Changes in the expression of drug-metabolizing enzymes can alter the therapeutic or toxicologic response to a drug and lead to serious adverse drug interactions ? a major public health problem in the US. Cytochromes P450 (CYPs) are the most prevalent drug-metabolizing enzymes and the human pregnane X receptor (hPXR) plays a central role in activating the expression of CYP. CYP3A4 catalyzes the metabolism of more than 50% of clinically used drugs. hPXR activation and CYP3A4-mediated drug metabolism occur primarily in hepatocytes. Importantly, expression of CYPs, including CYP3A4, is significantly lower in proliferating hepatocytes of regenerating livers than in quiescent hepatocytes in the normal livers. However, the molecular mechanism responsible for the reduction in levels of drug-metabolizing enzymes in proliferating hepatocytes is unknown. Repressed levels of drug-metabolizing enzymes can have a profound effect on drug efficacy and toxicity. Because liver regeneration is triggered by liver injuries caused by a large range of insults, which affect people of all ages and backgrounds, it is important to study the regulation of PXR in proliferating hepatocytes in order to predict and prevent adverse drug interactions in patients with regenerating livers. The objective of the proposed study is to determine the mechanism by which hPXR activity and CYP3A4 expression are repressed in proliferating hepatocytes. Our central hypothesis is that Cdks phosphorylate and repress hPXR activity, thereby causing the reduction of CYP expression in proliferating hepatocytes. The rationale for conducting the proposed research is that elucidating the mechanisms by which hPXR activity and CYP3A4 expression are repressed in proliferating hepatocytes will provide fundamentally novel insights into the regulation of drug metabolism and improve our understanding of the xenobiotic response, ultimately aid the prediction of the accurate dosage of drugs, and reduce the risk of adverse drug interactions. Our long-term goal is to understand how hPXR is regulated by cellular signaling pathways through phosphorylation in both normal and diseased livers in order to design more effective therapies. We plan to test our central hypothesis by the following 4 Specific Aims: (1) Identify Cdk-regulated phosphorylation sites on hPXR;(2) Determine which phosphorylation sites are causally responsible for the inhibitory effect of Cdks and how the phosphorylation affects the activity of hPXR;(3) Determine the extent to which hPXR-mediated CYP3A4 expression is repressed by Cdk during the cell cycle in proliferating hepatocytes;and (4) Determine the in vivo effect of phosphorylation on the activity of hPXR. The outcomes are expected to provide fundamental novel information on the regulation of drug metabolism and disposition in proliferating hepatocytes, and will also lay the foundation for the effective design of drug safety evaluations and therapeutic strategies.

Public Health Relevance

The proposed studies aim to fill the knowledge gap in an important area of drug metabolism and disposition, which will considerably improve our understanding of xenobiotic responses as well as risk prediction and prevention of adverse drug interactions in regenerating livers that affect people of all ages and backgrounds. The proposed studies are relevant to public health because adverse drug interactions contribute considerably to therapeutic-related morbidity and mortality, a major public health problem in the US. The results from the proposed studies will ultimately contribute to improving the health of human beings.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM086415-03
Application #
8269058
Study Section
Xenobiotic and Nutrient Disposition and Action Study Section (XNDA)
Program Officer
Okita, Richard T
Project Start
2010-06-01
Project End
2014-05-31
Budget Start
2012-06-01
Budget End
2013-05-31
Support Year
3
Fiscal Year
2012
Total Cost
$315,530
Indirect Cost
$122,216
Name
St. Jude Children's Research Hospital
Department
Type
DUNS #
067717892
City
Memphis
State
TN
Country
United States
Zip Code
38105
Lin, Wenwei; Chen, Taosheng (2018) Using TR-FRET to Investigate Protein-Protein Interactions: A Case Study of PXR-Coregulator Interaction. Adv Protein Chem Struct Biol 110:31-63
Lin, Wenwei; Wang, Yue-Ming; Chai, Sergio C et al. (2017) SPA70 is a potent antagonist of human pregnane X receptor. Nat Commun 8:741
Lin, Wenwei; Goktug, Asli N; Wu, Jing et al. (2017) High-Throughput Screening Identifies 1,4,5-Substituted 1,2,3-Triazole Analogs as Potent and Specific Antagonists of Pregnane X Receptor. Assay Drug Dev Technol 15:383-394
Lolodi, Ogheneochukome; Wang, Yue-Ming; Wright, William C et al. (2017) Differential Regulation of CYP3A4 and CYP3A5 and its Implication in Drug Discovery. Curr Drug Metab 18:1095-1105
Oladimeji, Peter O; Lin, Wenwei; Brewer, C Trent et al. (2017) Glucose-dependent regulation of pregnane X receptor is modulated by AMP-activated protein kinase. Sci Rep 7:46751
Bakke, Jesse; Wright, William C; Zamora, Anthony E et al. (2017) Transcription factor ZNF148 is a negative regulator of human muscle differentiation. Sci Rep 7:8138
Oladimeji, Peter; Cui, Hongmei; Zhang, Chen et al. (2016) Regulation of PXR and CAR by protein-protein interaction and signaling crosstalk. Expert Opin Drug Metab Toxicol 12:997-1010
Robbins, D; Bakke, J; Cherian, M T et al. (2016) PXR interaction with p53: a meeting of two masters. Cell Death Dis 7:e2218
Robbins, D; Cherian, M; Wu, J et al. (2016) Human pregnane X receptor compromises the function of p53 and promotes malignant transformation. Cell Death Discov 2:16023
Banerjee, Monimoy; Chai, Sergio C; Wu, Jing et al. (2016) Tryptophan 299 is a conserved residue of human pregnane X receptor critical for the functional consequence of ligand binding. Biochem Pharmacol 104:131-8

Showing the most recent 10 out of 41 publications