168
Views
5
CrossRef citations to date
0
Altmetric
Research Article

Exacerbated liver injury of antithyroid drugs in endotoxin-treated mice

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 615-623 | Received 11 Oct 2015, Accepted 26 Mar 2018, Published online: 03 May 2018

References

  • Abboud, G., and Kaplowitz, N., 2007. Drug-induced liver injury. Drug Safety, 30, 277–294.
  • Akai, S., et al., 2015. Kupffer cell-mediated exacerbation of methimazole-induced acute liver injury in rats. Journal of Applied Toxicology, 36, 702–715.
  • Akmal, A., and Kung, J., 2014. Propylthiouracil, and methimazole, and carbimazole-related hepatotoxicity. Expert Opinion on Drug Safety, 13, 1397–1406.
  • Antoine, D.J., Williams, D.P., and Park, B.K., 2008. Understanding the role of reactive metabolites in drug-induced hepatotoxicity: state of the science. Expert Opinion on Drug Metabolism and Toxicology, 4, 1415–1427.
  • Bachour-El Azzi, P., et al., and Others. 2014. Impact of inflammation on chlorpromazine-induced cytotoxicity and cholestatic features in HepaRG cells. Drug Metabolism and Disposition, 42, 1556–1566.
  • Bandyopadhyay, U., Biswas, K., and Banerjee, R.K., 2002. Extrathyroidal actions of antithyroid thionamides. Toxicol Letter, 128, 117–127.
  • Barton, C.C., et al., 2000. Bacterial lipopolysaccharide exposure augments aflatoxin B1-induced liver injury. Toxicological Sciences, 55, 444–452.
  • Carrion, A.F., et al., 2010. Propylthiouracil-induced acute liver failure: role of liver transplantation. International Journal of Endocrinology, 1, 1–5.
  • Chaudhari, N., et al., 2014. A molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress. Frontiers in Cellular Neuroscience, 8, 1–15.
  • Cooper, D.S., 1999. The side effects of antithyroid drugs. The Endocrinologist, 9, 457–478.
  • Cooper, D.S., 2005. Antithyroid drugs. New England Journal of Medicine, 352, 905–917.
  • Deng, X., et al., 2006. Modest inflammation enhances diclofenac hepatotoxicity in rats: role of neutrophils and bacterial translocation. Journal of Pharmacology and Expert Therapeutics, 319, 1191–1199.
  • Deng, X., et al., 2009. Inflammatory stress and idiosyncratic hepatotoxicity: hints from animal models. Pharmacological Reviews, 61, 262–282.
  • Dixon, L. J., et al., 2013. Kupffer cells in the liver, Hoboken, NJ: John Wiley & Sons, Inc.
  • Eghbal, M.A., et al., 2004. Peroxidase catalysed formation of cytotoxic prooxidant phenothiazine free radicals at physiological pH. Chemico-Biological Interactions, 151, 43–51.
  • Gallelli, L., et al., 2009. Hepatotoxicity induced by methimazole in a previously health patient. Current Drug Safety, 4, 204–206.
  • Ganey, P.E., et al., 2004. Adverse hepatic drug reactions: inflammatory episodes as consequence and contributor. Chemico-Biological Interactions, 150, 35–51.
  • Gupta, N.K., and Lewis, J.H., 2008. Review article: the use of potentially hepatotoxic drugs in patients with liver disease. Alimentary Pharmacology and Therapeutics, 28, 1021–1041.
  • Heidari, R., et al., 2014a. effects of enzyme induction and/or glutathione depletion on methimazole-induced hepatotoxicity in mice and the protective role of N-acetylcysteine. Advanced Pharmaceutical Bulletin, 4, 21–28.
  • Heidari, R., et al., 2014b. Mitigation of methimazole-induced hepatic injury by taurine in mice. Scientia Pharmaceutica, 83, 143–158.
  • Heidari, R., et al., 2014c. Factors affecting drug-induced liver injury: antithyroid drugs as instances. Clinical and Molecular Hepatology, 20, 237–248.
  • Heidari, R., et al., 2015a. Carbonyl traps as potential protective agents against methimazole-induced liver injury. Journal of Biochemical and Molecular Toxicology, 29, 173–181.
  • Heidari, R., et al., 2015b. An overview on the proposed mechanisms of antithyroid drugs-induced liver injury. Advanced Pharmaceutical Bullettin 5, 1–11.
  • Heidari, R., Babaei, H., and Eghbal, M., 2012a. Mechanisms of methimazole cytotoxicity in isolated rat hepatocytes. Drug and Chemical Toxicology, 36, 403–411.
  • Heidari, R., Babaei, H., and Eghbal, M.A., 2012b. Ameliorative effects of taurine against methimazole-induced cytotoxicity in isolated rat hepatocytes. Scientia Pharmaceutica, 80, 987–999.
  • Holt, M.P., and Ju, C., 2006. Mechanisms of drug-induced liver injury. The AAPS Journal, 8, E48–E54.
  • Jeon, S.J., et al., 2012. A case report of methimazole-induced acute liver failure successfully treated with liver transplantation. Korean Journal of Medicine, 83, 363–368.
  • Kang, H., et al., 1990. A case of methimazole-induced acute hepatic failure in a patient with chronic hepatitis B carrier. The Korean Journal of Internal Medicine, 5, 69–73.
  • Karaa, A., et al., 2008. S-adenosyl-L-methionine attenuates oxidative stress and hepatic stellate cell activation in an ethanol-LPS-induced fibrotic rat model. Shock, 30, 197–205.
  • Karamikhah, R., et al., 2015. Propylthiouracil-induced liver injury in mice and the protective role of taurine. Pharmaceutical Sciences, 21, 94–101.
  • Kobayashi, M., et al., 2012. Th2 cytokine-mediated methimazole-induced acute liver injury in mice. Journal of Applied Toxicology, 32, 823–833.
  • Kolios, G., Valatas, V., and Kouroumalis, E., & Others. 2006. Role of Kupffer cells in the pathogenesis of liver disease. World Journal of Gastroenterology, 12, 7413–7420.
  • Lee, E., et al., 1988. Oxidative metabolism of propylthiouracil by peroxidases from rat bone marrow. Xenobiotica, 18, 1135–1142.
  • Lee, K.Y., et al., 2000. Hepatic injury during treatment with antithyroid drugs in patients with hyperthyroidism. Journal of Korean Society of Endocrinology, 15, 554–560.
  • Lee, P.W., and Neal, R.A., 1978. Metabolism of methimazole by rat liver cytochrome P-450-containing monoxygenases. Drug Metabolism and Disposition, 6, 591–600.
  • Liu, S.F., Ye, X., and Malik, A.B., 1999. Pyrrolidine dithiocarbamate prevents I-κB degradation and reduces microvascular injury induced by lipopolysaccharide in multiple organs. Molecular Pharmacology, 55, 658–667.
  • Luyendyk, J.P., et al., 2003. Ranitidine treatment during a modest inflammatory response precipitates idiosyncrasy-like liver injury in rats. Journal of Pharmacology and Experimental Therapeutics, 307, 9–16.
  • Luyendyk, J.P., et al., 2004. Role of hepatic fibrin in idiosyncrasy-like liver injury from lipopolysaccharide-ranitidine coexposure in rats. Hepatology, 40, 1342–1351.
  • Macallister, S.L., et al., 2013. Modeling xenobiotic susceptibility to hepatotoxicity using an in vitro oxidative stress inflammation model 1. Canadian Journal of Physiology and Pharmacology, 91, 236–240.
  • Malozowski, S., and Chiesa, A., 2010. Propylthiouracil-induced hepatotoxicity and death. Hopefully, never more. The Journal of Clinical Endocrinology and Metabolism, 95, 3161–3163.
  • Mcgirr, L.G., Jatoe, S.D., and O'brien, P.J., 1990. Myeloperoxidase catalysed cooxidative metabolism of methimazole: Oxidation of glutathione and NADH by free radical intermediates. Chemico-Biological Interactions, 73, 279–295.
  • Mignon, A., et al., 1999. LPS challenge in D-galactosamine-sensitized mice accounts for caspase-dependent fulminant hepatitis, not for septic shock. American Journal of Respiratory and Critical Care Medicine, 159, 1308–1315.
  • Mittal, M., et al., 2014. Reactive Oxygen Species in Inflammation and Tissue Injury. Antioxidant Redox Signal, 20, 1126–1167.
  • Mizutani, T., et al., 1999. Metabolism-dependent hepatotoxicity of methimazole in mice depleted of glutathione. Journal of Applied Toxicology, 19, 193–198.
  • Mizutani, T., et al., 2000. Evidence for the involvement of N-methylthiourea, a ring cleavage metabolite, in the hepatotoxicity of methimazole in glutathione-depleted mice: structure-toxicity and metabolic studies. Chemical Research in Toxicology, 13, 170–176.
  • Moezi, L., et al., 2013. Enhanced anti-ulcer effect of pioglitazone on gastric ulcers in cirrhotic rats: The role of nitric oxide and IL-1β. Pharmacological Reports, 65, 134–143.
  • Moulin, F., et al., 2001. Hepatic and extrahepatic factors critical for liver injury during lipopolysaccharide exposure. American Journal of Physiology, 281, G1423–G1431.
  • Niknahad, H., et al., 2016. Concurrent inflammation augments antimalarial drugs-induced liver injury in rats. Advanced Pharmaceutical Bulletin, 6, 617–625.
  • Poulsen, L.L., Hyslop, R.M., and Ziegler, D.M., 1974. S-oxidation of thioureylenes catalyzed by a microsomal flavoprotein mixed-function oxidase. Biochemical Pharmacology, 23, 3431–3440.
  • Rivkees, S.A., and Mattison, D.R., 2009. Ending propylthiouracil-induced liver failure in children. The New England Journal. Medicine, 360, 1574–1575.
  • Rizzardini, M., et al., 1998. Kupffer cell depletion partially prevents hepatic heme oxygenase 1 messenger RNA accumulation in systemic inflammation in mice: role of interleukin 1beta. Hepatology, 27, 703–710.
  • Roth, R.A., et al., 2003. Inflammation and drug idiosyncrasy-is there a connection? Journal of Pharmacology and Experimental Therapeutics, 307, 1–8.
  • Roth, R.A., and Ganey, P.E., 2011. Animal models of idiosyncratic drug-induced liver injury-current status. Critical Reviews in Toxicology, 41, 723–739.
  • Sedlak, J., and Lindsay, R., 1968. Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman’s reagent. Analytical Biochemistry, 25, 192–205.
  • Su, G.L., 2002. Lipopolysaccharides in liver injury: molecular mechanisms of Kupffer cell activation. Am. J. Physiol. Gastrointest. Liver Physiol, 283, G256–G265.
  • Su, Y., et al., 2014. Lipopolysaccharide exposure augments isoniazide-induced liver injury. Journal of Applied Toxicology, 34, 1436–1442.
  • Tafazoli, S., and O'brien, P.J., 2005. Peroxidases: a role in the metabolism and side effects of drugs. Drug Discovery Today, 10, 617–625.
  • Uchiyama, M., and Mihara, M., 1978. Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Analytical Biochemistry, 86, 271–278.
  • Waldhauser, L., and Uetrecht, J., 1991. Oxidation of propylthiouracil to reactive metabolites by activated neutrophils. Implications for agranulocytosis. Drug Metabolism and Disposition, 19, 354–359.
  • Webb, G.J., and Adams, D.H., 2015. Modeling idiosyncrasy: a novel animal model of drug-induced liver injury. Hepatology, 61, 1124–1126.
  • Woeber, K.A., 2002. Methimazole-induced hepatotoxicity. Endocrine Practice, 8, 222–224.
  • Yang, J., et al., 2015a. Analysis of 90 cases of antithyroid drug-induced severe hepatotoxicity over 13 years in China. Thyroid, 25, 278–283.
  • Yang, J., et al., 2015b. Unusual synchronous methimazole-induced agranulocytosis and severe hepatotoxicity in patient with hyperthyroidism: a case report and review of the literature. International Journal of Endocrinology, 5, 1–6.
  • Zou, W., Roth, R. A., and Ganey, P. E., 2012. Animal models of idiosyncratic, drug-induced liver injury: emphasis on the inflammatory stress hypothesis. Encyclopedia of Drug Metabolism and Interaction. John Wiley & Sons, Inc. doi:10.1002/9780470921920.edm088

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.