Drug-induced liver injury is a major safety issue in anti-tuberculosis (TB) chemotherapy. The long- term goal of our research is to improve the safety profile of medication with anti-TB drugs. The objective of this application is to determine the mechanism of liver injury associated with rifampicin and isoniazid co-therapy. Extensive studies have been conducted previously to investigate the liver injury caused by rifampicin and isoniazid in mice or rats;however, none of these studies mimicked the hepatotoxicity in humans. Species differences between rodents and humans in responding to rifampicin and/or isoniazid are expected. Pregnane X receptor (PXR) is a transcription factor regulating a gene network involved in the metabolism of xenobiotics and endobiotics. The ability of chemicals to activate PXR is species- dependent. Rifampicin is a human specific PXR activator, which strongly activates human PXR, but has a very weak effect on mouse PXR. To overcome the species differences in ligand-dependent PXR activation, we generated a PXR-humanized mouse model. By using these PXR-humanized mice, we noted that rifampicin and isoniazid-induced liver injury is human PXR-dependent. However, rifampicin- mediated PXR activation does not alter isoniazid metabolism. By using a metabolomic approach, we found that rifampicin and isoniazid co-treatment caused protoporphyrin IX (PP-IX) accumulation, specifically in liver, and this is human PXR-dependent. PP-IX is an intermediate in porphyrin synthesis, and has been shown to be hepatotoxic in previous studies. Based upon our preliminary data and previous reports, we hypothesize that rifampicin and isoniazid co-treatment disturbs porphyrin synthesis, and the accumulation of PP-IX in liver mediates the hepatotoxicity. To test our hypothesis, we will pursue the following two specific aims: (1) identify the toxic mediator(s) in the liver injury caused by rifampicin and isoniazid co-therapy. Our working hypothesis is that the accumulation of PP-IX in liver is the key mediator of the hepatotoxicity caused by rifampicin and isoniazid co-therapy;and (2) determine the human PXR-dependent pathway(s) responsible for the hepatotoxicity in rifampicin and isoniazid co- therapy. Our working hypothesis is that human PXR-mediated up-regulation of aminolevulinic acid synthase 1, the rate-limiting enzyme in porphyrin synthesis in liver, is critical in the hepatotoxicity caused by rifampicin and isoniazid co-therapy. The results from these studies are expected to provide a new paradigm for the mechanistic understanding of rifampicin and isoniazid-induced hepatotoxicity. Novel strategies, based upon human PXR, aminolevulinic acid synthase 1, and PP-IX, can be developed to predict, prevent, and treat the liver injury caused by rifampicin and isoniazid co-therapy.

Public Health Relevance

Rifampicin and isoniazid co-therapy frequently causes liver injury, and even liver failure. This proposed project will provide a novel mechanistic understanding of the liver injury induced by rifampicin and isoniazid co-therapy, which can be applied to predict, prevent, and treat this toxic event.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK090305-02
Application #
8425062
Study Section
Xenobiotic and Nutrient Disposition and Action Study Section (XNDA)
Program Officer
Serrano, Jose
Project Start
2012-02-20
Project End
2013-06-30
Budget Start
2013-02-01
Budget End
2013-06-30
Support Year
2
Fiscal Year
2013
Total Cost
$167,986
Indirect Cost
$56,743
Name
University of Kansas
Department
Pharmacology
Type
Schools of Medicine
DUNS #
016060860
City
Kansas City
State
KS
Country
United States
Zip Code
66160
Bao, Yifan; Ma, Xiaochao; Rasmussen, Theodore P et al. (2018) Genetic Variations Associated with Anti-Tuberculosis Drug-Induced Liver Injury. Curr Pharmacol Rep 4:171-181
Zhu, Junjie; Wang, Pengcheng; Li, Feng et al. (2018) CYP1A1 and 1B1-mediated metabolic pathways of dolutegravir, an HIV integrase inhibitor. Biochem Pharmacol 158:174-184
Piekos, Stephanie C; Chen, Liming; Wang, Pengcheng et al. (2018) Consequences of Phenytoin Exposure on Hepatic Cytochrome P450 Expression during Postnatal Liver Maturation in Mice. Drug Metab Dispos 46:1241-1250
Wang, Pengcheng; Sachar, Madhav; Guo, Grace L et al. (2018) Liver metabolomics in a mouse model of erythropoietic protoporphyria. Biochem Pharmacol 154:474-481
Wang, Pengcheng; Shehu, Amina I; Lu, Jie et al. (2017) Deficiency of N-acetyltransferase increases the interactions of isoniazid with endobiotics in mouse liver. Biochem Pharmacol 145:218-225
Wang, Pengcheng; Shehu, Amina I; Ma, Xiaochao (2017) The Opportunities of Metabolomics in Drug Safety Evaluation. Curr Pharmacol Rep 3:10-15
Sachar, Madhav; Ma, Xiaochao (2016) Role of ABCG2 in liver injury associated with erythropoietic protoporphyria. Hepatology 64:305
Sachar, Madhav; Li, Feng; Liu, Ke et al. (2016) Chronic Treatment with Isoniazid Causes Protoporphyrin IX Accumulation in Mouse Liver. Chem Res Toxicol 29:1293-7
Li, Feng; Wang, Pengcheng; Liu, Ke et al. (2016) A High Dose of Isoniazid Disturbs Endobiotic Homeostasis in Mouse Liver. Drug Metab Dispos 44:1742-1751
Wang, Pengcheng; Pradhan, Komal; Zhong, Xiao-Bo et al. (2016) Isoniazid metabolism and hepatotoxicity. Acta Pharm Sin B 6:384-392

Showing the most recent 10 out of 20 publications