1
|
Lei S, Guo A, Lu J, Qi Q, Devanathan AS, Zhu J, Ma X. Activation of PXR causes drug interactions with Paxlovid in transgenic mice. Acta Pharm Sin B 2023; 13:4502-4510. [PMID: 37969744 PMCID: PMC10638548 DOI: 10.1016/j.apsb.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/11/2023] [Accepted: 07/28/2023] [Indexed: 11/17/2023] Open
Abstract
Paxlovid is a nirmatrelvir (NMV) and ritonavir (RTV) co-packaged medication used for the treatment of coronavirus disease 2019 (COVID-19). The active component of Paxlovid is NMV and RTV is a pharmacokinetic booster. Our work aimed to investigate the drug/herb-drug interactions associated with Paxlovid and provide mechanism-based guidance for the clinical use of Paxlovid. By using recombinant human cytochrome P450s (CYPs), we confirmed that CYP3A4 and 3A5 are the major enzymes responsible for NMV metabolism. The role of CYP3A in Paxlovid metabolism were further verified in Cyp3a-null mice, which showed that the deficiency of CYP3A significantly suppressed the metabolism of NMV and RTV. Pregnane X receptor (PXR) is a ligand-dependent transcription factor that upregulates CYP3A4/5 expression. We next explored the impact of drug- and herb-mediated PXR activation on Paxlovid metabolism in a transgenic mouse model expressing human PXR and CYP3A4/5. We found that PXR activation increased CYP3A4/5 expression, accelerated NMV metabolism, and reduced the systemic exposure of NMV. In summary, our work demonstrated that PXR activation can cause drug interactions with Paxlovid, suggesting that PXR-activating drugs and herbs should be used cautiously in COVID-19 patients receiving Paxlovid.
Collapse
Affiliation(s)
- Saifei Lei
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Alice Guo
- School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jie Lu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Qian Qi
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Aaron S. Devanathan
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
2
|
Miners JO, Polasek TM, Hulin JA, Rowland A, Meech R. Drug-drug interactions that alter the exposure of glucuronidated drugs: Scope, UDP-glucuronosyltransferase (UGT) enzyme selectivity, mechanisms (inhibition and induction), and clinical significance. Pharmacol Ther 2023:108459. [PMID: 37263383 DOI: 10.1016/j.pharmthera.2023.108459] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Drug-drug interactions (DDIs) arising from the perturbation of drug metabolising enzyme activities represent both a clinical problem and a potential economic loss for the pharmaceutical industry. DDIs involving glucuronidated drugs have historically attracted little attention and there is a perception that interactions are of minor clinical relevance. This review critically examines the scope and aetiology of DDIs that result in altered exposure of glucuronidated drugs. Interaction mechanisms, namely inhibition and induction of UDP-glucuronosyltransferase (UGT) enzymes and the potential interplay with drug transporters, are reviewed in detail, as is the clinical significance of known DDIs. Altered victim drug exposure arising from modulation of UGT enzyme activities is relatively common and, notably, the incidence and importance of UGT induction as a DDI mechanism is greater than generally believed. Numerous DDIs are clinically relevant, resulting in either loss of efficacy or an increased risk of adverse effects, necessitating dose individualisation. Several generalisations relating to the likelihood of DDIs can be drawn from the known substrate and inhibitor selectivities of UGT enzymes, highlighting the importance of comprehensive reaction phenotyping studies at an early stage of drug development. Further, rigorous assessment of the DDI liability of new chemical entities that undergo glucuronidation to a significant extent has been recommended recently by regulatory guidance. Although evidence-based approaches exist for the in vitro characterisation of UGT enzyme inhibition and induction, the availability of drugs considered appropriate for use as 'probe' substrates in clinical DDI studies is limited and this should be research priority.
Collapse
Affiliation(s)
- John O Miners
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Thomas M Polasek
- Certara, Princeton, NJ, USA; Centre for Medicines Use and Safety, Monash University, Melbourne, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Andrew Rowland
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
3
|
Gufford BT, Metzger IF, Bamfo NO, Benson EA, Masters AR, Lu JBL, Desta Z.
Influence of CYP2B6 Pharmacogenetics on Stereoselective Inhibition and Induction of Bupropion Metabolism by Efavirenz in Healthy Volunteers.
. J Pharmacol Exp Ther 2022; 382:JPET-AR-2022-001277. [PMID: 35798386 PMCID: PMC9426761 DOI: 10.1124/jpet.122.001277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/22/2022] Open
Abstract
We investigated the acute and chronic effects of efavirenz, a widely used antiretroviral drug, and CYP2B6 genotypes on the disposition of racemic and stereoisomers of bupropion (BUP) and its active metabolites, 4-hydroxyBUP, threohydroBUP and erythrohydroBUP. The primary objective of this study was to test how multiple processes unique to the efavirenz-CYP2B6 genotype interaction influence the extent of efavirenz-mediated drug-drug interaction (DDI) with the CYP2B6 probe substrate BUP. In a three-phase, sequential, open-label study, healthy volunteers (N=53) were administered a single 100 mg oral dose of BUP alone (control phase), with a single 600 mg oral efavirenz dose (inhibition phase), and after 17-days pretreatment with efavirenz (600 mg/day) (induction phase). Compared to the control phase, we show for the first time that efavirenz significantly decreases and chronically increases the exposure of hydroxyBUP and its diastereomers, respectively, and these interactions were CYP2B6 genotype dependent. Chronic efavirenz enhances the elimination of racemic BUP and its enantiomers as well as of threo- and erythro-hydroBUP and their diastereomers, suggesting additional novel mechanisms underlying efavirenz interaction with BUP. The effects of efavirenz and genotypes were nonstereospecific. In conclusion, acute and chronic administration of efavirenz inhibits and induces CYP2B6 activity. Efavirenz-BUP interaction is complex involving time- and CYP2B6 genotype-dependent inhibition and induction of primary and secondary metabolic pathways. Our findings highlight important implications to the safety and efficacy of BUP, study design considerations for future efavirenz interactions, and individualized drug therapy based on CYP2B6 genotypes. Significance Statement The effects of acute and chronic doses of efavirenz on the disposition of racemic and stereoisomers of BUP and its active metabolites were investigated in healthy volunteers. Efavirenz causes an acute inhibition, but chronic induction of CYP2B6 in a genotype dependent manner. Chronic efavirenz induces BUP reduction and the elimination of BUP active metabolites. Efavirenz's effects were non-stereospecific. These data reveal novel mechanisms underlying efavirenz DDI with BUP and provide important insights into time- and CYP2B6 genotype dependent DDIs.
Collapse
Affiliation(s)
| | | | - Nadia O Bamfo
- Division of Clinical Pharmacology, Indiana University School of Medicine, United States
| | - Eric A Benson
- Medicine, Indiana University School of Medicine, United States
| | - Andrea R Masters
- Melvin and Bren Simon Comprehensive Cancer Center Clinical Pharmacology Analytical Core, Indiana University School of Medicine, United States
| | - Jessica Bo Li Lu
- Division of Clinical Pharmacology, Indiana University School of Medicine, United States
| | - Zeruesenay Desta
- Medicine/Division of Clinical Pharmacology, Indiana University School of Medicine, United States
| |
Collapse
|
4
|
Wang X, Yu Y, Wang P, Yang K, Wang Y, Yan L, Zhong XB, Zhang L. Long Noncoding RNAs Hepatocyte Nuclear Factor 4A Antisense RNA 1 and Hepatocyte Nuclear Factor 1A Antisense RNA 1 Are Involved in Ritonavir-Induced Cytotoxicity in Hepatoma Cells. Drug Metab Dispos 2022; 50:704-715. [PMID: 34949673 PMCID: PMC9132102 DOI: 10.1124/dmd.121.000693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022] Open
Abstract
Ritonavir (RTV), a pharmacoenhancer used in anti-HIV regimens, can induce liver damage. RTV is primarily metabolized by cytochrome P450 3A4 (CYP3A4) in the liver. HNF4A antisense RNA 1 (HNF4A-AS1) and HNF1A antisense RNA 1 (HNF1A-AS1) are long noncoding RNAs that regulate the expression of pregnane X receptor (PXR) and CYP3A4. This study investigated the role and underlying mechanisms of HNF4A-AS1 and HNF1A-AS1 in RTV-induced hepatotoxicity. HNF4A-AS1 and HNF1A-AS1 were knocked down by small hairpin RNAs in Huh7 and HepG2 cells. Lactate dehydrogenase and reactive oxygen species assays were performed to assess RTV-induced hepatotoxicity. Chromatin immunoprecipitation quantitative real-time polymerase chain reaction was used to detect PXR enrichment and histone modifications in the CYP3A4 promoter. HNF4A-AS1 knockdown increased PXR and CYP3A4 expression and exacerbated RTV-induced cytotoxicity, whereas HNF1A-AS1 knockdown generated the opposite phenotype. Mechanistically, enrichment of PXR and trimethylation of histone 3 lysine 4 (H3K4me3) in the CYP3A4 promoter was increased, and trimethylation of histone 3 lysine 27 (H3K27me3) was decreased after HNF4A-AS1 knockdown. However, PXR and H3K4me3 enrichment decreased after HNF1A-AS1 knockdown. Alterations in RTV-induced hepatotoxicity caused by decreasing HNF4A-AS1 or HNF1A-AS1 were reversed by knockdown or overexpression of PXR. Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. SIGNIFICANCE STATEMENT: HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as long noncoding RNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. This discovery provides directions for further research on the mechanisms of RTV-induced liver injury.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Yihang Yu
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Kun Yang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Yiting Wang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Liang Yan
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Xiao-Bo Zhong
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| |
Collapse
|
5
|
Polymorphisms at CYP enzymes, NR1I2 and NR1I3 in association with virologic response to antiretroviral therapy in Brazilian HIV-positive individuals. THE PHARMACOGENOMICS JOURNAL 2022; 22:33-38. [PMID: 34504302 DOI: 10.1038/s41397-021-00254-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/17/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023]
Abstract
Virologic failure of antiretroviral therapy (ART) may be explained by single nucleotide polymorphisms (SNPs) in drug absorption and metabolism genes. Here, we characterized the associations between polymorphisms in cytochrome P450 enzymes' genes CYP2B6 and CYP3A4/A5, nuclear receptor genes NR1I2/3, and initial ART efficacy among 203 HIV-positive individuals from Rio de Janeiro. Association between SNPs and virologic control was evaluated after 6 and 12 months of follow-up using Cox regression models. The SNP rs2307424 (NR1I3) was associated with increased virologic response after 12 months of treatment, while rs1523127 (NR1I2), rs3003596, and rs2502815 (NR1I3) were associated with decreased response. Increased virologic response after 12 months (adjHR = 1.54; p = 0.02) was also observed among carriers of the NR1I3 haplotype rs2502815G-rs3003596A-rs2307424A versus the reference haplotype G-A-G. Our results suggest that NR1I2 and NR1I3 variants are associated with virologic responses to ART among Brazilians.
Collapse
|
6
|
Karpale M, Hukkanen J, Hakkola J. Nuclear Receptor PXR in Drug-Induced Hypercholesterolemia. Cells 2022; 11:cells11030313. [PMID: 35159123 PMCID: PMC8833906 DOI: 10.3390/cells11030313] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a major global health concern. The central modifiable risk factors and causative agents of the disease are high total and low-density lipoprotein (LDL) cholesterol. To reduce morbidity and mortality, a thorough understanding of the factors that influence an individual’s cholesterol status during the decades when the arteria-narrowing arteriosclerotic plaques are forming is critical. Several drugs are known to increase cholesterol levels; however, the mechanisms are poorly understood. Activation of pregnane X receptor (PXR), the major regulator of drug metabolism and molecular mediator of clinically significant drug–drug interactions, has been shown to induce hypercholesterolemia. As a major sensor of the chemical environment, PXR may in part mediate hypercholesterolemic effects of drug treatment. This review compiles the current knowledge of PXR in cholesterol homeostasis and discusses the role of PXR in drug-induced hypercholesterolemia.
Collapse
Affiliation(s)
- Mikko Karpale
- Research Unit of Biomedicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland;
| | - Janne Hukkanen
- Research Unit of Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland;
| | - Jukka Hakkola
- Research Unit of Biomedicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland;
- Correspondence:
| |
Collapse
|
7
|
Lv Y, Luo YY, Ren HW, Li CJ, Xiang ZX, Luan ZL. The role of pregnane X receptor (PXR) in substance metabolism. Front Endocrinol (Lausanne) 2022; 13:959902. [PMID: 36111293 PMCID: PMC9469194 DOI: 10.3389/fendo.2022.959902] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
As a member of the nuclear receptor (NR) superfamily, pregnane X receptor (PXR; NR1I2) is a ligand-activated transcription factor that plays a crucial role in the metabolism of xenobiotics and endobiotics in mammals. The tissue distribution of PXR is parallel to its function with high expression in the liver and small intestine and moderate expression in the kidney, stomach, skin, and blood-brain barrier, which are organs and tissues in frequent contact with xenobiotics. PXR was first recognized as an exogenous substance receptor regulating metabolizing enzymes and transporters and functioning in detoxification and drug metabolism in the liver. However, further research revealed that PXR acts as an equally important endogenous substance receptor in the metabolism and homeostasis of endogenous substances. In this review, we summarized the functions of PXR in metabolism of different substances such as glucose, lipid, bile acid, vitamin, minerals, and endocrines, and also included insights of the application of PXR ligands (drugs) in specific diseases.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian Medical University, Dalian, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian Medical University, Dalian, China
- *Correspondence: Zhi-Lin Luan,
| |
Collapse
|
8
|
Rigalli JP, Theile D, Nilles J, Weiss J. Regulation of PXR Function by Coactivator and Corepressor Proteins: Ligand Binding Is Just the Beginning. Cells 2021; 10:cells10113137. [PMID: 34831358 PMCID: PMC8625645 DOI: 10.3390/cells10113137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/13/2022] Open
Abstract
The pregnane X receptor (PXR, NR1I2) is a nuclear receptor which exerts its regulatory function by heterodimerization with the retinoid-X-receptor α (RXRα, NR2B1) and binding to the promoter and enhancer regions of diverse target genes. PXR is involved in the regulation of drug metabolism and excretion, metabolic and immunological functions and cancer pathogenesis. PXR activity is strongly regulated by the association with coactivator and corepressor proteins. Coactivator proteins exhibit histone acetyltransferase or histone methyltransferase activity or associate with proteins having one of these activities, thus promoting chromatin decondensation and activation of the gene expression. On the contrary, corepressor proteins promote histone deacetylation and therefore favor chromatin condensation and repression of the gene expression. Several studies pointed to clear cell- and ligand-specific differences in the activation of PXR. In this article, we will review the critical role of coactivator and corepressor proteins as molecular determinants of the specificity of PXR-mediated effects. As already known for other nuclear receptors, understanding the complex mechanism of PXR activation in each cell type and under particular physiological and pathophysiological conditions may lead to the development of selective modulators with therapeutic potential.
Collapse
|
9
|
Benedicto AM, Fuster-Martínez I, Tosca J, Esplugues JV, Blas-García A, Apostolova N. NNRTI and Liver Damage: Evidence of Their Association and the Mechanisms Involved. Cells 2021; 10:cells10071687. [PMID: 34359857 PMCID: PMC8303744 DOI: 10.3390/cells10071687] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Due to the improved effectiveness and safety of combined antiretroviral therapy, human immunodeficiency virus (HIV) infection has become a manageable, chronic condition rather than a mortal disease. However, HIV patients are at increased risk of experiencing non-AIDS-defining illnesses, with liver-related injury standing out as one of the leading causes of death among these patients. In addition to more HIV-specific processes, such as antiretroviral drug-related toxicity and direct injury to the liver by the virus itself, its pathogenesis is related to conditions that are also common in the general population, such as alcoholic and non-alcoholic fatty liver disease, viral hepatitis, and ageing. Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are essential components of combined anti-HIV treatment due to their unique antiviral activity, high specificity, and acceptable toxicity. While first-generation NNRTIs (nevirapine and efavirenz) have been related largely to liver toxicity, those belonging to the second generation (etravirine, rilpivirine and doravirine) seem to be generally safe for the liver. Indeed, there is preclinical evidence of rilpivirine being hepatoprotective in different models of liver injury, independently of the presence of HIV. The present study aims to review the mechanisms by which currently available anti-HIV drugs belonging to the NNRTI family may participate in the development of liver disease.
Collapse
Affiliation(s)
- Ana M. Benedicto
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (A.M.B.); (I.F.-M.); (N.A.)
| | - Isabel Fuster-Martínez
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (A.M.B.); (I.F.-M.); (N.A.)
| | - Joan Tosca
- Digestive Medicine Department, University Clinical Hospital of Valencia, 46010 Valencia, Spain;
| | - Juan V. Esplugues
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (A.M.B.); (I.F.-M.); (N.A.)
- FISABIO–University Hospital Dr Peset, 46017 Valencia, Spain;
- Center for Biomedical Research Network–Hepatic and Digestive Diseases (CIBERehd), 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-963-864-167; Fax: +34-963-983-879
| | - Ana Blas-García
- FISABIO–University Hospital Dr Peset, 46017 Valencia, Spain;
- Center for Biomedical Research Network–Hepatic and Digestive Diseases (CIBERehd), 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Nadezda Apostolova
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (A.M.B.); (I.F.-M.); (N.A.)
- FISABIO–University Hospital Dr Peset, 46017 Valencia, Spain;
- Center for Biomedical Research Network–Hepatic and Digestive Diseases (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
10
|
Mast N, El-Darzi N, Petrov AM, Li Y, Pikuleva IA. CYP46A1-dependent and independent effects of efavirenz treatment. Brain Commun 2020; 2:fcaa180. [PMID: 33305262 PMCID: PMC7713991 DOI: 10.1093/braincomms/fcaa180] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/22/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Cholesterol excess in the brain is mainly disposed via cholesterol 24-hydroxylation catalysed by cytochrome P450 46A1, a CNS-specific enzyme. Cytochrome P450 46A1 is emerging as a promising therapeutic target for various brain diseases with both enzyme activation and inhibition having therapeutic potential. The rate of cholesterol 24-hydroxylation determines the rate of brain cholesterol turnover and the rate of sterol flux through the plasma membranes. The latter was shown to affect membrane properties and thereby membrane proteins and membrane-dependent processes. Previously we found that treatment of 5XFAD mice, an Alzheimer's disease model, with a small dose of anti-HIV drug efavirenz allosterically activated cytochrome P450 46A1 in the brain and mitigated several disease manifestations. Herein, we generated Cyp46a1-/- 5XFAD mice and treated them, along with 5XFAD animals, with efavirenz to ascertain cytochrome P450 46A1-dependent and independent drug effects. Efavirenz-treated versus control Cyp46a1-/- 5XFAD and 5XFAD mice were compared for the brain sterol and steroid hormone content, amyloid β burden, protein and mRNA expression as well as synaptic ultrastructure. We found that the cytochrome P450 46A1-dependent efavirenz effects included changes in the levels of brain sterols, steroid hormones, and such proteins as glial fibrillary acidic protein, Iba1, Munc13-1, post-synaptic density-95, gephyrin, synaptophysin and synapsin-1. Changes in the expression of genes involved in neuroprotection, neurogenesis, synaptic function, inflammation, oxidative stress and apoptosis were also cytochrome P450 46A1-dependent. The total amyloid β load was the same in all groups of animals, except lack of cytochrome P450 46A1 decreased the production of the amyloid β40 species independent of treatment. In contrast, altered transcription of genes from cholinergic, monoaminergic, and peptidergic neurotransmission, steroid sulfation and production as well as vitamin D3 activation was the main CYP46A1-independent efavirenz effect. Collectively, the data obtained reveal that CYP46A1 controls cholesterol availability for the production of steroid hormones in the brain and the levels of biologically active neurosteroids. In addition, cytochrome P450 46A1 activity also seems to affect the levels of post-synaptic density-95, the main postsynaptic density protein, possibly by altering the calcium/calmodulin-dependent protein kinase II inhibitor 1 expression and activity of glycogen synthase kinase 3β. Even at a small dose, efavirenz likely acts as a transcriptional regulator, yet this regulation may not necessarily lead to functional effects. This study further confirmed that cytochrome P450 46A1 is a key enzyme for cholesterol homeostasis in the brain and that the therapeutic efavirenz effects on 5XFAD mice are likely realized via cytochrome P450 46A1 activation.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Nicole El-Darzi
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Young Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
11
|
Creamer BA, Sloan SNB, Dennis JF, Rogers R, Spencer S, McCuen A, Persaud P, Staudinger JL. Associations between Pregnane X Receptor and Breast Cancer Growth and Progression. Cells 2020; 9:cells9102295. [PMID: 33076284 PMCID: PMC7602492 DOI: 10.3390/cells9102295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
Pregnane X receptor (PXR, NR1I2) is a member of the ligand-activated nuclear receptor superfamily. This receptor is promiscuous in its activation profile and is responsive to a broad array of both endobiotic and xenobiotic ligands. PXR is involved in pivotal cellular detoxification processes to include the regulation of genes that encode key drug-metabolizing cytochrome-P450 enzymes, oxidative stress response, as well as enzymes that drive steroid and bile acid metabolism. While PXR clearly has important regulatory roles in the liver and gastrointestinal tract, this nuclear receptor also has biological functions in breast tissue. In this review, we highlight current knowledge of PXR’s role in mammary tumor carcinogenesis. The elevated level of PXR expression in cancerous breast tissue suggests a likely interface between aberrant cell division and xeno-protection in cancer cells. Moreover, PXR itself exerts positive effect on the cell cycle, thereby predisposing tumor cells to unchecked proliferation. Activation of PXR also plays a key role in regulating apoptosis, as well as in acquired resistance to chemotherapeutic agents. The repressive role of PXR in regulating inflammatory mediators along with the existence of genetic polymorphisms within the sequence of the PXR gene may predispose individuals to developing breast cancer. Further investigations into the role that PXR plays in driving tumorigenesis are needed.
Collapse
|
12
|
Martí-Rodrigo A, Alegre F, Moragrega ÁB, García-García F, Martí-Rodrigo P, Fernández-Iglesias A, Gracia-Sancho J, Apostolova N, Esplugues JV, Blas-García A. Rilpivirine attenuates liver fibrosis through selective STAT1-mediated apoptosis in hepatic stellate cells. Gut 2020; 69:920-932. [PMID: 31530714 DOI: 10.1136/gutjnl-2019-318372] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Liver fibrosis constitutes a major health problem worldwide due to its rapidly increasing prevalence and the lack of specific and effective treatments. Growing evidence suggests that signalling through cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathways regulates liver fibrosis and regeneration. Rilpivirine (RPV) is a widely used anti-HIV drug not reported to produce hepatotoxicity. We aimed to describe the potential hepatoprotective effects of RPV in different models of chronic liver injury, focusing on JAK-STAT signalling regulation. DESIGN The effects of RPV on hepatic steatosis, inflammation and fibrogenesis were studied in a nutritional mouse model of non-alcoholic fatty liver disease, carbon tetrachloride-induced fibrosis and bile duct ligation-induced fibrosis. Primary human hepatic stellate cells (hHSC) and human cell lines LX-2 and Hep3B were used to investigate the underlying molecular mechanisms. RESULTS RPV exerted a clear anti-inflammatory and antifibrotic effect in all the in vivo models of liver injury employed, and enhanced STAT3-dependent proliferation in hepatocytes and apoptosis in HSC through selective STAT1 activation. These results were reproduced in vitro; RPV undermined STAT3 activation and triggered STAT1-mediated pathways and apoptosis in HSC. Interestingly, this selective pro-apoptotic effect completely disappeared when STAT1 was silenced. Conditioned medium experiments showed that HSC apoptosis activated STAT3 in hepatocytes in an interleukin-6-dependent mechanism. CONCLUSION RPV ameliorates liver fibrosis through selective STAT1-dependent induction of apoptosis in HSC, which exert paracrinal effects in hepatocytes, thus promoting liver regeneration. RPV's actions may represent an effective strategy to treat chronic liver diseases of different aetiologies and help identify novel therapeutic targets.
Collapse
Affiliation(s)
- Alberto Martí-Rodrigo
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain
| | - Fernando Alegre
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain.,FISABIO-Hospital Universitario Dr. Peset, Valencia, Spain
| | - Ángela B Moragrega
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain
| | | | - Pablo Martí-Rodrigo
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain
| | - Anabel Fernández-Iglesias
- Liver Vascular Biology Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-CIBERehd, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-CIBERehd, Barcelona, Spain.,Hepatology, Department of Biomedical Research, Inselspital, University of Bern, Bern, Switzerland
| | - Nadezda Apostolova
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain
| | - Juan V Esplugues
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain.,FISABIO-Hospital Universitario Dr. Peset, Valencia, Spain
| | - Ana Blas-García
- Department of Pharmacology, Faculty of Medicine, University of Valencia-CIBERehd, Valencia, Spain
| |
Collapse
|
13
|
Denault JS, Cabot JF, Langlois H, Marcotte S, Sheehan NL. Optimizing concentrations of concomitant antiretrovirals by reducing etravirine doses: two case reports of complex drug-drug interactions. Antivir Ther 2020; 24:73-76. [PMID: 30353885 DOI: 10.3851/imp3274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2018] [Indexed: 10/28/2022]
Abstract
We report the cases of two treatment-experienced HIV-infected patients with complex antiretroviral regimens that showed significant drug-drug interactions with etravirine. Unexpectedly high etravirine concentrations likely caused subtherapeutic levels of darunavir, elvitegravir and dolutegravir through concentration-dependent metabolic induction. Therapeutic drug monitoring allowed safe etravirine dose decreases to manage these interactions.
Collapse
Affiliation(s)
- Jean-Simon Denault
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Pharmacy Department, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Jean-François Cabot
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Pharmacy Department, McGill University Health Centre, Montréal, QC, Canada
| | - Hugo Langlois
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Pharmacy Department, Hôpital du Sacré-Coeur de Montréal, Montréal, QC, Canada
| | - Suzanne Marcotte
- Pharmacy Department, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada.,Unité hospitalière de recherche, d'enseignement et de soins sur le sida, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Nancy L Sheehan
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Pharmacy Department, McGill University Health Centre, Montréal, QC, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
14
|
Collins KS, Metzger IF, Gufford BT, Lu JB, Medeiros EB, Pratt VM, Skaar TC, Desta Z. Influence of Uridine Diphosphate Glucuronosyltransferase Family 1 Member A1 and Solute Carrier Organic Anion Transporter Family 1 Member B1 Polymorphisms and Efavirenz on Bilirubin Disposition in Healthy Volunteers. Drug Metab Dispos 2020; 48:169-175. [PMID: 31888882 PMCID: PMC7011111 DOI: 10.1124/dmd.119.089052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic administration of efavirenz is associated with decreased serum bilirubin levels, probably through induction of UGT1A1 We assessed the impact of efavirenz monotherapy and UGT1A1 phenotypes on total, conjugated, and unconjugated serum bilirubin levels in healthy volunteers. Healthy volunteers were enrolled into a clinical study designed to address efavirenz pharmacokinetics, drug interactions, and pharmacogenetics. Volunteers received multiple oral doses (600 mg/day for 17 days) of efavirenz. Serum bilirubin levels were obtained at study entry and 1 week after completion of the study. DNA genotyping was performed for UGT1A1 [*80 (C>T), *6 (G>A), *28 (TA7), *36 (TA5), and *37 (TA8)] and for SLCO1B1 [*5 (521T>C) and *1b (388A>G] variants. Diplotype predicted phenotypes were classified as normal, intermediate, and slow metabolizers. Compared with bilirubin levels at screening, treatment with efavirenz significantly reduced total, conjugated, and unconjugated bilirubin. After stratification by UGT1A1 phenotypes, there was a significant decrease in total bilirubin among all phenotypes, conjugated bilirubin among intermediate metabolizers, and unconjugated bilirubin among normal and intermediate metabolizers. The data also show that UGT1A1 genotype predicts serum bilirubin levels at baseline, but this relationship is lost after efavirenz treatment. SLCO1B1 genotypes did not predict bilirubin levels at baseline or after efavirenz treatment. Our data suggest that efavirenz may alter bilirubin disposition mainly through induction of UGT1A1 metabolism and efflux through multidrug resistance-associated protein 2. SIGNIFICANCE STATEMENT: Efavirenz likely alters the pharmacokinetics of coadministered drugs, potentially causing lack of efficacy or increased adverse effects, as well as the disposition of endogenous compounds relevant in homeostasis through upregulation of UGT1A1 and multidrug resistance-associated protein 2. Measurement of unconjugated and conjugated bilirubin during new drug development may provide mechanistic understanding regarding enzyme and transporters modulated by the new drug.
Collapse
Affiliation(s)
- Kimberly S Collins
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Ingrid F Metzger
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Brandon T Gufford
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica B Lu
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Elizabeth B Medeiros
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Victoria M Pratt
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Todd C Skaar
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Zeruesenay Desta
- Department of Medicine, Division of Clinical Pharmacology (K.S.C., I.F.M., B.T.G., J.L., T.C.S., Z.D.), and Department of Medical and Molecular Genetics (E.B.M., V.M.P.), Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
15
|
Metzger IF, Dave N, Kreutz Y, Lu JB, Galinsky RE, Desta Z. CYP2B6 Genotype-Dependent Inhibition of CYP1A2 and Induction of CYP2A6 by the Antiretroviral Drug Efavirenz in Healthy Volunteers. Clin Transl Sci 2019; 12:657-666. [PMID: 31339646 PMCID: PMC6853154 DOI: 10.1111/cts.12671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/10/2019] [Indexed: 01/11/2023] Open
Abstract
We investigated the effect of efavirenz on the activities of cytochrome P450 (CYP)1A2, CYP2A6, xanthine oxidase (XO), and N-acetyltransferase 2 (NAT2), using caffeine as a probe. A single 150 mg oral dose of caffeine was administered to healthy volunteers (n = 58) on two separate occasions; with a single 600 mg oral dose of efavirenz and after treatment with 600 mg/day efavirenz for 17 days. Caffeine and its metabolites in plasma and urine were quantified using liquid chromatography/tandem-mass spectrometry. DNA was genotyped for CYP2B6*4 (785A>G), CYP2B6*9 (516G>T), and CYP2B6*18 (983T>C) alleles using TaqMan assays. Relative to single-dose efavirenz treatment, multiple doses of efavirenz decreased CYP1A2 (by 38%) and increased CYP2A6 (by 85%) activities (P < 0.05); XO and NAT2 activities were unaffected. CYP2B6*6*6 genotype was associated with lower CYP1A2 activity following both single and multiple doses of efavirenz. No similar association was noted for CYP2A6 activity. This is the first report showing that efavirenz reduces hepatic CYP1A2 and suggesting chronic efavirenz exposure likely enhances the elimination of CYP2A6 substrates. This is also the first to report the extent of efavirenz-CYP1A2 interaction may be efavirenz exposure-dependent and CYP2B6 genotype-dependent.
Collapse
Affiliation(s)
- Ingrid F. Metzger
- Division of Clinical PharmacologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Nimita Dave
- Division of Clinical PharmacologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
- Blueprint MedicinesCambridgeMassachusettsUSA
| | - Yvonne Kreutz
- Division of Clinical PharmacologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jessica B.L. Lu
- Division of Clinical PharmacologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Raymond E. Galinsky
- Division of Clinical PharmacologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
- School of PharmacyPurdue UniversityWest LafayetteIndianaUSA
| | - Zeruesenay Desta
- Division of Clinical PharmacologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
16
|
Villanueva S, Zhang W, Zecchinati F, Mottino A, Vore M. ABC Transporters in Extrahepatic Tissues: Pharmacological Regulation in Heart and Intestine. Curr Med Chem 2019; 26:1155-1184. [PMID: 29589524 DOI: 10.2174/0929867325666180327092639] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 12/17/2022]
Abstract
ATP binding cassette (ABC) transporters are transmembrane proteins expressed in secretory epithelia like the liver, kidneys and intestine, in the epithelia exhibiting barrier function such as the blood-brain barrier and placenta, and to a much lesser extent, in tissues like reproductive organs, lungs, heart and pancreas, among others. They regulate internal distribution of endogenous metabolites and xenobiotics including drugs of therapeutic use and also participate in their elimination from the body. We here describe the function and regulation of ABC transporters in the heart and small intestine, as examples of extrahepatic tissues, in which ABC proteins play clearly different roles. In the heart, they are involved in tissue pathogenesis as well as in protecting this organ against toxic compounds and druginduced oxidative stress. The small intestine is highly exposed to therapeutic drugs taken orally and, consequently, ABC transporters localized on its surface strongly influence drug absorption and pharmacokinetics. Examples of the ABC proteins currently described are Multidrug Resistance-associated Proteins 1 and 2 (MRP1 and 2) for heart and small intestine, respectively, and P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) for both organs.
Collapse
Affiliation(s)
- Silvina Villanueva
- Instituto de Fisiologia Experimental, Facultad de Ciencias Bioquimicas y Farmaceuticas, CONICET-UNR. 2000 Rosario, Argentina
| | - Wei Zhang
- Department of Toxicology & Cancer Biology, University of Kentucky, Lexington, KY 40536-0305, United States
| | - Felipe Zecchinati
- Instituto de Fisiologia Experimental, Facultad de Ciencias Bioquimicas y Farmaceuticas, CONICET-UNR. 2000 Rosario, Argentina
| | - Aldo Mottino
- Instituto de Fisiologia Experimental, Facultad de Ciencias Bioquimicas y Farmaceuticas, CONICET-UNR. 2000 Rosario, Argentina
| | - Mary Vore
- Department of Toxicology & Cancer Biology, University of Kentucky, Lexington, KY 40536-0305, United States
| |
Collapse
|
17
|
Shehu AI, Lu J, Wang P, Zhu J, Wang Y, Yang D, McMahon D, Xie W, Gonzalez FJ, Ma X. Pregnane X receptor activation potentiates ritonavir hepatotoxicity. J Clin Invest 2019; 129:2898-2903. [PMID: 31039134 DOI: 10.1172/jci128274] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ritonavir (RTV) is on the World Health Organization's List of Essential Medicines for antiretroviral therapy, but can cause hepatotoxicity by unknown mechanisms. Multiple clinical studies found that hepatotoxicity occurred in 100% of participants who were pretreated with rifampicin or efavirenz followed by RTV-containing regimens. Both rifampicin and efavirenz are activators of the pregnane X receptor (PXR), a transcription factor with significant inter-species differences in ligand-dependent activation. Using PXR-humanized mouse models, we recapitulated the RTV hepatotoxicity observed in the clinic. PXR was found to modulate RTV hepatotoxicity through CYP3A4-dependent pathways involved in RTV bioactivation, oxidative stress, and endoplasmic reticulum stress. In summary, the current work demonstrated the essential roles of human PXR and CYP3A4 in RTV hepatotoxicity, which can be applied to guide the safe use of RTV-containing regimens in the clinic.
Collapse
Affiliation(s)
- Amina I Shehu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Jie Lu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Pengcheng Wang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Yue Wang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Deborah McMahon
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, and
| |
Collapse
|
18
|
Heck CJS, Hamlin AN, Bumpus NN. Efavirenz and Efavirenz-like Compounds Activate Human, Murine, and Macaque Hepatic IRE1 α-XBP1. Mol Pharmacol 2019; 95:183-195. [PMID: 30442673 PMCID: PMC6324649 DOI: 10.1124/mol.118.113647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
Efavirenz (EFV), a widely used antiretroviral drug, is associated with idiosyncratic hepatotoxicity and dyslipidemia. Here we demonstrate that EFV stimulates the activation in primary hepatocytes of key cell stress regulators: inositol-requiring 1α (IRE1α) and X-box binding protein 1 (XBP1). Following EFV exposure, XBP1 splicing (indicating activation) was increased 35.7-fold in primary human hepatocytes. In parallel, XBP1 splicing and IRE1α phosphorylation (p-IRE1α, active IRE1α) were elevated 36.4-fold and 4.9-fold, respectively, in primary mouse hepatocytes. Of note, with EFV treatment, 47.2% of mouse hepatocytes were apoptotic; which was decreased to 23.9% in the presence of STF 083010, an inhibitor of XBP1 splicing. Experiments performed using pregnane X receptor (PXR)-null mouse hepatocytes revealed that EFV-mediated XBP1 splicing and hepatocyte death were not dependent on PXR, which is a nuclear receptor transcription factor that plays a crucial role in the cellular response to xenobiotics. Interestingly, incubation with the primary metabolite of EFV, 8-hydroxyefavirenz (8-OHEFV), only resulted in 10.3- and 2.9-fold increased XBP1 splicing in human and mouse hepatocytes and no change in levels of p-IRE1α in mouse hepatocytes. To further probe the structure-activity relationship of IRE1α-XBP1 activation by EFV, 16 EFV analogs were employed. Of these, an analog in which the EFV alkyne is replaced with an alkene and an analog in which the oxazinone oxygen is replaced by a carbon stimulated XBP1 splicing in human, mouse, and macaque hepatocytes. These data demonstrate that EFV and compounds sharing the EFV scaffold can activate IRE1α-XBP1 across human, mouse, and macaque species.
Collapse
Affiliation(s)
- Carley J S Heck
- Department of Pharmacology and Molecular Sciences (C.J.S.H., N.N.B.) and Department of Medicine, Division of Clinical Pharmacology (A.N.H., N.N.B.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Allyson N Hamlin
- Department of Pharmacology and Molecular Sciences (C.J.S.H., N.N.B.) and Department of Medicine, Division of Clinical Pharmacology (A.N.H., N.N.B.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Namandjé N Bumpus
- Department of Pharmacology and Molecular Sciences (C.J.S.H., N.N.B.) and Department of Medicine, Division of Clinical Pharmacology (A.N.H., N.N.B.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Narayanan B, Lade JM, Heck CJ, Dietz KD, Wade H, Bumpus NN. Probing Ligand Structure-Activity Relationships in Pregnane X Receptor (PXR): Efavirenz and 8-Hydroxyefavirenz Exhibit Divergence in Activation. ChemMedChem 2018; 13:736-747. [PMID: 29430850 PMCID: PMC6081956 DOI: 10.1002/cmdc.201700730] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/06/2018] [Indexed: 12/24/2022]
Abstract
Efavirenz (EFV), an antiretroviral that interacts clinically with co-administered drugs via activation of the pregnane X receptor (PXR), is extensively metabolized by the cytochromes P450. We tested whether its primary metabolite, 8-hydroxyEFV (8-OHEFV) can activate PXR and potentially contribute to PXR-mediated drug-drug interactions attributed to EFV. Luciferase reporter assays revealed that despite only differing from EFV by an oxygen atom, 8-OHEFV does not activate PXR. Corroborating this, treatment with EFV for 72 h elevated the mRNA abundance of the PXR target gene, Cyp3a11, by approximately 28-fold in primary hepatocytes isolated from PXR-humanized mice, whereas treatment with 8-OHEFV did not result in a change in Cyp3A11 mRNA levels. FRET-based competitive binding assays and isothermal calorimetry demonstrated that even with the lack of ability to activate PXR, 8-OHEFV displays an affinity for PXR (IC50 12.1 μm; KD 7.9 μm) nearly identical to that of EFV (IC50 18.7 μm; KD 12.5 μm). The use of 16 EFV analogues suggest that other discreet changes to the EFV structure beyond the 8-position are well tolerated. Molecular docking simulations implicate an 8-OHEFV binding mode that may underlie its divergence in PXR activation from EFV.
Collapse
Affiliation(s)
- Bhargavi Narayanan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe Street Hunterian 709 Baltimore, MD, USA
| | - Julie M. Lade
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St Biophysics 307 Baltimore, MD, USA
| | - Carley J.S. Heck
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St Biophysics 307 Baltimore, MD, USA
| | - Kevin D. Dietz
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe Street Hunterian 709 Baltimore, MD, USA
| | - Herschel Wade
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe Street Hunterian 709 Baltimore, MD, USA
| | - Namandjé N. Bumpus
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, 725 N Wolfe St Biophysics 307 Baltimore, MD, USA
| |
Collapse
|
20
|
Rigalli JP, Reichel M, Reuter T, Tocchetti GN, Dyckhoff G, Herold-Mende C, Theile D, Weiss J. The pregnane X receptor (PXR) and the nuclear receptor corepressor 2 (NCoR2) modulate cell growth in head and neck squamous cell carcinoma. PLoS One 2018; 13:e0193242. [PMID: 29470550 PMCID: PMC5823449 DOI: 10.1371/journal.pone.0193242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/07/2018] [Indexed: 01/19/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most frequent cancer worldwide. The pregnane X receptor (PXR) is a nuclear receptor regulating several target genes associated with cancer malignancy. We here demonstrated a significant effect of PXR on HNSCC cell growth, as evidenced in PXR knock-down experiments. PXR transcriptional activity is more importantly regulated by the presence of coactivators and corepressors than by PXR protein expression. To date, there is scarce information on the regulation of PXR in HNSCC and on its role in the pathogenesis of this disease. Coactivator and corepressor expression was screened through qRT-PCR in 8 HNSCC cell lines and correlated to PXR activity, determined by using a reporter gene assay. All cell lines considerably expressed all the cofactors assessed. PXR activity negatively correlated with nuclear receptor corepressor 2 (NCoR2) expression, indicating a major role of this corepressor in PXR modulation and suggesting its potential as a surrogate for PXR activity in HNSCC. To test the association of NCoR2 with the malignant phenotype, a subset of three cell lines was transfected with an over-expression plasmid for this corepressor. Subsequently, cell growth and chemoresistance assays were performed. To elucidate the mechanisms underlying NCoR2 effects on cell growth, caspase 3/7 activity and protein levels of cleaved caspase 3 and PARP were evaluated. In HNO97 cells, NCoR2 over-expression decreased cell growth, chemoresistance and increased cleaved caspase 3 levels, caspase activity and cleaved PARP levels. On the contrary, in HNO124 and HNO210 cells, NCoR2 over-expression increased cell growth, drug resistance and decreased cleaved caspase 3 levels, caspase activity and cleaved PARP levels. In conclusion, we demonstrated a role of PXR and NCoR2 in the modulation of cell growth in HNSCC. This may contribute to a better understanding of the highly variable HNSCC therapeutic response.
Collapse
Affiliation(s)
- Juan Pablo Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Matthias Reichel
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Tasmin Reuter
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Guillermo Nicolás Tocchetti
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
- Institute of Experimental Physiology (IFISE-CONICET), Rosario, Argentina
| | - Gerhard Dyckhoff
- Molecular Cell Biology Group, Department of Otorhinolaryngology, Head and Neck Surgery, University of Heidelberg, Heidelberg, Germany
| | - Christel Herold-Mende
- Molecular Cell Biology Group, Department of Otorhinolaryngology, Head and Neck Surgery, University of Heidelberg, Heidelberg, Germany
- Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Dirk Theile
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
21
|
Apostolova N, Blas-Garcia A, Galindo MJ, Esplugues JV. Efavirenz: What is known about the cellular mechanisms responsible for its adverse effects. Eur J Pharmacol 2017; 812:163-173. [PMID: 28690189 DOI: 10.1016/j.ejphar.2017.07.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 02/08/2023]
Abstract
The HIV infection remains an important health problem worldwide. However, due to the efficacy of combined antiretroviral therapy (cART), it has ceased to be a mortal condition, becoming a chronic disease instead. Efavirenz, the most prescribed non-nucleoside analogue reverse transcriptase inhibitor (NNRTI), has been a key component of cART since its commercialization in 1998. Though still a drug of choice in many countries, its primacy has been challenged by the arrival of newer antiretroviral agents with better toxicity profiles and treatment adherence. The major side effects related to EFV have been widely described in clinical studies, however the mechanisms that participate in their pathogenesis remain largely ununderstood. This review provides an insight into the cellular and molecular mechanisms responsible for the development of the most significant undesired effects induced by efavirenz, both short- and long-term, revealed by in vitro and in vivo experimental pharmacological research. Growing evidence implicates the drug in energy metabolism, mitochondrial function, and other cellular processes involved in stress responses including oxidative stress, inflammation and autophagy.
Collapse
Affiliation(s)
- Nadezda Apostolova
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia-Centro de Investigación Biomédica en Red-Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain.
| | - Ana Blas-Garcia
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia-Centro de Investigación Biomédica en Red-Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain
| | - Maria J Galindo
- Unidad de Enfermedades Infecciosas - Medicina Interna, Hospital Clínico Universitario de Valencia, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia-Centro de Investigación Biomédica en Red-Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain; FISABIO-Hospital Universitario Dr. Peset, Valencia, Spain
| |
Collapse
|
22
|
Sharma D, Turkistani AA, Chang W, Hu C, Xu Z, Chang TKH. Negative Regulation of Human Pregnane X Receptor by MicroRNA-18a-5p: Evidence for Suppression of MicroRNA-18a-5p Expression by Rifampin and Rilpivirine. Mol Pharmacol 2017; 92:48-56. [PMID: 28408657 DOI: 10.1124/mol.116.107003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 04/04/2017] [Indexed: 08/30/2023] Open
Abstract
Small noncoding microRNAs act as post-transcriptional regulators of gene expression involved in diverse biologic functions. Pregnane X receptor (PXR, NR1I2), a member of the superfamily of nuclear receptors, is a transcription factor governing the transport and biotransformation of various drugs and other chemicals. In the present study, we identified a specific microRNA (miR) involved in regulating the expression and functionality of human PXR (hPXR). According to bioinformatics analysis employing three commonly used algorithms (TargetScan, miRanda, and DIANA-microT-CDS), miR-18a-5p was predicted to be the top candidate microRNA regulator of hPXR. Consequently, this microRNA was selected for detailed experimental investigation. As shown in cell-based dual-luciferase reporter gene assays, functional interaction occurred between miR-18a-5p and the microRNA recognition element of miR-18a-5p in the 3'-untranslated region of hPXR mRNA. Transfection of LS180 human colorectal adenocarcinoma cells with an miR-18a-5p mimic decreased hPXR mRNA and protein expression, whereas transfection of LS180 cells with an miR-18a-5p inhibitor increased hPXR mRNA and protein expression. The decrease in hPXR expression by the miR-18a-5p mimic was associated with a reduction in the extent of hPXR target gene (CYP3A4) induction by rifampin and rilpivirine. Treatment of untransfected LS180 cells with either of these hPXR agonists decreased endogenous expression of miR-18a-5p, and this preceded the onset of CYP3A4 induction. In conclusion, miR-18a-5p is a negative regulator of hPXR expression and the hPXR agonists rifampin and rilpivirine are chemical suppressors of miR-18a-5p expression.
Collapse
Affiliation(s)
- Devinder Sharma
- Faculty of Pharmaceutical Sciences, (D.S., A.A.T., C.H., T.K.H.C.), and Food, Nutrition, and Health Program, Faculty of Land and Food Systems (W.C., Z.X.), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Abdullah A Turkistani
- Faculty of Pharmaceutical Sciences, (D.S., A.A.T., C.H., T.K.H.C.), and Food, Nutrition, and Health Program, Faculty of Land and Food Systems (W.C., Z.X.), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Wenjun Chang
- Faculty of Pharmaceutical Sciences, (D.S., A.A.T., C.H., T.K.H.C.), and Food, Nutrition, and Health Program, Faculty of Land and Food Systems (W.C., Z.X.), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine Hu
- Faculty of Pharmaceutical Sciences, (D.S., A.A.T., C.H., T.K.H.C.), and Food, Nutrition, and Health Program, Faculty of Land and Food Systems (W.C., Z.X.), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhaoming Xu
- Faculty of Pharmaceutical Sciences, (D.S., A.A.T., C.H., T.K.H.C.), and Food, Nutrition, and Health Program, Faculty of Land and Food Systems (W.C., Z.X.), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Thomas K H Chang
- Faculty of Pharmaceutical Sciences, (D.S., A.A.T., C.H., T.K.H.C.), and Food, Nutrition, and Health Program, Faculty of Land and Food Systems (W.C., Z.X.), The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
23
|
Piekos S, Pope C, Ferrara A, Zhong XB. Impact of Drug Treatment at Neonatal Ages on Variability of Drug Metabolism and Drug-drug Interactions in Adult Life. ACTA ACUST UNITED AC 2017; 3:1-9. [PMID: 28344923 DOI: 10.1007/s40495-016-0078-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW As the number of patients taking more than one medication concurrently continues to increase, predicting and preventing drug-drug interactions (DDIs) is now more important than ever. Administration of one drug can cause changes in the expression and activity of drug metabolizing enzymes (DMEs) and alter the efficacy or toxicity of other medications that are substrates for these enzymes, resulting in a DDI. In today's medical practice, potential DDIs are evaluated based on the current medications a patient is taking with little regard to drugs the patient has been exposed to in the past. The purpose of this review is to discuss potential impacts of drug treatment at neonatal ages on the variability of drug metabolism and DDIs in adult life. RECENT FINDINGS Existing evidence from the last thirty years has shown that exposure to certain xenobiotics during neonatal life has the potential to persistently alter DME expression through adult life. With recent advancements in the understanding of epigenetic regulation on gene expression, this phenomenon is resurfacing in the scientific community in hopes of defining possible mechanisms. Exposure to compounds that have the ability to bind nuclear receptors and trigger epigenetic modifications at neonatal and pediatric ages may have long-term, if not permanent, consequences on gene expression and DME activity. SUMMARY The information summarized in this review should challenge the way current healthcare providers assess DDI potential and may offer an explanation to the significant interindividual variability in drug metabolism that is observed among patients.
Collapse
Affiliation(s)
- Stephanie Piekos
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA
| | - Chad Pope
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA
| | - Austin Ferrara
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA
| |
Collapse
|
24
|
Garg A, Zhao A, Erickson SL, Mukherjee S, Lau AJ, Alston L, Chang TKH, Mani S, Hirota SA. Pregnane X Receptor Activation Attenuates Inflammation-Associated Intestinal Epithelial Barrier Dysfunction by Inhibiting Cytokine-Induced Myosin Light-Chain Kinase Expression and c-Jun N-Terminal Kinase 1/2 Activation. J Pharmacol Exp Ther 2016; 359:91-101. [PMID: 27440420 PMCID: PMC5034705 DOI: 10.1124/jpet.116.234096] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
The inflammatory bowel diseases (IBDs) are chronic inflammatory disorders with a complex etiology. IBD is thought to arise in genetically susceptible individuals in the context of aberrant interactions with the intestinal microbiota and other environmental risk factors. Recently, the pregnane X receptor (PXR) was identified as a sensor for microbial metabolites, whose activation can regulate the intestinal epithelial barrier. Mutations in NR1I2, the gene that encodes the PXR, have been linked to IBD, and in animal models, PXR deletion leads to barrier dysfunction. In the current study, we sought to assess the mechanism(s) through which the PXR regulates barrier function during inflammation. In Caco-2 intestinal epithelial cell monolayers, tumor necrosis factor-α/interferon-γ exposure disrupted the barrier and triggered zonula occludens-1 relocalization, increased expression of myosin light-chain kinase (MLCK), and activation of c-Jun N-terminal kinase 1/2 (JNK1/2). Activation of the PXR [rifaximin and [[3,5-Bis(1,1-dimethylethyl)-4-hydroxyphenyl]ethenylidene]bis-phosphonic acid tetraethyl ester (SR12813); 10 μM] protected the barrier, an effect that was associated with attenuated MLCK expression and JNK1/2 activation. In vivo, activation of the PXR [pregnenolone 16α-carbonitrile (PCN)] attenuated barrier disruption induced by toll-like receptor 4 activation in wild-type, but not Pxr-/-, mice. Furthermore, PCN treatment protected the barrier in the dextran-sulfate sodium model of experimental colitis, an effect that was associated with reduced expression of mucosal MLCK and phosphorylated JNK1/2. Together, our data suggest that the PXR regulates the intestinal epithelial barrier during inflammation by modulating cytokine-induced MLCK expression and JNK1/2 activation. Thus, targeting the PXR may prove beneficial for the treatment of inflammation-associated barrier disruption in the context of IBD.
Collapse
Affiliation(s)
- Aditya Garg
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Angela Zhao
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Sarah L Erickson
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Subhajit Mukherjee
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Aik Jiang Lau
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Laurie Alston
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Thomas K H Chang
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Sridhar Mani
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Simon A Hirota
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| |
Collapse
|
25
|
Hohmann N, Reinhard R, Schnaidt S, Witt L, Carls A, Burhenne J, Mikus G, Haefeli WE. Treatment with rilpivirine does not alter plasma concentrations of the CYP3A substrates tadalafil and midazolam in humans. J Antimicrob Chemother 2016; 71:2241-2247. [DOI: 10.1093/jac/dkw125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
26
|
Modulation of expression and activity of intestinal multidrug resistance-associated protein 2 by xenobiotics. Toxicol Appl Pharmacol 2016; 303:45-57. [DOI: 10.1016/j.taap.2016.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/29/2016] [Accepted: 05/01/2016] [Indexed: 12/24/2022]
|
27
|
Physiological and pathophysiological factors affecting the expression and activity of the drug transporter MRP2 in intestine. Impact on its function as membrane barrier. Pharmacol Res 2016; 109:32-44. [DOI: 10.1016/j.phrs.2016.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 12/15/2022]
|
28
|
Yousuf S, Natesan S, Enoch IVMV. Chemico-biological interaction of Etravirine and its β-Cyclodextrin complex with macromolecular targets. J Biomol Struct Dyn 2016; 35:1006-1019. [DOI: 10.1080/07391102.2016.1166987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Sameena Yousuf
- Department of Nanosciences & Technology and Department of Chemistry, School of Science and Humanities, Karunya University , Coimbatore 641114, Tamil Nadu, India
| | - Sudha Natesan
- Department of Nanosciences & Technology and Department of Chemistry, School of Science and Humanities, Karunya University , Coimbatore 641114, Tamil Nadu, India
| | - Israel V. M. V. Enoch
- Department of Nanosciences & Technology and Department of Chemistry, School of Science and Humanities, Karunya University , Coimbatore 641114, Tamil Nadu, India
| |
Collapse
|
29
|
Cherian MT, Chai SC, Chen T. Small-molecule modulators of the constitutive androstane receptor. Expert Opin Drug Metab Toxicol 2015; 11:1099-114. [PMID: 25979168 DOI: 10.1517/17425255.2015.1043887] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION The constitutive androstane receptor (CAR) induces drug-metabolizing enzymes for xenobiotic metabolism. AREAS COVERED This review covers recent advances in elucidating the biological functions of CAR and its modulation by a growing number of agonists and inhibitors. EXPERT OPINION Extrapolation of animal CAR function to that of humans should be carefully scrutinized, particularly when rodents are used in evaluating the metabolic profile and carcinogenic properties of clinical drugs and environmental chemicals. Continuous efforts are needed to discover novel CAR inhibitors, with extensive understanding of their inhibitory mechanism, species selectivity, and discriminating power against other xenobiotic sensors.
Collapse
Affiliation(s)
- Milu T Cherian
- Postdoctoral fellow, St. Jude Children's Research Hospital, Department of Chemical Biology and Therapeutics , 262 Danny Thomas Place, Memphis, TN 38105 , USA
| | | | | |
Collapse
|
30
|
Sharma D, Lau AJ, Sherman MA, Chang TKH. Differential activation of human constitutive androstane receptor and its SV23 and SV24 splice variants by rilpivirine and etravirine. Br J Pharmacol 2015; 172:1263-76. [PMID: 25363652 DOI: 10.1111/bph.12997] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/22/2014] [Accepted: 10/27/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Rilpivirine and etravirine are second-generation non-nucleoside reverse transcriptase inhibitors (NNRTIs) indicated for the treatment of HIV/AIDS. The constitutive androstane receptor (CAR) regulates the expression of genes involved in various biological processes, including the transport and biotransformation of drugs. We investigated the effect of rilpivirine and etravirine on the activity of the wild-type human CAR (hCAR-WT) and its hCAR-SV23 and hCAR-SV24 splice variants, and compared it with first-generation NNRTIs (efavirenz, nevirapine, and delavirdine). EXPERIMENTAL APPROACH Receptor activation, ligand-binding domain (LBD) transactivation, and co-activator recruitment were investigated in transiently transfected, NNRTI-treated HepG2 cells. Nuclear translocation of green fluorescent protein-tagged hCAR-WT and CYP2B6 gene expression were assessed in NNRTI-treated human hepatocytes. KEY RESULTS Rilpivirine and etravirine activated hCAR-WT, but not hCAR-SV23 or hCAR-SV24, and without transactivating the LBD or recruiting steroid receptor coactivators SRC-1, SRC-2, or SRC-3. Among the first-generation NNRTIs investigated, only efavirenz activated hCAR-WT, hCAR-SV23, and hCAR-SV24, but none of them transactivated the LBD of these receptors or substantively recruited SRC-1, SRC-2, or SRC-3. Rilpivirine, etravirine, and efavirenz triggered nuclear translocation of hCAR-WT and increased hCAR target gene (CYP2B6) expression. CONCLUSION AND IMPLICATIONS NNRTIs activate hCAR-WT, hCAR-SV23, and hCAR-SV24 in a drug-specific and isoform-selective manner. The activation occurs by a mechanism that does not appear to involve binding to the LBD or recruitment of SRC-1, SRC-2, or SRC-3.
Collapse
Affiliation(s)
- Devinder Sharma
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
31
|
Banerjee M, Chen T. Thiazide-like diuretic drug metolazone activates human pregnane X receptor to induce cytochrome 3A4 and multidrug-resistance protein 1. Biochem Pharmacol 2014; 92:389-402. [PMID: 25181459 PMCID: PMC4252478 DOI: 10.1016/j.bcp.2014.08.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 08/22/2014] [Accepted: 08/25/2014] [Indexed: 01/14/2023]
Abstract
Human pregnane X receptor (hPXR) regulates the expression of drug-metabolizing enzyme cytochrome P450 3A4 (CYP3A4) and drug transporters such as multidrug-resistance protein 1 (MDR1). PXR can be modulated by small molecules, including Federal Drug Administration (FDA)-approved drugs, thus altering drug metabolism and causing drug-drug interactions. To determine the role of FDA-approved drugs in PXR-mediated regulation of drug metabolism and clearance, we screened 1481 FDA-approved small-molecule drugs by using a luciferase reporter assay in HEK293T cells and identified the diuretic drug metolazone as an activator of hPXR. Our data showed that metolazone activated hPXR-mediated expression of CYP3A4 and MDR1 in human hepatocytes and intestine cells and increased CYP3A4 promoter activity in various cell lines. Mammalian two-hybrid assays showed that hPXR recruits its co-activator SRC-1 upon metolazone binding in HepG2 cells, explaining the mechanism of hPXR activation. To understand the role of other commonly-used diuretics in hPXR activation and the structure-activity relationship of metolazone, thiazide and non-thiazide diuretics drugs were also tested but only metolazone activates hPXR. To understand the molecular mechanism, docking studies and mutational analysis were carried out and showed that metolazone binds in the ligand-binding pocket and interacts with mostly hydrophobic amino acid residues. This is the first report showing that metolazone activates hPXR. Because activation of hPXR might cause drug-drug interactions, metolazone should be used with caution for drug treatment in patients undergoing combination therapy.
Collapse
Affiliation(s)
- Monimoy Banerjee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Mail Stop 1000, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Mail Stop 1000, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA.
| |
Collapse
|
32
|
Substantial effect of efavirenz monotherapy on bilirubin levels in healthy volunteers. Curr Ther Res Clin Exp 2014; 76:64-9. [PMID: 25352936 PMCID: PMC4209507 DOI: 10.1016/j.curtheres.2014.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2014] [Indexed: 01/11/2023] Open
Abstract
Background Efavirenz exhibits multiple interactions with drug-metabolizing enzymes and transporters, and for this reason efavirenz-based HIV therapy is associated with altered pharmacokinetics of coadministered drugs. Probably by the same mechanism, efavirenz-based HIV therapy affects the disposition of endogenous compounds, but this effect is difficult to directly link with efavirenz because it is used in combination with other drugs. Objectives To explore the effect of efavirenz monotherapy on biochemical laboratory values in a clinical trial of healthy volunteers. Methods Men and women (aged 18–49 years) with body mass index ≤32 who were assessed to be healthy based on medical history, physical examination, and standard laboratory screening received a single (600 mg) and multiple doses (600 mg/d for 17 days) of efavirenz orally. This trial was designed to determine the pharmacokinetics and drug interactions of efavirenz. As part of this study, analysis of serum chemistries that were measured at study entry (screening) and 1 week after completion of the multiple dose study (exit) is reported. Results Data from 60 subjects who fully completed and 13 subjects who partially completed the study are presented. Total bilirubin was substantially reduced at exit (by ~30%, with large intersubject variability) compared with screening values (P < 0.0001). The percent changes were in part explained by the intersubject differences in baseline total bilirubin because there was a significant correlation between baseline (screening) values and percent change at exit (r = 0.50; P < 0.0001). Hemoglobin and absolute neutropenia were also substantially decreased at exit compared with screening, but this may be due to intensive blood sampling rather than direct effect of efavirenz on these parameters. No significant correlation was found between percent change in hemoglobin versus percent change in bilirubin, indicating the effect of efavirenz on bilirubin is independent of its effects on hemoglobin. Conclusions Efavirenz monotherapy significantly lowers plasma total bilirubin concentration in healthy volunteers independent of its effect on hemoglobin, probably through its effects on bilirubin metabolism and transport (uptake and efflux). These findings help explain reversal by efavirenz of hyperbilirubinemia induction observed by some protease inhibitor antiretroviral drugs (eg, atazanavir). Besides its well-documented role on drug interactions, efavirenz may alter the disposition of endogenous compounds relevant in physiologic homeostasis through its interaction with drug metabolizing enzymes and/or drug transporters. ClinicalTrials.gov identifier: NCT00668395.
Collapse
|
33
|
Human biotransformation of the nonnucleoside reverse transcriptase inhibitor rilpivirine and a cross-species metabolism comparison. Antimicrob Agents Chemother 2013; 57:5067-79. [PMID: 23917319 DOI: 10.1128/aac.01401-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Rilpivirine is a nonnucleoside reverse transcriptase inhibitor used to treat HIV-1. In the present study, the pathways responsible for the biotransformation of rilpivirine were defined. Using human liver microsomes, the formation of two mono- and two dioxygenated metabolites were detected via ultra high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Mass spectral analysis of the products suggested that these metabolites resulted from oxygenation of the 2,6-dimethylphenyl ring and methyl groups of rilpivirine. Chemical inhibition studies and cDNA-expressed cytochrome P450 (CYP) assays indicated that oxygenations were catalyzed primarily by CYP3A4 and CYP3A5. Glucuronide conjugates of rilpivirine and a monomethylhydroxylated metabolite of rilpivirine were also detected and were found to be formed by UDP-glucuronosyltransferases (UGTs) UGT1A4 and UGT1A1, respectively. All metabolites that were identified in vitro were detectable in vivo. Further, targeted UHPLC-MS/MS-based in vivo metabolomics screening revealed that rilpivirine treatment versus efavirenz treatment may result in differential levels of endogenous metabolites, including tyrosine, homocysteine, and adenosine. Rilpivirine biotransformation was also assessed across species using liver microsomes isolated from a range of mammals, and the metabolite profile identified using human liver microsomes was largely conserved for both oxidative and glucuronide metabolite formation. These studies provide novel insight into the metabolism of rilpivirine and the potential differential effects of rilpivirine- and efavirenz-containing antiretroviral regimens on the endogenous metabolome.
Collapse
|
34
|
Banerjee M, Chen T. Differential regulation of CYP3A4 promoter activity by a new class of natural product derivatives binding to pregnane X receptor. Biochem Pharmacol 2013; 86:824-35. [PMID: 23928187 DOI: 10.1016/j.bcp.2013.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 01/25/2023]
Abstract
The pregnane X receptor (PXR) regulates drug metabolism by regulating the expression of drug-metabolizing enzymes such as cytochrome P450 3A4 (CYP3A4), which is involved in the metabolism of >50% of clinically prescribed drugs. The activity of PXR can be controlled by the binding of small molecule agonists or antagonists. Because of its unique ligand binding pocket, PXR binds promiscuously to structurally diverse chemicals. To study the structure-activity relationship, novel modulators for PXR are needed. Here we report the virtual screening of ∼25,000 natural product derivatives from the ZINC database using the Molecular Operating Environment docking software tool against the PXR-rifampicin complex X-ray crystal structure. Our screening resulted in identification of compounds based on the lowest S score, which measures Gibbs free energy. Interestingly, we found that the compounds that bind directly to PXR, as revealed in an intrinsic tryptophan fluorescence assay, modulate CYP3A4 promoter activity differentially in HepG2 cells. Mutational analysis and docking studies showed that these compounds bind broadly in the ligand binding pocket but interact with different amino acid residues. We further investigated the mechanism of binding by analyzing the functional groups that are important for distinguishing agonists from antagonists. The approach we used to identify novel modulators that bind to PXR can be useful for finding novel modulators of PXR.
Collapse
Affiliation(s)
- Monimoy Banerjee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | |
Collapse
|