z, Duan W, Huang J, Zhang L, Hu L, He L, Li F, Xiao Y, Shu B, et al. 2008. Involvement of mitochondrial pathway in triptolide-induced cytotoxicity in human normal liver L-02 cells. Biol Pharm Bull. 31(4):592–597.
  • Yu Z, Feng Z, Fu L, Wang J, Li C, Zhu H, Xie T, Zhou J, Zhou L, Zhou X. 2022. Qingluotongbi formula regulates the LXRα-ERS-SREBP-1c pathway in hepatocytes to alleviate the liver injury caused by Tripterygium wilfordii Hook. f. J Ethnopharmacol. 287:114952.
  • Zhang Y, Mao X, Li W, Chen W, Wang X, Ma Z, Lin N. 2021. Tripterygium wilfordii: an inspiring resource for rheumatoid arthritis treatment. Med Res Rev. 41(3):1337–1374.
  • Zhou L, Zhou C, Feng Z, Liu Z, Zhu H, Zhou X. 2018. Triptolide-induced hepatotoxicity can be alleviated when combined with Panax notoginseng saponins and Catapol. J Ethnopharmacol. 214:232–239.
  • Zhou X, Zhou Z, Jin M, Wang H, Wu M, Song Y, Cheng H. 2004. Clinical study of Qingluo Tongbi granules in treating 63 patients with rheumatoid arthritis of the type of yin-deficiency and heat in collaterals. J Tradit Chin Med. 24(2):83–87.