1
|
Leow JWH, Chan ECY. CYP2J2-mediated metabolism of arachidonic acid in heart: A review of its kinetics, inhibition and role in heart rhythm control. Pharmacol Ther 2024; 258:108637. [PMID: 38521247 DOI: 10.1016/j.pharmthera.2024.108637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Cytochrome P450 2 J2 (CYP2J2) is primarily expressed extrahepatically and is the predominant epoxygenase in human cardiac tissues. This highlights its key role in the metabolism of endogenous substrates. Significant scientific interest lies in cardiac CYP2J2 metabolism of arachidonic acid (AA), an omega-6 polyunsaturated fatty acid, to regioisomeric bioactive epoxyeicosatrienoic acid (EET) metabolites that show cardioprotective effects including regulation of cardiac electrophysiology. From an in vitro perspective, the accurate characterization of the kinetics of CYP2J2 metabolism of AA including its inhibition and inactivation by drugs could be useful in facilitating in vitro-in vivo extrapolations to predict drug-AA interactions in drug discovery and development. In this review, background information on the structure, regulation and expression of CYP2J2 in human heart is presented alongside AA and EETs as its endogenous substrate and metabolites. The in vitro and in vivo implications of the kinetics of this endogenous metabolic pathway as well as its perturbation via inhibition and inactivation by drugs are elaborated. Additionally, the role of CYP2J2-mediated metabolism of AA to EETs in cardiac electrophysiology will be expounded.
Collapse
Affiliation(s)
- Jacqueline Wen Hui Leow
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
2
|
Zhang S, Wang T, Feng Y, Li F, Qu A, Guan X, Wang H, Xu D. Pregnenolone 16α-carbonitrile negatively regulates hippocampal cytochrome P450 enzymes and ameliorates phenytoin-induced hippocampal neurotoxicity. J Pharm Anal 2023; 13:1510-1525. [PMID: 38223454 PMCID: PMC10785155 DOI: 10.1016/j.jpha.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 01/16/2024] Open
Abstract
The central nervous system is susceptible to the modulation of various neurophysiological processes by the cytochrome P450 enzyme (CYP), which plays a crucial role in the metabolism of neurosteroids. The antiepileptic drug phenytoin (PHT) has been observed to induce neuronal side effects in patients, which could be attributed to its induction of CYP expression and testosterone (TES) metabolism in the hippocampus. While pregnane X receptor (PXR) is widely known for its regulatory function of CYPs in the liver, we have discovered that the treatment of mice with pregnenolone 16α-carbonitrile (PCN), a PXR agonist, has differential effects on CYP expression in the liver and hippocampus. Specifically, the PCN treatment resulted in the induction of cytochrome P450, family 3, subfamily a, polypeptide 11 (CYP3A11), and CYP2B10 expression in the liver, while suppressing their expression in the hippocampus. Functionally, the PCN treatment protected mice from PHT-induced hippocampal nerve injury, which was accompanied by the inhibition of TES metabolism in the hippocampus. Mechanistically, we found that the inhibition of hippocampal CYP expression and attenuation of PHT-induced neurotoxicity by PCN were glucocorticoid receptor dependent, rather than PXR independent, as demonstrated by genetic and pharmacological models. In conclusion, our study provides evidence that PCN can negatively regulate hippocampal CYP expression and attenuate PHT-induced hippocampal neurotoxicity independently of PXR. Our findings suggest that glucocorticoids may be a potential therapeutic strategy for managing the neuronal side effects of PHT.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| | - Tingting Wang
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| | - Ye Feng
- Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fei Li
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100069, China
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, 100069, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| | - Dan Xu
- Department of Obstetric, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
3
|
Rakateli L, Huchzermeier R, van der Vorst EPC. AhR, PXR and CAR: From Xenobiotic Receptors to Metabolic Sensors. Cells 2023; 12:2752. [PMID: 38067179 PMCID: PMC10705969 DOI: 10.3390/cells12232752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Traditionally, xenobiotic receptors are known for their role in chemical sensing and detoxification, as receptor activation regulates the expression of various key enzymes and receptors. However, recent studies have highlighted that xenobiotic receptors also play a key role in the regulation of lipid metabolism and therefore function also as metabolic sensors. Since dyslipidemia is a major risk factor for various cardiometabolic diseases, like atherosclerosis and non-alcoholic fatty liver disease, it is of major importance to understand the molecular mechanisms that are regulated by xenobiotic receptors. In this review, three major xenobiotic receptors will be discussed, being the aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Specifically, this review will focus on recent insights into the metabolic functions of these receptors, especially in the field of lipid metabolism and the associated dyslipidemia.
Collapse
Affiliation(s)
- Leonida Rakateli
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (L.R.); (R.H.)
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Rosanna Huchzermeier
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (L.R.); (R.H.)
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (L.R.); (R.H.)
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
4
|
Jin J, Zhong XB. Epigenetic Mechanisms Contribute to Intraindividual Variations of Drug Metabolism Mediated by Cytochrome P450 Enzymes. Drug Metab Dispos 2023; 51:672-684. [PMID: 36973001 PMCID: PMC10197210 DOI: 10.1124/dmd.122.001007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Significant interindividual and intraindividual variations on cytochrome P450 (CYP)-mediated drug metabolism exist in the general population globally. Genetic polymorphisms are one of the major contribution factors for interindividual variations, but epigenetic mechanisms mainly contribute to intraindividual variations, including DNA methylation, histone modifications, microRNAs, and long non-coding RNAs. The current review provides analysis of advanced knowledge in the last decade on contributions of epigenetic mechanisms to intraindividual variations on CYP-mediated drug metabolism in several situations, including (1) ontogeny, the developmental changes of CYP expression in individuals from neonates to adults; (2) increased activities of CYP enzymes induced by drug treatment; (3) increased activities of CYP enzymes in adult ages induced by drug treatment at neonate ages; and (4) decreased activities of CYP enzymes in individuals with drug-induced liver injury (DILI). Furthermore, current challenges, knowledge gaps, and future perspective of the epigenetic mechanisms in development of CYP pharmacoepigenetics are discussed. In conclusion, epigenetic mechanisms have been proven to contribute to intraindividual variations of drug metabolism mediated by CYP enzymes in age development, drug induction, and DILI conditions. The knowledge has helped understanding how intraindividual variation are generated. Future studies are needed to develop CYP-based pharmacoepigenetics to guide clinical applications for precision medicine with improved therapeutic efficacy and reduced risk of adverse drug reactions and toxicity. SIGNIFICANCE STATEMENT: Understanding epigenetic mechanisms in contribution to intraindividual variations of CYP-mediated drug metabolism may help to develop CYP-based pharmacoepigenetics for precision medicine to improve therapeutic efficacy and reduce adverse drug reactions and toxicity for drugs metabolized by CYP enzymes.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
5
|
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
|
6
|
McCune JS, Navarro SL, Baker KS, Risler LJ, Phillips BR, Randolph TW, Shireman L, Schoch G, Deeg HJ, Zhang Y, Men A, Maton L, Huitema ADR. Prediction of Busulfan Clearance by Predose Plasma Metabolomic Profiling. Clin Pharmacol Ther 2023; 113:370-379. [PMID: 36369996 PMCID: PMC9888309 DOI: 10.1002/cpt.2794] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Intravenous busulfan doses are often personalized to a target plasma exposure (targeted busulfan) using an individual's busulfan clearance (BuCL). We evaluated whether BuCL could be predicted by a predose plasma panel of 841 endogenous metabolomic compounds (EMCs). In this prospective cohort of 132 hematopoietic cell transplantation (HCT) patients, all had samples collected immediately before busulfan administration (preBU) and 96 had samples collected 2 weeks before busulfan (2-week-preBU). BuCL was significantly associated with 37 EMCs after univariate linear regression analysis and controlling for false discovery (< 0.05) in the 132 preBU samples. In parallel, with preBU samples, we included all 841 EMCs in a least absolute shrinkage and selection operator-penalized regression which selected 13 EMCs as predominantly associated with BuCL. Then, we constructed a prediction model by estimating coefficients for these 13 EMCs, along with sex, using ordinary least-squares. When the resulting linear prediction model was applied to the 2-week-preBU samples, it explained 40% of the variation in BuCL (adjusted R2 = 0.40). Pathway enrichment analysis revealed 18 pathways associated with BuCL. Lysine degradation followed by steroid biosynthesis, which aligned with the univariate analysis, were the top two pathways. BuCL can be predicted before busulfan administration with a linear regression model of 13 EMCs. This pharmacometabolomics method should be prioritized over use of a busulfan test dose or pharmacogenomics to guide busulfan dosing. These results highlight the potential of pharmacometabolomics as a precision medicine tool to improve or replace pharmacokinetics to personalize busulfan doses.
Collapse
Affiliation(s)
- Jeannine S. McCune
- City of Hope, Department of Hematologic Malignancies Translational Sciences, Duarte, California (CA), 91010, United States of America (USA)
| | - Sandi L. Navarro
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (WA), 98109, USA
| | - K. Scott Baker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (WA), 98109, USA,Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Linda J. Risler
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Brian R. Phillips
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Timothy W. Randolph
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (WA), 98109, USA
| | - Laura Shireman
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Gary Schoch
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (WA), 98109, USA
| | - H. Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (WA), 98109, USA,Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Yuzheng Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (WA), 98109, USA
| | - Alex Men
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Loes Maton
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Alwin D. R. Huitema
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands,Department of Pharmacology, Princes Maxima & Pharmacology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands,Department of Clinical Pharmacy, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
7
|
Meirinho S, Rodrigues M, Fortuna A, Falcão A, Alves G. Study of the metabolic stability profiles of perampanel, rufinamide and stiripentol and prediction of drug interactions using HepaRG cells as an in vitro human model. Toxicol In Vitro 2022; 82:105389. [PMID: 35597399 DOI: 10.1016/j.tiv.2022.105389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 04/11/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
New-generation antiepileptic drugs as perampanel, rufinamide and stiripentol emerged as alternatives in chronic epilepsy polytherapy. Hence, their metabolic stability and potential involvement in relevant drug-drug interactions (DDI) are of great clinical interest, being HepaRG cells herein used as an in vitro human model. To characterize their metabolic stability profiles, HepaRG cells were incubated with perampanel (1 μM), rufinamide (100 μM) or stiripentol (5 μM) for 12-h. HepaRG cells, pretreated with known CYP450 isoenzymes inducers (rifampicin, phenytoin, phenobarbital, omeprazole and carbamazepine), were also incubated with perampanel, rufinamide or stiripentol to assess possible DDI mediated by CYP induction. Results suggest a considerable decrease in perampanel and stiripentol concentrations over 12-h; contrary, rufinamide concentrations did not variated. Cells pretreatment with all inducers significantly decreased stiripentol concentrations (between 20.3% and 31.9%), suggesting a considerable potential for DDI. Rufinamide concentrations only decreased when preincubated with rifampicin and with the highest tested concentrations of the remaining inducers. Perampanel levels decreased with rifampicin, carbamazepine and phenobarbital, supporting the involvement of CYP3A4-mediated metabolism. Besides relevant information concerning the metabolic stability profile and potential DDIs of the new antiepileptics here studied, it was also reinforced the HepaRG cells suitability as a reliable in vitro model to foresee in vivo metabolism in humans.
Collapse
Affiliation(s)
- Sara Meirinho
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Márcio Rodrigues
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CPIRN-UDI-IPG - Center for Potential and Innovation of Natural Resources, Research Unit for Inland Development, Polytechnic Institute of Guarda, Av. Dr. Francisco de Sá Carneiro, 6300-559 Guarda, Portugal
| | - Ana Fortuna
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Pólo das Ciências e da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Amílcar Falcão
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Pólo das Ciências e da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Gilberto Alves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
8
|
Rey-Bedon C, Banik P, Gokaltun A, Hofheinz O, Yarmush ML, Uygun MK, Usta OB. CYP450 drug inducibility in NAFLD via an in vitro hepatic model: Understanding drug-drug interactions in the fatty liver. Biomed Pharmacother 2022; 146:112377. [PMID: 35062050 PMCID: PMC8792443 DOI: 10.1016/j.biopha.2021.112377] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/20/2022] Open
Abstract
Drug-drug-interactions (DDIs) occur when a drug alters the metabolic rate, efficacy, and toxicity of concurrently used drugs. While almost 1 in 4 adults now use at least 3 concurrent prescription drugs in the United States, the Non-alcoholic fatty liver disease (NAFLD) prevalence has also risen over 25%. The effect of NALFD on DDIs is largely unknown. NAFLD is characterized by lipid vesicle accumulation in the liver, which can progress to severe steatohepatitis (NASH), fibrosis, cirrhosis, and hepatic carcinoma. The CYP450 enzyme family dysregulation in NAFLD, which might already alter the efficacy and toxicity of drugs, has been partially characterized. Nevertheless, the drug-induced dysregulation of CYP450 enzymes has not been studied in the fatty liver. These changes in enzymatic inducibility during NAFLD, when taking concurrent drugs, could cause unexpected fatalities through inadvertent DDIs. We have, thus, developed an in vitro model to investigate the CYP450 transcriptional regulation in NAFLD. Specifically, we cultured primary human hepatocytes in a medium containing free fatty acids, high glucose, and insulin for seven days. These cultures displayed intracellular macro-steatosis after 5 days and cytokine secretion resembling NAFLD patients. We further verified the model's dysregulation in the transcription of key CYP450 enzymes. We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. We, thus, conclude that the fatty liver could cause aggravated drug-drug interactions in NAFLD or NASH patients related to CYP2B6 and CYP2C9 enzymes.
Collapse
Affiliation(s)
- Camilo Rey-Bedon
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, United States
| | - Peony Banik
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States
| | - Aslihan Gokaltun
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States; Department of Chemical Engineering, Hacettepe University, 06532 Beytepe, Ankara, Turkey
| | - O Hofheinz
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States
| | - Martin L Yarmush
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, United States; Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, United States
| | - M Korkut Uygun
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States
| | - O Berk Usta
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States.
| |
Collapse
|
9
|
Frequencies of CYP2B6 ∗4, ∗5, and ∗6 Alleles within an Iranian Population (Mazandaran). Genet Res (Camb) 2021; 2021:8703812. [PMID: 34949964 PMCID: PMC8660211 DOI: 10.1155/2021/8703812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/15/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023] Open
Abstract
Background The human CYP2B subfamily consists of one functional gene (CYP2B6) and one pseudogene (CYP2B7P). Cytochrome P450 2B6 (CYP2B6) is a highly polymorphic enzyme that shows marked interindividual and interethnic variations. Currently, 38 alleles have been described, and some of the allelic variants have been associated with low enzyme activity. The aim of this study was to investigate the frequencies of CYP2B6∗4, CYP2B6∗5, and CYP2B6∗6 alleles in the Mazani ethnic group among Iranian Population. Methods The study was conducted in 289 unrelated healthy volunteers. DNA was extracted from peripheral blood and analyzed by the PCR-RFLP protocol. The PCR product was digested with restriction enzymes and then separated using agarose gel electrophoresis. Results The frequency of CYP2B6∗4, CYP2B6∗5, and CYP2B6∗6 in this study was 34.60%, 7.26%, and 34.54%, respectively. Conclusion The frequency of the CYP2B6∗4 allele in the Mazani ethnic group was much higher (34.60%) than other population. The frequency of CYP2B6∗6 (34.54%) also was higher than its frequency in other previously reported population. But the frequency of CYP2B6∗5 in this study was lower than expected. These results will be useful in understanding the ethnic diversity in Iranian population and offer a preliminary basis for more rational use of drugs that are substrates for CYP2B6 in this population.
Collapse
|
10
|
Li Z, Li L, Heyward S, Men S, Xu M, Sueyoshi T, Wang H. Phenobarbital Induces SLC13A5 Expression through Activation of PXR but Not CAR in Human Primary Hepatocytes. Cells 2021; 10:cells10123381. [PMID: 34943889 PMCID: PMC8699749 DOI: 10.3390/cells10123381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 02/05/2023] Open
Abstract
Phenobarbital (PB), a widely used antiepileptic drug, is known to upregulate the expression of numerous drug-metabolizing enzymes and transporters in the liver primarily via activation of the constitutive androstane receptor (CAR, NR1I3). The solute carrier family 13 member 5 (SLC13A5), a sodium-coupled citrate transporter, plays an important role in intracellular citrate homeostasis that is associated with a number of metabolic syndromes and neurological disorders. Here, we show that PB markedly elevates the expression of SLC13A5 through a pregnane X receptor (PXR)-dependent but CAR-independent signaling pathway. In human primary hepatocytes, the mRNA and protein expression of SLC13A5 was robustly induced by PB treatment, while genetic knockdown or pharmacological inhibition of PXR significantly attenuated this induction. Utilizing genetically modified HepaRG cells, we found that PB induces SLC13A5 expression in both wild type and CAR-knockout HepaRG cells, whereas such induction was fully abolished in the PXR-knockout HepaRG cells. Mechanistically, we identified and functionally characterized three enhancer modules located upstream from the transcription start site or introns of the SLC13A5 gene that are associated with the regulation of PXR-mediated SLC13A5 induction. Moreover, metformin, a deactivator of PXR, dramatically suppressed PB-mediated induction of hepatic SLC13A5 as well as its activation of the SLC13A5 luciferase reporter activity via PXR. Collectively, these data reveal PB as a potent inducer of SLC13A5 through the activation of PXR but not CAR in human primary hepatocytes.
Collapse
Affiliation(s)
- Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA; (Z.L.); (L.L.); (S.M.)
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA; (Z.L.); (L.L.); (S.M.)
| | - Scott Heyward
- BioIVT, 1450 S Rolling Road, Halethorpe, MD 21227, USA;
| | - Shuaiqian Men
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA; (Z.L.); (L.L.); (S.M.)
| | - Meishu Xu
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Tatsuya Sueyoshi
- Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA;
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA; (Z.L.); (L.L.); (S.M.)
- Correspondence: ; Tel.: +1-410-706-1280
| |
Collapse
|
11
|
Preiss LC, Liu R, Hewitt P, Thompson D, Georgi K, Badolo L, Lauschke VM, Petersson C. Deconvolution of Cytochrome P450 Induction Mechanisms in HepaRG Nuclear Hormone Receptor Knockout Cells. Drug Metab Dispos 2021; 49:668-678. [PMID: 34035124 DOI: 10.1124/dmd.120.000333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Pregnane X receptor (PXR), constitutive androstane receptor (CAR), and PXR/CAR knockout (KO) HepaRG cells, as well as a PXR reporter gene assay, were used to investigate the mechanism of CYP3A4 and CYP2B6 induction by prototypical substrates and a group of compounds from the Merck KGaA oncology drug discovery pipeline. The basal and inducible gene expression of CYP3A4 and CYP2B6 of nuclear hormone receptor (NHR) KO HepaRG relative to control HepaRG was characterized. The basal expression of CYP3A4 was markedly higher in the PXR (10-fold) and CAR (11-fold) KO cell lines compared with control HepaRG, whereas inducibility was substantially lower. Inversely, basal expression of CYP3A4 in PXR/CAR double KO (dKO) was low (10-fold reduction). Basal CYP2B6 expression was high in PXR KO (9-fold) cells which showed low inducibility, whereas the basal expression remained unchanged in CAR and dKO cell lines compared with control cells. Most of the test compounds induced CYP3A4 and CYP2B6 via PXR and, to a lesser extent, via CAR. Furthermore, other non-NHR-driven induction mechanisms were implicated, either alone or in addition to NHRs. Notably, 5 of the 16 compounds (31%) that were PXR inducers in HepaRG did not activate PXR in the reporter gene assay, illustrating the limitations of this system. This study indicates that HepaRG is a highly sensitive system fit for early screening of cytochrome P450 (P450) induction in drug discovery. Furthermore, it shows the applicability of HepaRG NHR KO cells as tools to deconvolute mechanisms of P450 induction using novel compounds representative for oncology drug discovery. SIGNIFICANCE STATEMENT: This work describes the identification of induction mechanisms of CYP3A4 and CYP2B6 for an assembly of oncology drug candidates using HepaRG nuclear hormone receptor knockout and displays its advantages compared to a pregnane X receptor reporter gene assay. With this study, risk assessment of drug candidates in early drug development can be improved.
Collapse
Affiliation(s)
- Lena C Preiss
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Ruoqi Liu
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Philip Hewitt
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - David Thompson
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Katrin Georgi
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Lassina Badolo
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Volker M Lauschke
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Carl Petersson
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| |
Collapse
|
12
|
Niu C, Smith B, Lai Y. Transporter Gene Regulation in Sandwich Cultured Human Hepatocytes Through the Activation of Constitutive Androstane Receptor (CAR) or Aryl Hydrocarbon Receptor (AhR). Front Pharmacol 2021; 11:620197. [PMID: 33551819 PMCID: PMC7859440 DOI: 10.3389/fphar.2020.620197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/15/2020] [Indexed: 01/11/2023] Open
Abstract
The induction potentials of ligand-activated nuclear receptors on metabolizing enzyme genes are routinely tested for new chemical entities. However, regulations of drug transporter genes by the nuclear receptor ligands are underappreciated, especially in differentiated human hepatocyte cultures. In this study, gene induction by the ligands of constitutive androstane receptor (CAR) and aryl hydrocarbon receptor (AhR) was characterized in sandwich-cultured human hepatocytes (SCHH) from multiple donors. The cells were treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), omeprazole (OP), 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime (CITCO) and phenobarbital (PB) for three days. RNA samples were analyzed by qRT-PCR method. As expected, CITCO, the direct activator, and PB, the indirect activator of CAR, induced CYP3A4 (31 and 40-fold), CYP2B6 (24 and 28-fold) and UGT1A1 (2.9 and 4.2-fold), respectively. Conversely, TCDD and OP, the activators of AhR, induced CYP1A1 (38 and 37-fold), and UGT1A1 (4.3 and 5.0-fold), respectively. In addition, OP but not TCDD induced CY3A4 by about 61-fold. Twenty-four hepatic drug transporter genes were characterized, and of those, SLC51B was induced the most by PB and OP by about 3.3 and 6.5 fold, respectively. Marginal inductions (about 2-fold) of SLC47A1 and SLCO4C1 genes by PB, and ABCG2 gene by TCDD were observed. In contrast, SLC10A1 gene was suppressed about 2-fold by TCDD and CITCO. While clinical relevance of SLC51B gene induction or SLC10A1 gene suppression warrants further investigation, the results verified that the assessment of transporter gene inductions are not required for new drug entities, when a drug does not remarkably induce metabolizing enzyme genes by CAR and AhR activation.
Collapse
Affiliation(s)
- Congrong Niu
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA, United States
| | - Bill Smith
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA, United States
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA, United States
| |
Collapse
|
13
|
Oliviero F, Lukowicz C, Boussadia B, Forner-Piquer I, Pascussi JM, Marchi N, Mselli-Lakhal L. Constitutive Androstane Receptor: A Peripheral and a Neurovascular Stress or Environmental Sensor. Cells 2020; 9:E2426. [PMID: 33171992 PMCID: PMC7694609 DOI: 10.3390/cells9112426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
Xenobiotic nuclear receptors (NR) are intracellular players involved in an increasing number of physiological processes. Examined and characterized in peripheral organs where they govern metabolic, transport and detoxification mechanisms, accumulating data suggest a functional expression of specific NR at the neurovascular unit (NVU). Here, we focus on the Constitutive Androstane Receptor (CAR), expressed in detoxifying organs such as the liver, intestines and kidneys. By direct and indirect activation, CAR is implicated in hepatic detoxification of xenobiotics, environmental contaminants, and endogenous molecules (bilirubin, bile acids). Importantly, CAR participates in physiological stress adaptation responses, hormonal and energy homeostasis due to glucose and lipid sensing. We next analyze the emerging evidence supporting a role of CAR in NVU cells including the blood-brain barrier (BBB), a key vascular interface regulating communications between the brain and the periphery. We address the emerging concept of how CAR may regulate specific P450 cytochromes at the NVU and the associated relevance to brain diseases. A clear understanding of how CAR engages during pathological conditions could enable new mechanistic, and perhaps pharmacological, entry-points within a peripheral-brain axis.
Collapse
Affiliation(s)
- Fabiana Oliviero
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| | - Céline Lukowicz
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| | - Badreddine Boussadia
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Isabel Forner-Piquer
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Jean-Marc Pascussi
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Laila Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| |
Collapse
|
14
|
Loerracher AK, Braunbeck T. Inducibility of cytochrome P450-mediated 7-methoxycoumarin-O-demethylase activity in zebrafish (Danio rerio) embryos. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 225:105540. [PMID: 32569997 DOI: 10.1016/j.aquatox.2020.105540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/31/2020] [Accepted: 06/07/2020] [Indexed: 06/11/2023]
Abstract
The zebrafish (Danio rerio) embryo has increasingly been used as an alternative model in human and environmental toxicology. Since the cytochrome P450 (CYP) system is of fundamental importance for the understanding and correct interpretation of the outcome of toxicological studies, constitutive and xenobiotic-induced 7-methoxycoumarin-O-demethylase (MCOD), i.e. 'mammalian CYP2-like', activities were monitored in vivo in zebrafish embryos via confocal laser scanning microscopy. In order to elucidate molecular mechanisms underlying the MCOD induction, dose-dependent effects of the prototypical CYP inducers β-naphthoflavone (aryl hydrocarbon receptor (AhR) agonist), rifampicin (pregnane X receptor (PXR) agonist), carbamazepine and phenobarbital (constitutive androstane receptor (CAR) agonists) were analyzed in zebrafish embryos of varying age. Starting from 36 h of age, all embryonic stages of zebrafish could be shown to have constitutive MCOD activity, albeit with spatial variation and at distinct levels. Whereas carbamazepine, phenobarbital and rifampicin had no effect on in vivo MCOD activity in 96 h old zebrafish embryos, the model aryl hydrocarbon receptor agonist β-naphthoflavone significantly induced MCOD activity in 96 h old zebrafish embryos at 46-734 nM, however, without a clear concentration-effect relationship. Induction of MCOD activity by β-naphthoflavone gradually decreased with progression of embryonic development. By in vivo characterization of constitutive and xenobiotic-induced MCOD activity patterns in 36, 60, 84 and 108 h old zebrafish embryos, this decrease could primarily be attributed to an age-related decline in the induction of MCOD activity in the cardiovascular system. Results of this study provide novel insights into the mechanism and extent, by which specific CYP activities in early life-stages of zebrafish can be influenced by exposure to xenobiotics. The study thus lends further support to the view that zebrafish embryos- at least from an age of 36 h - have an elaborate and inducible biotransformation system.
Collapse
Affiliation(s)
- Ann-Kathrin Loerracher
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120, Heidelberg, Germany.
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120, Heidelberg, Germany
| |
Collapse
|
15
|
Elmeliegy M, Vourvahis M, Guo C, Wang DD. Effect of P-glycoprotein (P-gp) Inducers on Exposure of P-gp Substrates: Review of Clinical Drug-Drug Interaction Studies. Clin Pharmacokinet 2020; 59:699-714. [PMID: 32052379 PMCID: PMC7292822 DOI: 10.1007/s40262-020-00867-1] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding transporter-mediated drug-drug interactions (DDIs) for investigational agents is important during drug development to assess DDI liability, its clinical relevance, and to determine appropriate DDI management strategies. P-glycoprotein (P-gp) is an efflux transporter that influences the pharmacokinetics (PK) of various compounds. Assessing transporter induction in vitro is challenging and is not always predictive of in vivo effects, and hence there is a need to consider clinical DDI studies; however, there is no clear guidance on when clinical evaluation of transporter induction is required. Furthermore, there is no proposed list of index transporter inducers to be used in clinical studies. This review evaluated DDI studies with known P-gp inducers to better understand the mechanism and site of P-gp induction, as well as the magnitude of induction effect on the exposure of P-gp substrates. Our review indicates that P-gp and cytochrome P450 (CYP450) enzymes are co-regulated via the pregnane xenobiotic receptor (PXR) and the constitutive androstane receptor (CAR). The magnitude of the decrease in substrate drug exposure by P-gp induction is generally less than that of CYP3A. Most P-gp inducers reduced total bioavailability with a minor impact on renal clearance, despite known expression of P-gp at the apical membrane of the kidney proximal tubules. Rifampin is the most potent P-gp inducer, resulting in an average reduction in substrate exposure ranging between 20 and 67%. For other inducers, the reduction in P-gp substrate exposure ranged from 12 to 42%. A lower reduction in exposure of the P-gp substrate was observed with a lower dose of the inducer and/or if the administration of the inducer and substrate was simultaneous, i.e. not staggered. These findings suggest that clinical evaluation of the impact of P-gp inducers on the PK of investigational agents that are substrates for P-gp might be warranted only for compounds with a relatively steep exposure-efficacy relationship.
Collapse
Affiliation(s)
- Mohamed Elmeliegy
- Clinical Pharmacology, Global Product Development, Pfizer Inc., 10555 Science Center Dr., San Diego, CA, 92121, USA.
| | - Manoli Vourvahis
- Clinical Pharmacology, Global Product Development, Pfizer Inc., New York, NY, USA
| | - Cen Guo
- Clinical Pharmacology, Global Product Development, Pfizer Inc., 10555 Science Center Dr., San Diego, CA, 92121, USA
| | - Diane D Wang
- Clinical Pharmacology, Global Product Development, Pfizer Inc., 10555 Science Center Dr., San Diego, CA, 92121, USA
| |
Collapse
|
16
|
Mertansine Inhibits mRNA Expression and Enzyme Activities of Cytochrome P450s and Uridine 5′-Diphospho-Glucuronosyltransferases in Human Hepatocytes and Liver Microsomes. Pharmaceutics 2020; 12:pharmaceutics12030220. [PMID: 32131538 PMCID: PMC7150891 DOI: 10.3390/pharmaceutics12030220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 11/16/2022] Open
Abstract
Mertansine, a tubulin inhibitor, is used as the cytotoxic component of antibody–drug conjugates (ADCs) for cancer therapy. The effects of mertansine on uridine 5′-diphospho-glucuronosyltransferase (UGT) activities in human liver microsomes and its effects on the mRNA expression of cytochrome P450s (CYPs) and UGTs in human hepatocytes were evaluated to assess the potential for drug–drug interactions (DDIs). Mertansine potently inhibited UGT1A1-catalyzed SN-38 glucuronidation, UGT1A3-catalyzed chenodeoxycholic acid 24-acyl-β-glucuronidation, and UGT1A4-catalyzed trifluoperazine N-β-d-glucuronidation, with Ki values of 13.5 µM, 4.3 µM, and 21.2 µM, respectively, but no inhibition of UGT1A6, UGT1A9, and UGT2B7 enzyme activities was observed in human liver microsomes. A 48 h treatment of mertansine (1.25–2500 nM) in human hepatocytes resulted in the dose-dependent suppression of mRNA levels of CYP1A2, CYP2B6, CYP3A4, CYP2C8, CYP2C9, CYP2C19, UGT1A1, and UGT1A9, with IC50 values of 93.7 ± 109.1, 36.8 ± 18.3, 160.6 ± 167.4, 32.1 ± 14.9, 578.4 ± 452.0, 539.5 ± 233.4, 856.7 ± 781.9, and 54.1 ± 29.1 nM, respectively, and decreased the activities of CYP1A2-mediated phenacetin O-deethylase, CYP2B6-mediated bupropion hydroxylase, and CYP3A4-mediated midazolam 1′-hydroxylase. These in vitro DDI potentials of mertansine with CYP1A2, CYP2B6, CYP2C8/9/19, CYP3A4, UGT1A1, and UGT1A9 substrates suggest that it is necessary to carefully characterize the DDI potentials of ADC candidates with mertansine as a payload in the clinic.
Collapse
|
17
|
Fashe M, Hashiguchi T, Negishi M, Sueyoshi T. Ser100-Phosphorylated ROR α Orchestrates CAR and HNF4 α to Form Active Chromatin Complex in Response to Phenobarbital to Regulate Induction of CYP2B6. Mol Pharmacol 2020; 97:191-201. [PMID: 31924695 PMCID: PMC6978708 DOI: 10.1124/mol.119.118273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/16/2019] [Indexed: 01/11/2023] Open
Abstract
We have previously shown that the retinoid-related orphan receptor alpha (RORα) phosphorylation plays a pivotal role in sulfotransferase 1E1 gene regulation within mouse liver. Here, we found serine 100-phosphorylated RORα orchestrates constitutive androstane receptor (CAR) and hepatocyte nuclear factor 4 alpha (HNF4α) to induce CYP2B6 by phenobarbital (PB) in human primary hepatocytes (HPHs). RORα knockdown using small interfering RNAs suppressed CYP2B6 mRNAs in HPH, whereas transient expression of RORα in COS-1 cells activated CYP2B6 promoter activity in reporter assays. Through chromatin immunoprecipitation (IP) and gel shift assays, we found that RORα in the form of phosphorylated (p-) S100 directly bound to a newly identified RORα response element (RORα response element on CYP2B6 promoter, -660/-649) within the CYP2B6 promoter in untreated or treated HPH. In PB-treated HPH, p-Ser100 RORα was both enriched in the distal phenobarbital response element module (PBREM) and the proximal okadaic acid response element (OARE), a known HNF4α binding site. Chromatin conformation capture assay revealed direct contact between the PBREM and OARE only in PB-treated HPH. Moreover, CAR preferably interacted with phosphomimetically mutated RORα at Ser100 residue in co-IP assay. A gel shift assay with a radiolabeled OARE module and nuclear extracts prepared from PB-treated mouse liver confirmed that HNF4α formed a complex with Ser 100-phosphorylated RORα, as shown by supershifted complexes with anti-p-Ser100 RORα and anti-HNF4α antibodies. Altogether, the results established that p-Ser100 RORα bridging the PBREM and OARE orchestrates CAR and HNF4α to form active chromatin complex during PB-induced CYP2B6 expression in human primary hepatocytes. SIGNIFICANCE STATEMENT: CYP2B6 is a vital enzyme for the metabolic elimination of xenobiotics, and it is prone to induction by xenobiotics, including phenobarbital via constitutive androstane receptor (CAR) and hepatocyte nuclear factor 4 alpha (HNF4α). Here, we show that retinoid-related orphan receptor alpha (RORα), through phosphorylated S100 residue, orchestrated CAR-HNF4α interaction on the CYP2B6 promoter in human primary hepatocyte cultures. These results signify not only the role of RORα in the molecular process of CYP2B6 induction, but it also reveals the importance of conserved phosphorylation sites within the DNA-binding domain of the receptor.
Collapse
Affiliation(s)
- Muluneh Fashe
- Pharmacogenetics section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Takuyu Hashiguchi
- Pharmacogenetics section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Masahiko Negishi
- Pharmacogenetics section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Tatsuya Sueyoshi
- Pharmacogenetics section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
18
|
Bernasconi C, Pelkonen O, Andersson TB, Strickland J, Wilk-Zasadna I, Asturiol D, Cole T, Liska R, Worth A, Müller-Vieira U, Richert L, Chesne C, Coecke S. Validation of in vitro methods for human cytochrome P450 enzyme induction: Outcome of a multi-laboratory study. Toxicol In Vitro 2019; 60:212-228. [PMID: 31158489 PMCID: PMC6718736 DOI: 10.1016/j.tiv.2019.05.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
CYP enzyme induction is a sensitive biomarker for phenotypic metabolic competence of in vitro test systems; it is a key event associated with thyroid disruption, and a biomarker for toxicologically relevant nuclear receptor-mediated pathways. This paper summarises the results of a multi-laboratory validation study of two in vitro methods that assess the potential of chemicals to induce cytochrome P450 (CYP) enzyme activity, in particular CYP1A2, CYP2B6, and CYP3A4. The methods are based on the use of cryopreserved primary human hepatocytes (PHH) and human HepaRG cells. The validation study was coordinated by the European Union Reference Laboratory for Alternatives to Animal Testing of the European Commission's Joint Research Centre and involved a ring trial among six laboratories. The reproducibility was assessed within and between laboratories using a validation set of 13 selected chemicals (known human inducers and non-inducers) tested under blind conditions. The ability of the two methods to predict human CYP induction potential was assessed. Chemical space analysis confirmed that the selected chemicals are broadly representative of a diverse range of chemicals. The two methods were found to be reliable and relevant in vitro tools for the assessment of human CYP induction, with the HepaRG method being better suited for routine testing. Recommendations for the practical application of the two methods are proposed.
Collapse
Affiliation(s)
| | - Olavi Pelkonen
- Research Unit of Biomedicine/Pharmacology and Toxicology, Faculty of Medicine, Aapistie 5B, University of Oulu, FIN-90014, Finland; Clinical Research Center, Oulu University Hospital, Finland
| | - Tommy B Andersson
- Drug Metabolism and Pharmacokinetics, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden; Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Judy Strickland
- Integrated Laboratory Systems (contractor supporting NICEATM), Research Triangle Park, North, Carolina, 27709, USA
| | | | - David Asturiol
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Thomas Cole
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Roman Liska
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Ursula Müller-Vieira
- Boehringer Ingelheim, Germany. Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, an der Riss, Germany
| | - Lysiane Richert
- KaLy-Cell, 20A, rue du Général Leclerc, 67115 Plobsheim, France(g) Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760 Saint Grégoire, France
| | - Christophe Chesne
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760 Saint Grégoire, France
| | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|
19
|
Li L, Welch MA, Li Z, Mackowiak B, Heyward S, Swaan PW, Wang H. Mechanistic Insights of Phenobarbital-Mediated Activation of Human but Not Mouse Pregnane X Receptor. Mol Pharmacol 2019; 96:345-354. [PMID: 31436536 PMCID: PMC6701513 DOI: 10.1124/mol.119.116616] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/07/2019] [Indexed: 12/12/2022] Open
Abstract
Phenobarbital (PB), a broadly used antiseizure drug, was the first to be characterized as an inducer of cytochrome P450 by activation of the constitutive androstane receptor (CAR). Although PB is recognized as a conserved CAR activator among species via a well-documented indirect activation mechanism, conflicting results have been reported regarding PB regulation of the pregnane X receptor (PXR), a sister receptor of CAR, and the underlying mechanisms remain elusive. Here, we show that in a human CAR (hCAR)-knockout (KO) HepaRG cell line, PB significantly induces the expression of CYP2B6 and CYP3A4, two shared target genes of hCAR and human PXR (hPXR). In human primary hepatocytes and hCAR-KO HepaRG cells, PB-induced expression of CYP3A4 was markedly repressed by genetic knockdown or pharmacological inhibition of hPXR. Mechanistically, PB concentration dependently activates hPXR but not its mouse counterpart in cell-based luciferase assays. Mammalian two-hybrid assays demonstrated that PB selectively increases the functional interaction between the steroid receptor coactivator-1 and hPXR but not mouse PXR. Moreover, surface plasmon resonance binding affinity assay showed that PB directly binds to the ligand binding domain of hPXR (KD = 1.42 × 10-05). Structure-activity analysis further revealed that the amino acid tryptophan-299 within the ligand binding pocket of hPXR plays a key role in the agonistic binding of PB and mutation of tryptophan-299 disrupts PB activation of hPXR. Collectively, these data reveal that PB, a selective mouse CAR activator, activates both hCAR and hPXR, and provide novel mechanistic insights for PB-mediated activation of hPXR.
Collapse
Affiliation(s)
- Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| | - Matthew A Welch
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| | - Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| | - Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| | - Peter W Swaan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., M.A.W., Z.L., B.M., P.W.S., H.W.); and BioIVT, Halethorpe, Maryland (S.H.)
| |
Collapse
|
20
|
Mackowiak B, Li L, Lynch C, Ziman A, Heyward S, Xia M, Wang H. High-content analysis of constitutive androstane receptor (CAR) translocation identifies mosapride citrate as a CAR agonist that represses gluconeogenesis. Biochem Pharmacol 2019; 168:224-236. [PMID: 31306645 DOI: 10.1016/j.bcp.2019.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/10/2019] [Indexed: 12/31/2022]
Abstract
The constitutive androstane receptor (CAR) plays an important role in hepatic drug metabolism and detoxification but has recently been projected as a potential drug target for metabolic disorders due to its repression of lipogenesis and gluconeogenesis. Thus, identification of physiologically-relevant CAR modulators has garnered significant interest. Here, we adapted the previously characterized human CAR (hCAR) nuclear translocation assay in human primary hepatocytes (HPH) to a high-content format and screened an FDA-approved drug library containing 978 compounds. Comparison of hCAR nuclear translocation results with the Tox21 hCAR luciferase reporter assay database in 643 shared compounds revealed significant overlap between these two assays, with approximately half of hCAR agonists also mediating nuclear translocation. Further validation of these compounds in HPH and/or using published data from literature demonstrated that hCAR translocation exhibits a higher correlation with the induction of hCAR target genes, such as CYP2B6, than the luciferase assay. In addition, some CAR antagonists which repress CYP2B6 mRNA expression in HPH, such as sorafenib, rimonabant, and CINPA1, were found to translocate hCAR to the nucleus of HPH. Notably, both the translocation assay and the luciferase assay identified mosapride citrate (MOS), a gastroprokinetic agent that is known to reduce fasting blood glucose levels in humans, as a novel hCAR activator. Further studies with MOS in HPH uncovered that MOS can repress the expression of gluconeogenic genes and decrease glucose output from hepatocytes, providing a previously unidentified liver-specific mechanism by which MOS modulates blood glucose levels.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, United States
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, United States
| | - Caitlin Lynch
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, United States
| | - Andrew Ziman
- Nikon Instruments Inc., 1300 Walt Whitman Road, Melville, NY 11747, United States
| | - Scott Heyward
- Bioreclamation In Vitro Technologies, 1450 S Rolling Rd, Halethorpe, MD 21227, United States
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, United States
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, United States.
| |
Collapse
|
21
|
Carazo A, Dusek J, Holas O, Skoda J, Hyrsova L, Smutny T, Soukup T, Dosedel M, Pávek P. Teriflunomide Is an Indirect Human Constitutive Androstane Receptor (CAR) Activator Interacting With Epidermal Growth Factor (EGF) Signaling. Front Pharmacol 2018; 9:993. [PMID: 30364229 PMCID: PMC6193428 DOI: 10.3389/fphar.2018.00993] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/13/2018] [Indexed: 01/19/2023] Open
Abstract
The constitutive androstane receptor (CAR) is a nuclear receptor involved mainly in xenobiotic and endobiotic metabolism regulation. CAR is activated directly by its ligands via the ligand binding domain (LBD) or indirectly by inhibition of the epidermal growth factor (EGF) signaling. We found that leflunomide (LEF) and its main metabolite teriflunomide (TER), both used for autoimmune diseases treatment, induce the prototype CAR target gene CYP2B6 in primary human hepatocytes. As TER was discovered to be an EGF receptor antagonist, we sought to determine if TER is an indirect activator of CAR. In primary human hepatocytes and in differentiated HepaRG cells, we found that LEF and TER up-regulate CAR target genes CYP2B6 and CYP3A4 mRNAs and enzymatic activities. TER stimulated CAR+A mutant translocation into the nucleus but neither LEF nor TER activated the CAR LBD, CAR3 variant or pregnane X receptor (PXR) in gene reporter assays. Interestingly, TER significantly up-regulated CAR mRNA expression, a result which could be a consequence of both EGF receptor and ELK-1 transcription factor inhibition by TER or by TER-mediated activation of glucocorticoid receptor (GR), an upstream hormonal regulator of CAR. We can conclude that TER is a novel indirect CAR activator which through EGF inhibition and GR activation controls both detoxification and some intermediary metabolism genes.
Collapse
Affiliation(s)
- Alejandro Carazo
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czechia.,Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czechia
| | - Jan Dusek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czechia
| | - Ondrej Holas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Charles University, Prague, Czechia
| | - Josef Skoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czechia
| | - Lucie Hyrsova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czechia
| | - Tomas Smutny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czechia
| | - Tomas Soukup
- Division of Rheumatology, 2nd Department of Internal Medicine - Gastroenterology, Faculty of Medicine, University Hospital in Hradec Kralove, Charles University, Prague, Czechia
| | - Martin Dosedel
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy, Charles University, Prague, Czechia
| | - Petr Pávek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Prague, Czechia
| |
Collapse
|
22
|
Piekos SC, Chen L, Wang P, Shi J, Yaqoob S, Zhu HJ, Ma X, Zhong XB. Consequences of Phenytoin Exposure on Hepatic Cytochrome P450 Expression during Postnatal Liver Maturation in Mice. Drug Metab Dispos 2018; 46:1241-1250. [PMID: 29884652 PMCID: PMC6053591 DOI: 10.1124/dmd.118.080861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/01/2018] [Indexed: 12/15/2022] Open
Abstract
The induction of cytochrome P450 (P450) enzymes in response to drug treatment is a significant contributing factor to drug-drug interactions, which may reduce therapeutic efficacy and/or cause toxicity. Since most studies on P450 induction are performed in adults, enzyme induction at neonatal, infant, and adolescent ages is not well understood. Previous work defined the postnatal ontogeny of drug-metabolizing P450s in human and mouse livers; however, there are limited data on the ontogeny of the induction potential of each enzyme in response to drug treatment. Induction of P450s at the neonatal age may also cause permanent alterations in P450 expression in adults. The goal of this study was to investigate the short- and long-term effects of phenytoin treatment on mRNA and protein expressions and enzyme activities of CYP2B10, 2C29, 3A11, and 3A16 at different ages during postnatal liver maturation in mice. Induction of mRNA immediately following phenytoin treatment appeared to depend on basal expression of the enzyme at a specific age. While neonatal mice showed the greatest fold changes in CYP2B10, 2C29, and 3A11 mRNA expression following treatment, the levels of induced protein expression and enzymatic activity were much lower than that of induced levels in adults. The expression of fetal CYP3A16 was repressed by phenytoin treatment. Neonatal treatment with phenytoin did not permanently induce enzyme expression in adulthood. Taken together, our data suggest that inducibility of drug-metabolizing P450s is much lower in neonatal mice than it is in adults and neonatal induction by phenytoin is not permanent.
Collapse
Affiliation(s)
- Stephanie C Piekos
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Liming Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Pengcheng Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Jian Shi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Sharon Yaqoob
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Hao-Jie Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (S.C.P., L.C., S.Y., X.-b.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W., X.M.); and Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.)
| |
Collapse
|
23
|
Cellular adaptation to xenobiotics: Interplay between xenosensors, reactive oxygen species and FOXO transcription factors. Redox Biol 2017; 13:646-654. [PMID: 28818793 PMCID: PMC5558470 DOI: 10.1016/j.redox.2017.07.015] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/20/2017] [Accepted: 07/28/2017] [Indexed: 12/22/2022] Open
Abstract
Cells adapt to an exposure to xenobiotics by upregulating the biosynthesis of proteins involved in xenobiotic metabolism. This is achieved largely via activation of cellular xenosensors that modulate gene expression. Biotransformation of xenobiotics frequently comes with the generation of reactive oxygen species (ROS). ROS, in turn, are known modulators of signal transduction processes. FOXO (forkhead box, class O) transcription factors are among the proteins deeply involved in the cellular response to stress, including oxidative stress elicited by the formation of ROS. On the one hand, FOXO activity is modulated by ROS, while on the other, FOXO target genes include many that encode antioxidant proteins – thereby establishing a regulatory circuit. Here, the role of ROS and of FOXOs in the regulation of xenosensor transcriptional activities will be discussed. Constitutive androstane receptor (CAR), pregnane X receptor (PXR), peroxisome proliferator-activated receptors (PPARs), arylhydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2) all interact with FOXOs and/or ROS. The two latter not only fine-tune the activities of xenosensors but also mediate interactions between them. As a consequence, the emerging picture of an interplay between xenosensors, ROS and FOXO transcription factors suggests a modulatory role of ROS and FOXOs in the cellular adaptive response to xenobiotics. Exposure of cells to xenobiotics may trigger formation of reactive oxygen species. Xenosensors respond to xenobiotics by upregulation of xenobiotic metabolism. FOXO transcription factors modulate the activities of several xenosensors. ROS affect FOXO activity, and FOXO target genes include antioxidant proteins. FOXOs bridge xenobiotic-induced ROS generation and xenosensor regulation.
Collapse
|
24
|
Mackowiak B, Li L, Welch MA, Li D, Jones JW, Heyward S, Kane MA, Swaan PW, Wang H. Molecular Basis of Metabolism-Mediated Conversion of PK11195 from an Antagonist to an Agonist of the Constitutive Androstane Receptor. Mol Pharmacol 2017; 92:75-87. [PMID: 28442602 PMCID: PMC5452073 DOI: 10.1124/mol.117.108621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
The constitutive androstane receptor (CAR) plays an important role in xenobiotic metabolism, energy homeostasis, and cell proliferation. Antagonism of the CAR represents a key strategy for studying its function and may have potential clinical applications. However, specific human CAR (hCAR) antagonists are limited and conflicting data on the activity of these compounds have been reported. 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide (PK11195), a typical peripheral benzodiazepine receptor ligand, has been established as a potent hCAR deactivator in immortalized cells; whether it inhibits hCAR activity under physiologically relevant conditions remains unclear. Here, we investigated the effects of PK11195 on hCAR in metabolically competent human primary hepatocytes (HPH) and HepaRG cells. We show that although PK11195 antagonizes hCAR in HepG2 cells, it induces the expression of CYP2B6 and CYP3A4, targets of hCAR and the pregnane X receptor (PXR), in HPH, HepaRG, and PXR-knockout HepaRG cells. Utilizing a HPH-HepG2 coculture model, we demonstrate that inclusion of HPH converts PK11195 from an antagonist to an agonist of hCAR, and such conversion was attenuated by potent CYP3A4 inhibitor ketoconazole. Metabolically, we show that the N-desmethyl metabolite is responsible for PK11195-mediated hCAR activation by facilitating hCAR interaction with coactivators and enhancing hCAR nuclear translocation in HPHs. Structure-activity analysis revealed that N-demethylation alters the interaction of PK11195 with the binding pocket of hCAR to favor activation. Together, these results indicate that removal of a methyl group switches PK11195 from a potent antagonist of hCAR to an agonist in HPH and highlights the importance of physiologically relevant metabolism when attempting to define the biologic action of small molecules.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Matthew A Welch
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Daochuan Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Peter W Swaan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (B.M., L.L., M.A.W., D.L., J.W.J., M.A.K., P.W.S., H.W.); and Bioreclamation In Vitro Technologies, Halethorpe, Maryland (S.H.)
| |
Collapse
|
25
|
Kato H, Yamaotsu N, Iwazaki N, Okamura S, Kume T, Hirono S. Precise prediction of activators for the human constitutive androstane receptor using structure-based three-dimensional quantitative structure–activity relationship methods. Drug Metab Pharmacokinet 2017; 32:179-188. [DOI: 10.1016/j.dmpk.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/24/2017] [Accepted: 02/01/2017] [Indexed: 02/07/2023]
|
26
|
Dong JQ, Gosset JR, Fahmi OA, Lin Z, Chabot JR, Terra SG, Le V, Chidsey K, Nouri P, Kim A, Buckbinder L, Kalgutkar AS. Examination of the Human Cytochrome P4503A4 Induction Potential of PF-06282999, an Irreversible Myeloperoxidase Inactivator: Integration of Preclinical, In Silico, and Biomarker Methodologies in the Prediction of the Clinical Outcome. Drug Metab Dispos 2017; 45:501-511. [PMID: 28254951 DOI: 10.1124/dmd.116.074476] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/28/2017] [Indexed: 12/11/2022] Open
Abstract
The propensity for CYP3A4 induction by 2-(6-(5-chloro-2-methoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide (PF-06282999), an irreversible inactivator of myeloperoxidase, was examined in the present study. Studies using human hepatocytes revealed moderate increases in CYP3A4 mRNA and midazolam-1'-hydroxylase activity in a PF-06282999 dose-dependent fashion. At the highest tested concentration of 300 μM, PF-06282999 caused maximal induction in CYP3A4 mRNA and enzyme activity ranging from 56% to 86% and 47% t0 72%, respectively, of rifampicin response across the three hepatocyte donor pools. In a clinical drug-drug interaction (DDI) study, the mean midazolam Cmax and area under the curve (AUC) values following 14-day treatment with PF-06282999 decreased in a dose-dependent fashion with a maximum decrease in midazolam AUC0-inf and Cmax of ∼57.2% and 41.1% observed at the 500 mg twice daily dose. The moderate impact on midazolam pharmacokinetics at the 500 mg twice daily dose of PF-06282999 was also reflected in statistically significant changes in plasma 4β-hydroxycholesterol/cholesterol and urinary 6β-hydroxycortisol/cortisol ratios. Changes in plasma and urinary CYP3A4 biomarkers did not reach statistical significance at the 125 mg three times daily dose of PF-06282999, despite a modest decrease in midazolam systemic exposure. Predicted DDI magnitude based on the in vitro induction parameters and simulated pharmacokinetics of perpetrator (PF-06282999) and victim (midazolam) using the Simcyp (Simcyp Ltd., Sheffield, United Kingdom) population-based simulator were in reasonable agreement with the observed clinical data. Since the magnitude of the 4β-hydroxycholesterol or 6β-hydroxycortisol ratio change was generally smaller than the magnitude of the midazolam AUC change with PF-06282999, a pharmacokinetic interaction study with midazolam ultimately proved important for assessment of DDI via CYP3A4 induction.
Collapse
Affiliation(s)
- Jennifer Q Dong
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - James R Gosset
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Odette A Fahmi
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Zhiwu Lin
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Jeffrey R Chabot
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Steven G Terra
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Vu Le
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Kristin Chidsey
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Parya Nouri
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Albert Kim
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Leonard Buckbinder
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| | - Amit S Kalgutkar
- Clinical Pharmacology (J.Q.D.), Pharmacokinetics, Pharmacodynamics, and Metabolism (J.R.G., J.R.C., A.S.K.), Statistics (V.L.), Early Clinical Development (K.C., A.K.), and Cardiovascular and Metabolic Disease Research Unit (L.B.), Pfizer Inc., Cambridge, Massachusetts; and Pharmacokinetics, Pharmacodynamics, and Metabolism (O.A.F., Z.L.), Clinical Development (S.G.T.), and Clinical Assay Group (P.N.), Pfizer Inc., Groton, Connecticut
| |
Collapse
|
27
|
Panomvana D, Methaneethorn J, Vachirayonstien T. Correlation Between Elimination Parameters of Phenytoin and Carbamazepine in Patients with Epilepsy Receiving Both Drugs Concomitantly: A Preliminary Study. Pharmaceut Med 2017. [DOI: 10.1007/s40290-017-0182-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
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
|
29
|
Lee K, You H, Choi J, No KT. Development of pharmacophore-based classification model for activators of constitutive androstane receptor. Drug Metab Pharmacokinet 2016; 32:172-178. [PMID: 28366619 DOI: 10.1016/j.dmpk.2016.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/21/2016] [Accepted: 11/10/2016] [Indexed: 10/20/2022]
Abstract
Constitutive androstane receptor (CAR) is predominantly expressed in the liver and is important for regulating drug metabolism and transport. Despite its biological importance, there have been few attempts to develop in silico models to predict the activity of CAR modulated by chemical compounds. The number of in silico studies of CAR may be limited because of CAR's constitutive activity under normal conditions, which makes it difficult to elucidate the key structural features of the interaction between CAR and its ligands. In this study, to address these limitations, we introduced 3D pharmacophore-based descriptors with an integrated ligand and structure-based pharmacophore features, which represent the receptor-ligand interaction. Machine learning methods (support vector machine and artificial neural network) were applied to develop an in silico model with the descriptors containing significant information regarding the ligand binding positions. The best classification model built with a solvent accessibility volume-based filter and the support vector machine showed good predictabilities of 87%, and 85.4% for the training set and validation set, respectively. This demonstrates that our model can be used to accurately predict CAR activators and offers structural information regarding ligand/protein interactions.
Collapse
Affiliation(s)
- Kyungro Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Hwan You
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Jiwon Choi
- Bioinformatics & Molecular Design Research Center, Yonsei University, Seoul 03722, South Korea
| | - Kyoung Tai No
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea; Bioinformatics & Molecular Design Research Center, Yonsei University, Seoul 03722, South Korea.
| |
Collapse
|
30
|
Pinne M, Ponce E, Raucy JL. Transactivation Assays to Assess Canine and Rodent Pregnane X Receptor (PXR) and Constitutive Androstane Receptor (CAR) Activation. PLoS One 2016; 11:e0164642. [PMID: 27732639 PMCID: PMC5061317 DOI: 10.1371/journal.pone.0164642] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/28/2016] [Indexed: 11/21/2022] Open
Abstract
The pregnane X receptor (PXR/SXR, NR1I2) and constitutive androstane receptor (CAR, NR1I3) are nuclear receptors (NRs) involved in the regulation of many genes including cytochrome P450 enzymes (CYPs) and transporters important in metabolism and uptake of both endogenous substrates and xenobiotics. Activation of these receptors can lead to adverse drug effects as well as drug-drug interactions. Depending on which nuclear receptor is activated will determine which adverse effect could occur, making identification important. Screening for NR activation by New Molecular Entities (NMEs) using cell-based transactivation assays is the singular high throughput method currently available for identifying the activation of a particular NR. Moreover, screening for species-specific NR activation can minimize the use of animals in drug development and toxicology studies. With this in mind, we have developed in vitro transactivation assays to identify compounds that activate canine and rat PXR and CAR3. We found differences in specificity for canine and rat PXR, with the best activator for canine PXR being 10 μM SR12813 (60.1 ± 3.1-fold) and for rat PXR, 10 μM dexamethasone (60.9 ± 8.4 fold). Of the 19 test agents examined, 10 and 9 significantly activated rat and canine PXR at varying degrees, respectively. In contrast, 5 compounds exhibited statistically significant activation of rat CAR3 and 4 activated the canine receptor. For canine CAR3, 50 μM artemisinin proved to be the best activator (7.3 ± 1.8 and 10.5 ± 2.2 fold) while clotrimazole (10 μM) was the primary activator of the rat variant (13.7 ± 0.8 and 26.9 ± 1.3 fold). Results from these studies demonstrated that cell-based transactivation assays can detect species-specific activators and revealed that PXR was activated by at least twice as many compounds as was CAR3, suggesting that there are many more agonists for PXR than CAR.
Collapse
Affiliation(s)
- Marija Pinne
- Puracyp, Inc., Carlsbad, California, United States of America
- * E-mail:
| | - Elsa Ponce
- Puracyp, Inc., Carlsbad, California, United States of America
| | - Judy L. Raucy
- Puracyp, Inc., Carlsbad, California, United States of America
| |
Collapse
|
31
|
Kawase A, Tanaka H, Otori T, Matsuyama K, Iwaki M. Effects of duration of phenytoin administration on mRNA expression of cytochrome P450 and P-glycoprotein in the liver and small intestine of rats. Asian J Pharm Sci 2016. [DOI: 10.1016/j.ajps.2016.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
32
|
Williamson B, Lorbeer M, Mitchell MD, Brayman TG, Riley RJ. Evaluation of a novel PXR-knockout in HepaRG ™ cells. Pharmacol Res Perspect 2016; 4:e00264. [PMID: 27713827 PMCID: PMC5045942 DOI: 10.1002/prp2.264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/10/2016] [Indexed: 11/08/2022] Open
Abstract
The nuclear pregnane X receptor (PXR) regulates the expression of genes involved in the metabolism, hepatobiliary disposition, and toxicity of drugs and endogenous compounds. PXR is a promiscuous nuclear hormone receptor (NHR) with significant ligand and DNA‐binding crosstalk with the constitutive androstane receptor (CAR); hence, defining the precise role of PXR in gene regulation is challenging. Here, utilising a novel PXR‐knockout (KO) HepaRG cell line, real‐time PCR analysis was conducted to determine PXR involvement for a range of inducers. The selective PXR agonist rifampicin, a selective CAR activator, 6‐(4‐chlorophenyl)imidazo[2,1‐b][1,3]thiazole‐5‐carbaldehyde O‐(3,4‐dichlorobenzyl)oxime (CITCO), and dual activators of CAR and PXR including phenobarbital (PB) were analyzed. HepaRG control cells (5F clone) were responsive to prototypical inducers of CYP2B6 and CYP3A4. No response was observed in the PXR‐KO cells treated with rifampicin. Induction of CYP3A4 by PB, artemisinin, and phenytoin was also much reduced in PXR‐KO cells, while the response to CITCO was maintained. This finding is in agreement with the abolition of functional PXR expression. The apparent EC50 values for PB were in agreement between the cell lines; however, CITCO was ~threefold (0.3 μmol/L vs. 1 μmol/L) lower in the PXR‐KO cells compared with the 5F cells for CYP2B6 induction. Results presented support the application of the novel PXR‐KO cells in the definitive assignment of PXR‐mediated CYP2B6 and CYP3A4 induction. Utilization of such cell lines will allow advancement in composing structure activity relationships rather than relying predominantly on pharmacological manipulations and provide in‐depth mechanistic evaluation.
Collapse
Affiliation(s)
- Beth Williamson
- Evotec (UK) Ltd 114 Innovation Drive Abingdon Oxfordshire OX14 4RZ United Kingdom
| | - Mathias Lorbeer
- Evotec (UK) Ltd 114 Innovation Drive Abingdon Oxfordshire OX14 4RZ United Kingdom
| | | | | | - Robert J Riley
- Evotec (UK) Ltd 114 Innovation Drive Abingdon Oxfordshire OX14 4RZ United Kingdom
| |
Collapse
|
33
|
Chai SC, Cherian MT, Wang YM, Chen T. Small-molecule modulators of PXR and CAR. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1859:1141-1154. [PMID: 26921498 PMCID: PMC4975625 DOI: 10.1016/j.bbagrm.2016.02.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/06/2016] [Accepted: 02/06/2016] [Indexed: 12/27/2022]
Abstract
Two nuclear receptors, the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR), participate in the xenobiotic detoxification system by regulating the expression of drug-metabolizing enzymes and transporters in order to degrade and excrete foreign chemicals or endogenous metabolites. This review aims to expand the perceived relevance of PXR and CAR beyond their established role as master xenosensors to disease-oriented areas, emphasizing their modulation by small molecules. Structural studies of these receptors have provided much-needed insight into the nature of their binding promiscuity and the important elements that lead to ligand binding. Reports of species- and isoform-selective activation highlight the need for further scrutiny when extrapolating from animal data to humans, as animal models are at the forefront of early drug discovery. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
Affiliation(s)
- Sergio C Chai
- Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Milu T Cherian
- Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yue-Ming Wang
- Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
34
|
Nagai M, Konno Y, Satsukawa M, Yamashita S, Yoshinari K. Establishment of In Silico Prediction Models for CYP3A4 and CYP2B6 Induction in Human Hepatocytes by Multiple Regression Analysis Using Azole Compounds. Drug Metab Dispos 2016; 44:1390-8. [PMID: 27208383 DOI: 10.1124/dmd.115.068619] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 05/18/2016] [Indexed: 11/22/2022] Open
Abstract
Drug-drug interactions (DDIs) via cytochrome P450 (P450) induction are one clinical problem leading to increased risk of adverse effects and the need for dosage adjustments and additional therapeutic monitoring. In silico models for predicting P450 induction are useful for avoiding DDI risk. In this study, we have established regression models for CYP3A4 and CYP2B6 induction in human hepatocytes using several physicochemical parameters for a set of azole compounds with different P450 induction as characteristics as model compounds. To obtain a well-correlated regression model, the compounds for CYP3A4 or CYP2B6 induction were independently selected from the tested azole compounds using principal component analysis with fold-induction data. Both of the multiple linear regression models obtained for CYP3A4 and CYP2B6 induction are represented by different sets of physicochemical parameters. The adjusted coefficients of determination for these models were of 0.8 and 0.9, respectively. The fold-induction of the validation compounds, another set of 12 azole-containing compounds, were predicted within twofold limits for both CYP3A4 and CYP2B6. The concordance for the prediction of CYP3A4 induction was 87% with another validation set, 23 marketed drugs. However, the prediction of CYP2B6 induction tended to be overestimated for these marketed drugs. The regression models show that lipophilicity mostly contributes to CYP3A4 induction, whereas not only the lipophilicity but also the molecular polarity is important for CYP2B6 induction. Our regression models, especially that for CYP3A4 induction, might provide useful methods to avoid potent CYP3A4 or CYP2B6 inducers during the lead optimization stage without performing induction assays in human hepatocytes.
Collapse
Affiliation(s)
- Mika Nagai
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical, Kyoto, Japan (M.N., Y.K., M.S.); Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (M.N., K.Y.); and Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan (S.Y.)
| | - Yoshihiro Konno
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical, Kyoto, Japan (M.N., Y.K., M.S.); Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (M.N., K.Y.); and Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan (S.Y.)
| | - Masahiro Satsukawa
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical, Kyoto, Japan (M.N., Y.K., M.S.); Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (M.N., K.Y.); and Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan (S.Y.)
| | - Shinji Yamashita
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical, Kyoto, Japan (M.N., Y.K., M.S.); Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (M.N., K.Y.); and Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan (S.Y.)
| | - Kouichi Yoshinari
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical, Kyoto, Japan (M.N., Y.K., M.S.); Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (M.N., K.Y.); and Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan (S.Y.)
| |
Collapse
|
35
|
Zhang L, Miao XJ, Wang X, Pan HH, Li P, Ren H, Jia YR, Lu C, Wang HB, Yuan L, Zhang GL. Antiproliferation of berberine is mediated by epigenetic modification of constitutive androstane receptor (CAR) metabolic pathway in hepatoma cells. Sci Rep 2016; 6:28116. [PMID: 27311637 PMCID: PMC4911599 DOI: 10.1038/srep28116] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/26/2016] [Indexed: 01/07/2023] Open
Abstract
Constitutive androstane receptor (CAR) regulates hepatic xenobiotic and energy metabolism, as well as promotes cell growth and hepatocarcinogenesis. Berberine is an ancient multipotent alkaloid drug which derived from Coptis chinensis plants. Here we report that berberine is able to be cellular uptake and accessible to chromatin in human hepatoma HepG2 cells. Berberine induces more apoptosis, cell cycle arrest, but less ROS production in CAR overexpressed mCAR-HepG2 cells. Moreover, berberine inhibits expressions of CAR and its target genes CYP2B6 and CYP3A4. Furthermore, berberine enhances DNA methylation level in whole genome but reduces that in promoter regions CpG sites of CYP2B6 and CYP3A4 genes under the presence of CAR condition. These results indicated that the antiproliferation of berberine might be mediated by the unique epigenetic modifying mechanism of CAR metabolic pathway, suggesting that berberine is a promising candidate in anticancer adjuvant chemotherapy, due to its distinct pharmacological properties in clinic.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xiao-Jie Miao
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hai-Hui Pan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Pu Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hong Ren
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yong-Rui Jia
- Medical and Healthy Analytical Center, Peking University, Beijing, 100191, China
| | - Chuang Lu
- Department of Drug Metabolism &Pharmacokinetics, Biogen, Cambridge, Massachusetts, USA
| | - Hong-Bing Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, USA
| | - Lan Yuan
- Medical and Healthy Analytical Center, Peking University, Beijing, 100191, China
| | - Guo-Liang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
36
|
Tien YC, Liu K, Pope C, Wang P, Ma X, Zhong XB. Dose of Phenobarbital and Age of Treatment at Early Life are Two Key Factors for the Persistent Induction of Cytochrome P450 Enzymes in Adult Mouse Liver. Drug Metab Dispos 2015; 43:1938-45. [PMID: 26400395 PMCID: PMC4658495 DOI: 10.1124/dmd.115.066316] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/21/2015] [Indexed: 02/01/2023] Open
Abstract
Drug treatment of neonates and infants and its long-term consequences on drug responses have emerged in recent years as a major challenge for health care professionals. In the current study, we use phenobarbital as a model drug and mouse as an in vivo model to demonstrate that the dose of phenobarbital and age of treatment are two key factors for the persistent induction of gene expression and consequential increases of enzyme activities of Cyp2b, Cyp2c, and Cyp3a in adult livers. We show that phenobarbital treatment at early life of day 5 after birth with a low dose (<100 mg/kg) does not change expression and enzyme activities of Cyp2b, Cyp2c, and Cyp3a in adult mouse liver, whereas phenobarbital treatment with a high dose (>200 mg/kg) significantly increases expression and enzyme activities of these P450s in adult liver. We also demonstrate that phenobarbital treatment before day 10 after birth, but not at later ages, significantly increases mRNAs, proteins, and enzyme activities of the tested P450s. Such persistent induction of P450 gene expression and enzyme activities in adult livers by phenobarbital treatment only occurs within a sensitive age window early in life. The persistent induction in gene expression and enzyme activities is higher in female mice than in male mice for Cyp2b10 but not for Cyp2c29 and Cyp3a11. These results will stimulate studies to evaluate the long-term impacts of drug treatment with different doses at neonatal and infant ages on drug metabolism, therapeutic efficacy, and drug-induced toxicity throughout the rest of life.
Collapse
Affiliation(s)
- Yun-Chen Tien
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (Y.C.T., C.P., X.B.Z.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (K.L., P.W., X.M.)
| | - Ke Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (Y.C.T., C.P., X.B.Z.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (K.L., P.W., X.M.)
| | - Chad Pope
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (Y.C.T., C.P., X.B.Z.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (K.L., P.W., X.M.)
| | - Pengcheng Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (Y.C.T., C.P., X.B.Z.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (K.L., P.W., X.M.)
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (Y.C.T., C.P., X.B.Z.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (K.L., P.W., X.M.)
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (Y.C.T., C.P., X.B.Z.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (K.L., P.W., X.M.)
| |
Collapse
|
37
|
Ramachandran SD, Vivarès A, Klieber S, Hewitt NJ, Muenst B, Heinz S, Walles H, Braspenning J. Applicability of second-generation upcyte® human hepatocytes for use in CYP inhibition and induction studies. Pharmacol Res Perspect 2015; 3:e00161. [PMID: 26516577 PMCID: PMC4618636 DOI: 10.1002/prp2.161] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/08/2015] [Indexed: 11/08/2022] Open
Abstract
Human upcyte® hepatocytes are proliferating hepatocytes that retain many characteristics of primary human hepatocytes. We conducted a comprehensive evaluation of the application of second-generation upcyte® hepatocytes from four donors for inhibition and induction assays using a selection of reference inhibitors and inducers. CYP1A2, CYP2B6, CYP2C9, and CYP3A4 were reproducibly inhibited in a concentration-dependent manner and the calculated IC50 values for each compound correctly classified them as potent inhibitors. Upcyte® hepatocytes were responsive to prototypical CYP1A2, CYP2B6, CYP2C9, and CYP3A4 inducers, confirming that they have functional AhR-, CAR-, and PXR-mediated CYP regulation. A panel of 11 inducers classified as potent, moderate or noninducers of CYP3A4 and CYP2B6 were tested. There was a good fit of data from upcyte® hepatocytes to three different predictive models for CYP3A4 induction, namely the Relative Induction Score (RIS), AUCu/F2, and C max,u/Ind50. In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. In conclusion, these data support the use of second-generation upcyte® hepatocytes for CYP inhibition and induction assays. Under the culture conditions used, these cells expressed CYP activities that were equivalent to or higher than those measured in primary human hepatocyte cultures, which could be inhibited or induced by prototypical CYP inhibitors and inducers, respectively. Moreover, they can be used to predict in vivo CYP3A4 induction potential using three prediction models. Bulk availability of cells from multiple donors makes upcyte® hepatocytes suitable for DDI screening, as well as more in-depth mechanistic investigations.
Collapse
Affiliation(s)
| | - Aurélie Vivarès
- Sanofi – DSAR Drug Disposition – In Vitro models371, rue du Pr. Blayac, Montpellier, 34000, France
| | - Sylvie Klieber
- Sanofi – DSAR Drug Disposition – In Vitro models371, rue du Pr. Blayac, Montpellier, 34000, France
| | | | - Bernhard Muenst
- Medicyte GmbHIm Neuenheimer Feld 581, Heidelberg, D-69120, Germany
| | - Stefan Heinz
- Medicyte GmbHIm Neuenheimer Feld 581, Heidelberg, D-69120, Germany
| | - Heike Walles
- Tissue Engineering and Regenerative Medicine, University WuerzburgRoentgenring 11, Wuerzburg, D-97070, Germany
| | | |
Collapse
|
38
|
Rubinchik-Stern M, Shmuel M, Eyal S. Antiepileptic drugs alter the expression of placental carriers: An in vitro study in a human placental cell line. Epilepsia 2015; 56:1023-32. [DOI: 10.1111/epi.13037] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Miriam Rubinchik-Stern
- Institute for Drug Research; School of Pharmacy; The Hebrew University; Jerusalem Israel
| | - Miri Shmuel
- Institute for Drug Research; School of Pharmacy; The Hebrew University; Jerusalem Israel
| | - Sara Eyal
- Institute for Drug Research; School of Pharmacy; The Hebrew University; Jerusalem Israel
| |
Collapse
|
39
|
Kobayashi K, Hashimoto M, Honkakoski P, Negishi M. Regulation of gene expression by CAR: an update. Arch Toxicol 2015; 89:1045-55. [PMID: 25975989 DOI: 10.1007/s00204-015-1522-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/27/2015] [Indexed: 11/30/2022]
Abstract
The constitutive androstane receptor (CAR), a member of the nuclear receptor superfamily, is a well-known xenosensor that regulates hepatic drug metabolism and detoxification. CAR activation can be elicited by a large variety of xenobiotics, including phenobarbital (PB) which is not a directly binding CAR ligand. The mechanism of CAR activation is complex and involves translocation from the cytoplasm into the nucleus, followed by further activation steps in the nucleus. Recently, epidermal growth factor receptor (EGFR) has been identified as a PB-responsive receptor, and PB activates CAR by inhibiting the EGFR signaling. In addition to regulation of drug metabolism, activation of CAR has multiple biological end points such as modulation of xenobiotic-elicited liver injury, and the role of CAR in endobiotic functions such as glucose metabolism and cholesterol homeostasis is increasingly recognized. Thus, investigations on the molecular mechanism of CAR activation are critical for the real understanding of CAR-mediated processes. Here, we summarize the current understanding of mechanisms by which CAR activators regulate gene expression through cellular signaling pathways and the roles of CAR on xenobiotic-elicited hepatocellular carcinoma, liver injury, glucose metabolism and cholesterol homeostasis.
Collapse
Affiliation(s)
- Kaoru Kobayashi
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan,
| | | | | | | |
Collapse
|
40
|
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
|
41
|
Carazo Fernández A, Smutny T, Hyrsová L, Berka K, Pavek P. Chrysin, baicalein and galangin are indirect activators of the human constitutive androstane receptor (CAR). Toxicol Lett 2015; 233:68-77. [DOI: 10.1016/j.toxlet.2015.01.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 11/30/2022]
|
42
|
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
|
43
|
Yamada T, Kikumoto H, Lake BG, Kawamura S. Lack of effect of metofluthrin and sodium phenobarbital on replicative DNA synthesis and Ki-67 mRNA expression in cultured human hepatocytes. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00217b] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
High doses of metofluthrin have been shown to produce hepatocellular tumours in rats.
Collapse
Affiliation(s)
- Tomoya Yamada
- Environmental Health Science Laboratory
- Sumitomo Chemical Company
- Ltd
- Osaka 554-8558
- Japan
| | - Hiroko Kikumoto
- Environmental Health Science Laboratory
- Sumitomo Chemical Company
- Ltd
- Osaka 554-8558
- Japan
| | - Brian G. Lake
- Centre for Toxicology
- Faculty of Health and Medical Sciences
- University of Surrey
- Guildford
- UK
| | - Satoshi Kawamura
- Environmental Health Science Laboratory
- Sumitomo Chemical Company
- Ltd
- Osaka 554-8558
- Japan
| |
Collapse
|
44
|
Uchida Y, Ohtsuki S, Terasaki T. Pharmacoproteomics-based reconstruction of in vivo P-glycoprotein function at blood-brain barrier and brain distribution of substrate verapamil in pentylenetetrazole-kindled epilepsy, spontaneous epilepsy, and phenytoin treatment models. Drug Metab Dispos 2014; 42:1719-26. [PMID: 25061162 DOI: 10.1124/dmd.114.059055] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The purpose of this study was to demonstrate experimentally that alterations of in vivo transporter function at the blood-brain barrier (BBB) in disease and during pharmacotherapy can be reconstructed from in vitro data based on our established pharmacoproteomic concept of reconstructing in vivo function by integrating intrinsic transport activity per transporter molecule and absolute protein expression level at the BBB. Pentylenetetrazole (PTZ)-kindled and spontaneous model of epilepsy (EL) mice were used as models of chemically induced and spontaneous epilepsy, respectively. A mouse model of antiepileptic drug treatment was prepared by consecutive 5-week administration of phenytoin (PHT). Quantitative targeted absolute proteomic analysis of 31 membrane proteins showed that P-glycoprotein (P-gp/mdr1a) protein expression levels were significantly increased in brain capillaries of PTZ (129%), EL (143%), and PHT mice (192%) compared with controls. The brain-to-plasma concentration ratios (Kp brain) of P-gp/mdr1a substrate verapamil were 0.563, 0.394, 0.432, and 0.234 in control, PTZ, EL, and PHT mice, respectively. In vivo P-gp/mdr1a function at the BBB was reconstructed from the measured P-gp/mdr1a protein expression levels and intrinsic transport activity for verapamil per P-gp/mdr1a previously reported by our group. Then, the reconstructed P-gp/mdr1a functional activities were integrated with unbound fractions of verapamil in plasma and brain to reconstruct Kp brain of verapamil. In all mice, reconstructed Kp brain values agreed well with the observed values within a 1.21-fold range. These results demonstrate that altered P-gp functions at the BBB in epilepsy and during pharmacotherapy can be reconstructed from in vitro data by means of our pharmacoproteomic approach.
Collapse
Affiliation(s)
- Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.U., T.T.); and Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.)
| | - Sumio Ohtsuki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.U., T.T.); and Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.)
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.U., T.T.); and Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.)
| |
Collapse
|
45
|
Lau AJ, Chang TKH. Indirect activation of the SV23 and SV24 splice variants of human constitutive androstane receptor: analysis with 3-hydroxyflavone and its analogues. Br J Pharmacol 2014; 170:403-14. [PMID: 23809009 DOI: 10.1111/bph.12284] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/17/2013] [Accepted: 06/20/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Naturally occurring splice variants of human CAR (hCAR), including hCAR-SV23 (insertion of amino acids SPTV) and hCAR-SV24 (APYLT), have been shown to be expressed in liver. However, little is known regarding how hCAR-SV23 and hCAR-SV24 are activated. Therefore, we investigated the mode of activation of these hCAR splice variants. EXPERIMENTAL APPROACH Cell-based reporter gene assays, including ligand-binding domain transactivation assays and coactivator recruitment assays, were conducted on cultured HepG2 cells transfected with various constructs and treated with 3-hydroxyflavone or a hydroxylated (galangin, datiscetin, kaempferol, morin, quercetin or myricetin) or methylated (isorhamnetin, tamarixetin, or syringetin) analogue. KEY RESULTS Among the flavonols investigated, only 3-hydroxyflavone increased hCAR-SV23 and hCAR-SV24 activities. 3-Hydroxyflavone did not transactivate the ligand-binding domain of these isoforms or recruit steroid receptor coactivators (SRC-1, SRC-2, or SRC-3). By comparison, 3-hydroxyflavone, galangin, datiscetin, kaempferol, quercetin, isorhamnetin and tamarixetin activated hCAR-WT, whereas none of the flavonols activated hCAR-SV25 (both SPTV and APYLT insertions). The flavonols 3-Hydroxyflavone, galangin, quercetin and tamarixetin transactivated the ligand-binding domain of hCAR-WT, but only 3-hydroxyflavone recruited SRC-1, SRC-2 and SRC-3 to the receptor. CONCLUSION AND IMPLICATIONS hCAR-SV23 and hCAR-SV24 can be activated by a mechanism that does not involve the ligand-binding domain of the receptor or recruitment of SRC-1, SRC-2, or SRC-3. 3-Hydroxyflavone and its structural analogues activated hCAR in an isoform-selective and chemical-specific manner. Overall, our study provides insight into a novel mode of ligand activation of hCAR-SV23 and hCAR-SV24.
Collapse
Affiliation(s)
- Aik Jiang Lau
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
46
|
Schneider Aguirre R, Karpen SJ. Inflammatory mediators increase SUMOylation of retinoid X receptor α in a c-Jun N-terminal kinase-dependent manner in human hepatocellular carcinoma cells. Mol Pharmacol 2013; 84:218-26. [PMID: 23690070 PMCID: PMC3716323 DOI: 10.1124/mol.113.085555] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/20/2013] [Indexed: 12/31/2022] Open
Abstract
Retinoid X receptor α [RXRα; nuclear receptor (NR)2B1] is a crucial regulator in the expression of a broad array of hepatic genes under both normal and pathologic conditions. During inflammation, RXRα undergoes rapid post-translational modifications, including c-Jun N-terminal kinase (JNK)-mediated phosphorylation, which correlates with a reduction in RXRα function. A small ubiquitin-like modifier (SUMO) acceptor site was recently described in human RXRα, yet the contributors, regulators, and consequences of SUMO-RXRα are not well understood. Inflammation and other stressors alter nuclear receptor function in liver and induce SUMOylation of several NRs as part of proinflammatory gene regulation, but linkages between these two pathways in liver, or for RXRα directly, remain unexplored. We sought to determine if inflammation induces SUMOylation of RXRα in human liver-derived (HuH-7) cells. Lipopolysaccharide, interleukin-1β, and tumor necrosis factor α (TNFα) rapidly and substantially stimulated SUMOylation of RXRα. Two RXRα ligands, 9-cis retinoic acid (9cRA) and LG268, induced SUMOylation of RXRα, whereas both inflammation- and ligand-induced SUMOylation of RXRα require the K108 residue. Pretreatment with 1,9-pyrazoloanthrone (SP600125), a potent JNK inhibitor, abrogates TNFα- and 9cRA-stimulated RXRα SUMOylation. Pretreatment with SUMOylation inhibitors markedly augmented basal expression of several RXRα-regulated hepatobiliary genes. These results indicate that inflammatory signaling pathways rapidly induce SUMOylation of RXRα, adding to the repertoire of RXRα molecular species in the hepatocyte that respond to inflammation. SUMOylation, a newly described post-translational modification of RXRα, appears to contribute to the inflammation-induced reduction of RXRα-regulated gene expression in the liver that affects core hepatic functions, including hepatobiliary transport.
Collapse
Affiliation(s)
- Rebecca Schneider Aguirre
- Department of Molecular and Cellular Biology (R.S.A., S.J.K.) and Texas Children’s Liver Center (S.J.K.), Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
47
|
Paul KB, Thompson JT, Simmons SO, Vanden Heuvel JP, Crofton KM. Evidence for triclosan-induced activation of human and rodent xenobiotic nuclear receptors. Toxicol In Vitro 2013; 27:2049-60. [PMID: 23899473 DOI: 10.1016/j.tiv.2013.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/05/2013] [Accepted: 07/19/2013] [Indexed: 10/26/2022]
Abstract
The bacteriostat triclosan (2,4,4'-trichloro-2'-hydroxydiphenylether) (TCS) decreases rat serum thyroxine via putative nuclear receptor (NR) interaction(s) and subsequent transcriptional up-regulation of hepatic catabolism and clearance. However, due to the evolutionary divergence of the constitutive androstane and pregnane-X receptors (CAR, PXR), TCS-mediated downstream effects may be species-dependent. To test the hypothesis that TCS activates xenobiotic NRs across species, cell-based NR reporter assays were employed to assess potential activation of rat, mouse, and human PXR, and rat, mouse, and three splice variants of human CAR. TCS activated hPXR, acted as an inverse agonist of hCAR1, and as a weak agonist of hCAR3. TCS failed to activate rPXR in full-length receptor reporter assays, and instead acted as a modest inverse agonist of rCAR. Consistent with the rat data, TCS also failed to activate mPXR and was a modest inverse agonist of mCAR. These data suggest that TCS may interact with multiple NRs, including hPXR, hCAR1, hCAR3, and rCAR in order to potentially affect hepatic catabolism. Overall these data support the conclusion that TCS may interact with NRs to regulate hepatic catabolism and downstream thyroid hormone homeostasis in both rat and human models, though perhaps by divergent mechanisms.
Collapse
Affiliation(s)
- Katie B Paul
- University of North Carolina at Chapel Hill, Curriculum in Toxicology, CB 7270, Chapel Hill, NC 27599, United States; Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, United States
| | | | | | | | | |
Collapse
|
48
|
Honma M, Kozawa M, Suzuki H. Methods for the quantitative evaluation and prediction of CYP enzyme induction using human in vitro systems. Expert Opin Drug Discov 2012; 5:491-511. [PMID: 22823132 DOI: 10.1517/17460441003762717] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD For successful drug development, it is important to investigate the potency of candidate drugs causing drug-drug interactions (DDI) during the early stages of development. The most common mechanisms of DDIs are the inhibition and induction of CYP enzymes. Therefore, it is important to develop co.mpounds with lower potencies for CYP enzyme induction. AREAS COVERED IN THIS REVIEW The aim of the present paper is to present an overview of the current knowledge of CYP induction mechanisms, particularly focusing on the transcriptional gene activation mediated by pregnane X receptor, aryl hydrocarbon receptor and constitutive androstane receptor. The adoptable options of in vitro assay methods for evaluating CYP induction are also summarized. Finally, we introduce a method for the quantitative prediction of CYP3A4 induction considering the turnover of CYP3A4 mRNA and protein in hepatocytes based on the data obtained from a reporter gene assay. WHAT THE READER WILL GAIN In order to predict in vivo CYP enzyme induction quantitatively based on in vitro information, an understanding of the physiological induction mechanisms and the features of each in vitro assay system is essential. We also present the estimation method of in vivo CYP induction potency of each compound based on the in vitro data which are routinely obtained but not necessarily utilized maximally in pharmaceutical companies. TAKE HOME MESSAGE It is desirable to select compounds with lower potencies for the inductive effect. For this purpose, an accurate prioritization procedure to evaluate the induction potency of each compound in a quantitative manner considering the pharmacologically effective concentration of each compound is necessary.
Collapse
Affiliation(s)
- Masashi Honma
- The University of Tokyo Hospital, Faculty of Medicine, Department of Pharmacy, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan +81 3 3815 5411 ; +81 3 3816 6159 ;
| | | | | |
Collapse
|
49
|
Wang YM, Ong SS, Chai SC, Chen T. Role of CAR and PXR in xenobiotic sensing and metabolism. Expert Opin Drug Metab Toxicol 2012; 8:803-17. [PMID: 22554043 DOI: 10.1517/17425255.2012.685237] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The xenobiotic detoxification system, which protects the human body from external chemicals, comprises drug-metabolizing enzymes and transporters whose expressions are regulated by pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). The progress made in a large number of recent studies calls for a timely review to summarize and highlight these key discoveries. AREAS COVERED This review summarizes recent advances in elucidating the roles of PXR and CAR in the xenobiotic detoxification system. It also highlights the progress in understanding the regulation of PXR and CAR activity at the post-translational levels, as well as the structural basis for the regulation of these two xenobiotic sensors. EXPERT OPINION Future efforts are needed to discover novel agonists and antagonists with species and isoform selectivity, to systematically understand the regulation of PXR and CAR at multiple levels (transcriptional, post-transcriptional and post-translational levels) in response to xenobiotics exposure, and to solve the structures of the full-length receptors, which will be enabled by improved protein expression and purification techniques and approaches. In addition, more efforts will be needed to validate PXR and CAR as disease-related therapeutic targets and thus expand their roles as master xenobiotic sensors.
Collapse
Affiliation(s)
- Yue-Ming Wang
- St. Jude Children's Research Hospital, Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | | | | |
Collapse
|
50
|
Yang D, Yang J, Shi D, Deng R, Yan B. Scoparone potentiates transactivation of the bile salt export pump gene and this effect is enhanced by cytochrome P450 metabolism but abolished by a PKC inhibitor. Br J Pharmacol 2012; 164:1547-57. [PMID: 21649640 DOI: 10.1111/j.1476-5381.2011.01522.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Hyperbilirubinaemia and cholestasis are two major forms of liver abnormality. The Chinese herb Yin Chin has been used for thousands of years to treat liver dysfunctions. In mice, this herb and its principal ingredient scoparone were found to accelerate the clearance of bilirubin accompanied by the induction of uridine diphosphate-5'-glucuronosyltransferase-1A1 (UGT1A1), a bilirubin processing enzyme. The aim of this study was to determine whether scoparone induces the expression of human UGT1A1. In addition, the expression of the bile salt export pump (BSEP), a transporter of bile acids, was determined. EXPERIMENTAL APPROACH Primary human hepatocytes and hepatoma line Huh7 were treated with scoparone, chenodeoxycholic acid (CDCA) or both. The expression of UGT1A1 and BSEP mRNA was determined. The activation of the human BSEP promoter reporter by scoparone was determined in Huh7 cells by transient transfection and in mice by bioluminescent imaging. The metabolism of scoparone was investigated by recombinant CYP enzymes and pooled human liver microsomes. KEY RESULTS Scoparone did not enhance the expression of either human BSEP or, surprisingly, UGT1A1. However, scoparone significantly potentiated the expression of BSEP induced by CDCA. Consistent with this, scoparone potentiated the stimulant effect of CDCA on the human BSEP promoter. This potentiation was enhanced by co-transfection of cytochrome P4501A2 but abolished by the PKC inhibitor GF109203X. CONCLUSIONS AND IMPLICATIONS Scoparone and Yin Chin normalize liver function primarily by enhancing the secretion of bile acids, and this effect probably varies depending on the metabolic rate of scoparone.
Collapse
Affiliation(s)
- Dongfang Yang
- Department of Biomedical Sciences, Center for Pharmacogenomics and Molecular Therapy, University of Rhode Island, Kingston, RI 02881, USA
| | | | | | | | | |
Collapse
|