101
|
Cave MC, Clair HB, Hardesty JE, Falkner KC, Feng W, Clark BJ, Sidey J, Shi H, Aqel BA, McClain CJ, Prough RA. Nuclear receptors and nonalcoholic fatty liver disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1083-1099. [PMID: 26962021 DOI: 10.1016/j.bbagrm.2016.03.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
Nuclear receptors are transcription factors which sense changing environmental or hormonal signals and effect transcriptional changes to regulate core life functions including growth, development, and reproduction. To support this function, following ligand-activation by xenobiotics, members of subfamily 1 nuclear receptors (NR1s) may heterodimerize with the retinoid X receptor (RXR) to regulate transcription of genes involved in energy and xenobiotic metabolism and inflammation. Several of these receptors including the peroxisome proliferator-activated receptors (PPARs), the pregnane and xenobiotic receptor (PXR), the constitutive androstane receptor (CAR), the liver X receptor (LXR) and the farnesoid X receptor (FXR) are key regulators of the gut:liver:adipose axis and serve to coordinate metabolic responses across organ systems between the fed and fasting states. Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease and may progress to cirrhosis and even hepatocellular carcinoma. NAFLD is associated with inappropriate nuclear receptor function and perturbations along the gut:liver:adipose axis including obesity, increased intestinal permeability with systemic inflammation, abnormal hepatic lipid metabolism, and insulin resistance. Environmental chemicals may compound the problem by directly interacting with nuclear receptors leading to metabolic confusion and the inability to differentiate fed from fasting conditions. This review focuses on the impact of nuclear receptors in the pathogenesis and treatment of NAFLD. Clinical trials including PIVENS and FLINT demonstrate that nuclear receptor targeted therapies may lead to the paradoxical dissociation of steatosis, inflammation, fibrosis, insulin resistance, dyslipidemia and obesity. Novel strategies currently under development (including tissue-specific ligands and dual receptor agonists) may be required to separate the beneficial effects of nuclear receptor activation from unwanted metabolic side effects. The impact of nuclear receptor crosstalk in NAFLD is likely to be profound, but requires further elucidation. 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)
- Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA; The KentuckyOne Health Jewish Hospital Liver Transplant Program, Louisville, KY 40202, USA.
| | - Heather B Clair
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Josiah E Hardesty
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - K Cameron Falkner
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Wenke Feng
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Barbara J Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jennifer Sidey
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Hongxue Shi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bashar A Aqel
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Scottsdale, AZ 85054, USA
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA; The KentuckyOne Health Jewish Hospital Liver Transplant Program, Louisville, KY 40202, USA
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
102
|
Sousa C, Andrade PB, Valentão P. Relationships of Echium plantagineum L. bee pollen, dietary flavonoids and their colonic metabolites with cytochrome P450 enzymes and oxidative stress. RSC Adv 2016. [DOI: 10.1039/c5ra26736f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Echium plantagineumL. bee pollen is a dietary source of flavonoids, which can play a protective role in the gastrointestinal tract by modulating cytochrome P450 (CYP) biotransformation enzymes and by opposing oxidative stress.
Collapse
Affiliation(s)
- C. Sousa
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| | - P. B. Andrade
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| | - P. Valentão
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
103
|
Scheer N, Kapelyukh Y, Rode A, Oswald S, Busch D, McLaughlin LA, Lin D, Henderson CJ, Wolf CR. Defining Human Pathways of Drug Metabolism In Vivo through the Development of a Multiple Humanized Mouse Model. Drug Metab Dispos 2015; 43:1679-90. [PMID: 26265742 DOI: 10.1124/dmd.115.065656] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/10/2015] [Indexed: 11/22/2022] Open
Abstract
Variability in drug pharmacokinetics is a major factor in defining drug efficacy and side effects. There remains an urgent need, particularly with the growing use of polypharmacy, to obtain more informative experimental data predicting clinical outcomes. Major species differences in multiplicity, substrate specificity, and regulation of enzymes from the cytochrome P450-dependent mono-oxygenase system play a critical role in drug metabolism. To develop an in vivo model for predicting human responses to drugs, we generated a mouse, where 31 P450 genes from the Cyp2c, Cyp2d, and Cyp3a gene families were exchanged for their relevant human counterparts. The model has been improved through additional humanization for the nuclear receptors constitutive androgen receptor and pregnane X receptor that control the expression of key drug metabolizing enzymes and transporters. In this most complex humanized mouse model reported to date, the cytochromes P450 function as predicted and we illustrate how these mice can be applied to predict drug-drug interactions in humans.
Collapse
Affiliation(s)
- Nico Scheer
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Yury Kapelyukh
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Anja Rode
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Stefan Oswald
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Diana Busch
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Lesley A McLaughlin
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - De Lin
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Colin J Henderson
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - C Roland Wolf
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| |
Collapse
|
104
|
Mellor CL, Steinmetz FP, Cronin MTD. The identification of nuclear receptors associated with hepatic steatosis to develop and extend adverse outcome pathways. Crit Rev Toxicol 2015; 46:138-52. [PMID: 26451809 DOI: 10.3109/10408444.2015.1089471] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of adverse outcome pathways (AOPs) is becoming a key component of twenty-first century toxicology. AOPs provide a conceptual framework that links the molecular initiating event to an adverse outcome through organized toxicological knowledge, bridging the gap from chemistry to toxicological effect. As nuclear receptors (NRs) play essential roles for many physiological processes within the body, they are used regularly as drug targets for therapies to treat many diseases including diabetes, cancer and neurodegenerative diseases. Due to the heightened development of NR ligands, there is increased need for the identification of related AOPs to facilitate their risk assessment. Many NR ligands have been linked specifically to steatosis. This article reviews and summarizes the role of NR and their importance with links between NR examined to identify plausible putative AOPs. The following NRs are shown to induce hepatic steatosis upon ligand binding: aryl hydrocarbon receptor, constitutive androstane receptor, oestrogen receptor, glucocorticoid receptor, farnesoid X receptor, liver X receptor, peroxisome proliferator-activated receptor, pregnane X receptor and the retinoic acid receptor. A preliminary, putative AOP was formed for NR binding linked to hepatic steatosis as the adverse outcome.
Collapse
Affiliation(s)
- Claire L Mellor
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| | - Fabian P Steinmetz
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| | - Mark T D Cronin
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| |
Collapse
|
105
|
Richter I, Fidler AE. Tunicate pregnane X receptor (PXR) orthologs: Transcript characterization and natural variation. Mar Genomics 2015; 23:99-108. [DOI: 10.1016/j.margen.2015.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 12/12/2022]
|
106
|
Cain-Hom C, Pabalate R, Pham A, Patel HN, Wiler R, Cox JC. Mammalian Genotyping Using Acoustic Droplet Ejection for Enhanced Data Reproducibility, Superior Throughput, and Minimized Cross-Contamination. ACTA ACUST UNITED AC 2015; 21:37-48. [PMID: 26311060 DOI: 10.1177/2211068215601637] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Indexed: 12/20/2022]
Abstract
Genetically engineered animal models are major tools of a drug discovery pipeline because they facilitate understanding of the molecular and biochemical basis of disease. These highly complex models of human disease often require increasingly convoluted genetic analysis. With growing needs for throughput and consistency, we find that traditional aspiration-and-dispense liquid-handling robots no longer have the required speed, quality, or reproducibility.We present an adaptation and installation of an acoustic droplet ejection (ADE) liquid-handling system for ultra-high-throughput screening of genetically engineered models. An ADE system is fully integrated with existing laboratory processes and platforms to facilitate execution of PCR and quantitative PCR (qPCR) reactions. Such a configuration permits interrogation of highly complex genetic models in a variety of backgrounds. Our findings demonstrate that a single ADE system replaces 8-10 traditional liquid-handling robots while increasing quality and reproducibility.We demonstrate significant improvements achieved by transitioning to an ADE device: extremely low detectable cross-contamination in PCR and qPCR despite extensive use, greatly increased data reproducibility (large increases in data quality and Cq consistency), lowered reaction volumes for large cost savings, and nearly a magnitude increase in speed per instrument. We show several comparisons between traditional- and ADE-based pipetting for a qPCR-based workflow.
Collapse
Affiliation(s)
- Carol Cain-Hom
- Department of Transgenic Technology, Genentech Inc., San Francisco, CA, USA
| | - Ryan Pabalate
- Department of Transgenic Technology, Genentech Inc., San Francisco, CA, USA
| | - Anna Pham
- Department of Transgenic Technology, Genentech Inc., San Francisco, CA, USA
| | - Hetal N Patel
- Department of Transgenic Technology, Genentech Inc., San Francisco, CA, USA
| | - Rhonda Wiler
- Department of Transgenic Technology, Genentech Inc., San Francisco, CA, USA
| | - J Colin Cox
- Department of Transgenic Technology, Genentech Inc., San Francisco, CA, USA
| |
Collapse
|
107
|
Wang YM, Chai SC, Lin W, Chai X, Elias A, Wu J, Ong SS, Pondugula SR, Beard JA, Schuetz EG, Zeng S, Xie W, Chen T. Serine 350 of human pregnane X receptor is crucial for its heterodimerization with retinoid X receptor alpha and transactivation of target genes in vitro and in vivo. Biochem Pharmacol 2015; 96:357-68. [PMID: 26119819 DOI: 10.1016/j.bcp.2015.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022]
Abstract
The human pregnane X receptor (hPXR), a member of the nuclear receptor superfamily, senses xenobiotics and controls the transcription of genes encoding drug-metabolizing enzymes and transporters. The regulation of hPXR's transcriptional activation of its target genes is important for xenobiotic detoxification and endobiotic metabolism, and hPXR dysregulation can cause various adverse drug effects. Studies have implicated the putative phosphorylation site serine 350 (Ser(350)) in regulating hPXR transcriptional activity, but the mechanism of regulation remains elusive. Here we investigated the transactivation of hPXR target genes in vitro and in vivo by hPXR with a phosphomimetic mutation at Ser(350) (hPXR(S350D)). The S350D phosphomimetic mutation reduced the endogenous expression of cytochrome P450 3A4 (an hPXR target gene) in HepG2 and LS180 cells. Biochemical assays and structural modeling revealed that Ser(350) of hPXR is crucial for formation of the hPXR-retinoid X receptor alpha (RXRα) heterodimer. The S350D mutation abrogated heterodimerization in a ligand-independent manner, impairing hPXR-mediated transactivation. Further, in a novel humanized transgenic mouse model expressing the hPXR(S350D) transgene, we demonstrated that the S350D mutation alone is sufficient to impair hPXR transcriptional activity in mouse liver. This transgenic mouse model provides a unique tool to investigate the regulation and function of hPXR, including its non-genomic function, in vivo. Our finding that phosphorylation regulates hPXR activity has implications for development of novel hPXR antagonists and for safety evaluation during drug development.
Collapse
Affiliation(s)
- Yue-Ming Wang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiaojuan Chai
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ayesha Elias
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Su Sien Ong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Satyanarayana R Pondugula
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jordan A Beard
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Erin G Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Su Zeng
- Department of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
108
|
Sahasrabudhe V, Zhu T, Vaz A, Tse S. Drug Metabolism and Drug Interactions: Potential Application to Antituberculosis Drugs. J Infect Dis 2015; 211 Suppl 3:S107-14. [DOI: 10.1093/infdis/jiv009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
109
|
Finkbeiner S, Frumkin M, Kassner PD. Cell-based screening: extracting meaning from complex data. Neuron 2015; 86:160-74. [PMID: 25856492 PMCID: PMC4457442 DOI: 10.1016/j.neuron.2015.02.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 09/23/2014] [Accepted: 01/22/2015] [Indexed: 01/23/2023]
Abstract
Unbiased discovery approaches have the potential to uncover neurobiological insights into CNS disease and lead to the development of therapies. Here, we review lessons learned from imaging-based screening approaches and recent advances in these areas, including powerful new computational tools to synthesize complex data into more useful knowledge that can reliably guide future research and development.
Collapse
Affiliation(s)
- Steven Finkbeiner
- Director of the Taube/Koret Center for Neurodegenerative Disease and the Hellman Family Foundation Program in Alzheimer's Disease Research, Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Michael Frumkin
- Director of Engineering, Research, Google, Inc., 1600 Amphitheatre Parkway, Mountain View, CA 94043, USA
| | - Paul D Kassner
- Director of Research, Amgen, Inc., 1120 Veterans Boulevard South, San Francisco, CA 94080, USA
| |
Collapse
|
110
|
Gonzalez FJ, Fang ZZ, Ma X. Transgenic mice and metabolomics for study of hepatic xenobiotic metabolism and toxicity. Expert Opin Drug Metab Toxicol 2015; 11:869-81. [PMID: 25836352 DOI: 10.1517/17425255.2015.1032245] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The study of xenobiotic metabolism and toxicity has been greatly aided by the use of genetically modified mouse models and metabolomics. AREAS COVERED Gene knockout mice can be used to determine the enzymes responsible for the metabolism of xenobiotics in vivo and to examine the mechanisms of xenobiotic-induced toxicity. Humanized mouse models are especially important because there exist marked species differences in the xenobiotic-metabolizing enzymes and the nuclear receptors that regulate these enzymes. Humanized mice expressing CYPs and nuclear receptors including the pregnane X receptor, the major regulator of xenobiotic metabolism and transport were produced. With genetically modified mouse models, metabolomics can determine the metabolic map of many xenobiotics with a level of sensitivity that allows the discovery of even minor metabolites. This technology can be used for determining the mechanism of xenobiotic toxicity and to find early biomarkers for toxicity. EXPERT OPINION Metabolomics and genetically modified mouse models can be used for the study of xenobiotic metabolism and toxicity by: i) comparison of the metabolomics profiles between wild-type and genetically modified mice, and searching for genotype-dependent endogenous metabolites; ii) searching for and elucidating metabolites derived from xenobiotics; and iii) discovery of specific alterations of endogenous compounds induced by xenobiotics-induced toxicity.
Collapse
Affiliation(s)
- Frank J Gonzalez
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Metabolism , Bethesda, MD 20892 , USA +1 301 496 9067 ; +1 301 496 8419 ;
| | | | | |
Collapse
|
111
|
Li L, Li H, Garzel B, Yang H, Sueyoshi T, Li Q, Shu Y, Zhang J, Hu B, Heyward S, Moeller T, Xie W, Negishi M, Wang H. SLC13A5 is a novel transcriptional target of the pregnane X receptor and sensitizes drug-induced steatosis in human liver. Mol Pharmacol 2015; 87:674-82. [PMID: 25628225 PMCID: PMC4366797 DOI: 10.1124/mol.114.097287] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/26/2015] [Indexed: 12/20/2022] Open
Abstract
The solute carrier family 13 member 5 (SLC13A5) is a sodium-coupled transporter that mediates cellular uptake of citrate, which plays important roles in the synthesis of fatty acids and cholesterol. Recently, the pregnane X receptor (PXR, NR1I2), initially characterized as a xenobiotic sensor, has been functionally linked to the regulation of various physiologic processes that are associated with lipid metabolism and energy homeostasis. Here, we show that the SLC13A5 gene is a novel transcriptional target of PXR, and altered expression of SLC13A5 affects lipid accumulation in human liver cells. The prototypical PXR activator rifampicin markedly induced the mRNA and protein expression of SLC13A5 in human primary hepatocytes. Utilizing cell-based luciferase reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays, we identified and functionally characterized two enhancer modules located upstream of the SLC13A5 gene transcription start site that are associated with regulation of PXR-mediated SLC13A5 induction. Functional analysis further revealed that rifampicin can enhance lipid accumulation in human primary hepatocytes, and knockdown of SLC13A5 expression alone leads to a significant decrease of the lipid content in HepG2 cells. Overall, our results uncover SLC13A5 as a novel target gene of PXR and may contribute to drug-induced steatosis and metabolic disorders in humans.
Collapse
Affiliation(s)
- Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Haishan Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Brandy Garzel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Hui Yang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Tatsuya Sueyoshi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Qing Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Yan Shu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Junran Zhang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Bingfang Hu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Timothy Moeller
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Wen Xie
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Masahiko Negishi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| |
Collapse
|
112
|
Kliewer SA. Nuclear receptor PXR: discovery of a pharmaceutical anti-target. J Clin Invest 2015; 125:1388-9. [PMID: 25831443 DOI: 10.1172/jci81244] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Transcriptional induction of the gene encoding cytochrome P450 3A oxygenase (CYP3A) causes a prominent class of dangerous drug-drug interactions wherein one drug accelerates the metabolism of another. In our 1998 JCI paper, we reported the cloning of the human nuclear receptor PXR and demonstrated that it mediates CYP3A induction. We determined that PXR is expressed in liver, acts through a DNA response element located in the CYP3A promoter, and is activated by a structurally diverse collection of drugs that induce CYP3A. Our findings revealed the molecular basis for the CYP3A induction class of drug-drug interactions and provided a high-throughput means for screening out drug candidates with this activity.
Collapse
|
113
|
Lee SY, Lee JY, Kim YM, Kim SK, Oh SJ. Expression of hepatic cytochrome P450s and UDP-glucuronosyltransferases in PXR and CAR double humanized mice treated with rifampicin. Toxicol Lett 2015; 235:107-15. [PMID: 25835148 DOI: 10.1016/j.toxlet.2015.03.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 01/23/2023]
Abstract
Nuclear receptor humanized mice models have been developed to predict regulation of drug metabolizing enzyme by xenobiotics. However, limited information is available concerning xenobiotic-induced regulation of drug metabolizing enzymes in multiple nuclear receptor humanized mice. The present study investigated the hepatic regulation of cytochrome P450s (CYPs) and UDP-glucuronosyltransferases (UGTs) in the pregnane X receptor (PXR) and the constitutive androstane receptor double humanized mice treated with rifampicin (RIF; 10mg/kg) for 4 days. RIF increased hepatic microsomal protein and total CYP contents, and CYP reductase activity in the humanized mice, but not in normal mice. Moreover, hepatic induction of Cyp2b10, Cyp2c, and Cyp3a11 were observed only in the RIF-treated humanized mice, suggesting that the humanized mice are sensitive to RIF with respect to the regulation of the hepatic CYP system. Hepatic UGT activities using estradiol, serotonin, and mefenamic acid, but not chenodeoxycholic acid as substrates, increased in the RIF-treated humanized mice, and the glucuronidation activities of estradiol and chenodeoxycholic acid increased in RIF-treated normal mice. These results raise the possibility that a PXR-independent mechanism may be involved in hepatic regulation of UGTs by RIF.
Collapse
Affiliation(s)
- Sang Yoon Lee
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Ji-Yoon Lee
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Young-Mi Kim
- College of Pharmacy, Hanyang University, Ansan, Gyeonggido 426-791 Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Republic of Korea.
| | - Soo Jin Oh
- Bio-Evaluation Center, KRIBB, 685-1 Yangcheong-ri, Ochang-eup, Cheongwon-gun, Chungbuk 363-883, Republic of Korea.
| |
Collapse
|
114
|
Azuma K, Casey SC, Urano T, Horie-Inoue K, Ouchi Y, Blumberg B, Inoue S. Pregnane X receptor knockout mice display aging-dependent wearing of articular cartilage. PLoS One 2015; 10:e0119177. [PMID: 25749104 PMCID: PMC4352085 DOI: 10.1371/journal.pone.0119177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/10/2015] [Indexed: 11/24/2022] Open
Abstract
Steroid and xenobiotic receptor (SXR) and its murine ortholog, pregnane X receptor (PXR), are nuclear receptors that are expressed at high levels in the liver and the intestine where they function as xenobiotic sensors that induce expression of genes involved in detoxification and drug excretion. Recent evidence showed that SXR and PXR are also expressed in bone tissue where they mediate bone metabolism. Here we report that systemic deletion of PXR results in aging-dependent wearing of articular cartilage of knee joints. Histomorphometrical analysis showed remarkable reduction of width and an enlarged gap between femoral and tibial articular cartilage in PXR knockout mice. We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Lastly, we demonstrated the biological significance of Fam20a expression in chondrocytes by evaluating osteoarthritis-related gene expression of primary articular chondrocytes. Consistent with epidemiological findings, our results indicate that SXR/PXR protects against aging-dependent wearing of articular cartilage and that ligands for SXR/PXR have potential role in preventing osteoarthritis caused by aging.
Collapse
Affiliation(s)
- Kotaro Azuma
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Stephanie C. Casey
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, United States of America
| | - Tomohiko Urano
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Anti-aging Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Yasuyoshi Ouchi
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, United States of America
| | - Satoshi Inoue
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Anti-aging Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| |
Collapse
|
115
|
Cheng Q, Inaba Y, Lu P, Xu M, He J, Zhao Y, Guo GL, Kuruba R, de la Vega R, Evans RW, Li S, Xie W. Chronic activation of FXR in transgenic mice caused perinatal toxicity and sensitized mice to cholesterol toxicity. Mol Endocrinol 2015; 29:571-82. [PMID: 25719402 DOI: 10.1210/me.2014-1337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The nuclear receptor farnesoid X receptor (FXR) (nuclear receptor subfamily 1, group H, member 4, or NR1H4) is highly expressed in the liver and intestine. Previous reports have suggested beneficial functions of FXR in the homeostasis of bile acids, lipids, and glucose, as well as in promoting liver regeneration and inhibiting carcinogenesis. To investigate the effect of chronic FXR activation in vivo, we generated transgenic mice that conditionally and tissue specifically express the activated form of FXR in the liver and intestine. Unexpectedly, the transgenic mice showed several intriguing phenotypes, including partial neonatal lethality, growth retardation, and spontaneous liver toxicity. The transgenic mice also displayed heightened sensitivity to a high-cholesterol diet-induced hepatotoxicity but resistance to the gallstone formation. The phenotypes were transgene specific, because they were abolished upon treatment with doxycycline to silence the transgene expression. The perinatal toxicity, which can be rescued by a maternal vitamin supplement, may have resulted from vitamin deficiency due to low biliary bile acid output as a consequence of inhibition of bile acid formation. Our results also suggested that the fibroblast growth factor-inducible immediate-early response protein 14 (Fn14), a member of the proinflammatory TNF family, is a FXR-responsive gene. However, the contribution of Fn14 induction in the perinatal toxic phenotype of the transgenic mice remains to be defined. Because FXR is being explored as a therapeutic target, our results suggested that a chronic activation of this nuclear receptor may have an unintended side effect especially during the perinatal stage.
Collapse
Affiliation(s)
- Qiuqiong Cheng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Q.C., Y.I., P.L., M.X., J.H., Y.Z., R.K., S.L., W.X.), and Departments of Epidemiology (R.D.L.V., R.W.E.) and Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; and Department of Pharmacology and Toxicology (G.L.G.), Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Kubota A, Goldstone JV, Lemaire B, Takata M, Woodin BR, Stegeman JJ. Role of pregnane X receptor and aryl hydrocarbon receptor in transcriptional regulation of pxr, CYP2, and CYP3 genes in developing zebrafish. Toxicol Sci 2015; 143:398-407. [PMID: 25424564 PMCID: PMC4306721 DOI: 10.1093/toxsci/kfu240] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Ligand-activated receptors regulate numerous genes, and mediate effects of a broad set of endogenous and exogenous chemicals in vertebrates. Understanding the roles of these transcription factors in zebrafish (Danio rerio) is important to the use of this non-mammalian model in toxicological, pharmacological, and carcinogenesis research. Response to a potential agonist for the pregnane X receptor (Pxr) [pregnenolone (PN)] was examined in developing zebrafish, to assess involvement of Pxr in regulation of selected genes, including genes in cytochrome P450 subfamilies CYP2 and CYP3. We also examined interaction of Pxr and the aryl hydrocarbon receptor (Ahr) signaling pathways. Pregnenolone caused a dose-dependent increase in mRNA levels of pxr, ahr2, CYP1A, CYP2AA1, CYP2AA12, CYP3A65, and CYP3C1, most of which peaked at 3 µM PN. The well-known Ahr agonist 3,3',4,4',5-pentachlorobiphenyl (PCB126) also upregulated expression of pxr, ahr2, CYP1A, CYP2AA12, CYP3A65, and CYP3C1 in a dose-dependent manner. Inhibition of pxr translation by morpholino antisense oligonucleotides (MO) suppressed PN-induced expression of pxr, ahr2, CYP3A65, and CYP3C1 genes. Levels of CYP2AA1 and CYP2AA12 mRNA were increased in the control-MO group exposed to PN; this was prevented by knocking down Pxr. Similarly, Ahr2-MO treatment blocked PCB126-induced mRNA expression of pxr, CYP1A, CYP2AA12, CYP3A65, and CYP3C1. The present study shows self-regulation of pxr by PN in developing zebrafish. Selected zebrafish CYP1, CYP2 (including several CYP2AAs) and CYP3 genes appear to be under the regulation of both Pxr and Ahr2.
Collapse
Affiliation(s)
- Akira Kubota
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543
| | - Jared V Goldstone
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543
| | - Benjamin Lemaire
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543
| | - Matthew Takata
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543
| | - Bruce R Woodin
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543
| | - John J Stegeman
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543
| |
Collapse
|
117
|
Prakash C, Zuniga B, Song CS, Jiang S, Cropper J, Park S, Chatterjee B. Nuclear Receptors in Drug Metabolism, Drug Response and Drug Interactions. NUCLEAR RECEPTOR RESEARCH 2015; 2:101178. [PMID: 27478824 PMCID: PMC4963026 DOI: 10.11131/2015/101178] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Orally delivered small-molecule therapeutics are metabolized in the liver and intestine by phase I and phase II drug-metabolizing enzymes (DMEs), and transport proteins coordinate drug influx (phase 0) and drug/drug-metabolite efflux (phase III). Genes involved in drug metabolism and disposition are induced by xenobiotic-activated nuclear receptors (NRs), i.e. PXR (pregnane X receptor) and CAR (constitutive androstane receptor), and by the 1α, 25-dihydroxy vitamin D3-activated vitamin D receptor (VDR), due to transactivation of xenobiotic-response elements (XREs) present in phase 0-III genes. Additional NRs, like HNF4-α, FXR, LXR-α play important roles in drug metabolism in certain settings, such as in relation to cholesterol and bile acid metabolism. The phase I enzymes CYP3A4/A5, CYP2D6, CYP2B6, CYP2C9, CYP2C19, CYP1A2, CYP2C8, CYP2A6, CYP2J2, and CYP2E1 metabolize >90% of all prescription drugs, and phase II conjugation of hydrophilic functional groups (with/without phase I modification) facilitates drug clearance. The conjugation step is mediated by broad-specificity transferases like UGTs, SULTs, GSTs. This review delves into our current understanding of PXR/CAR/VDR-mediated regulation of DME and transporter expression, as well as effects of single nucleotide polymorphism (SNP) and epigenome (specified by promoter methylation, histone modification, microRNAs, long non coding RNAs) on the expression of PXR/CAR/VDR and phase 0-III mediators, and their impacts on variable drug response. Therapeutic agents that target epigenetic regulation and the molecular basis and consequences (overdosing, underdosing, or beneficial outcome) of drug-drug/drug-food/drug-herb interactions are also discussed. Precision medicine requires understanding of a drug's impact on DME and transporter activity and their NR-regulated expression in order to achieve optimal drug efficacy without adverse drug reactions. In future drug screening, new tools such as humanized mouse models and microfluidic organs-on-chips, which mimic the physiology of a multicellular environment, will likely replace the current cell-based workflow.
Collapse
Affiliation(s)
- Chandra Prakash
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- William Carey University College of Osteopathic Medicine, 498 Tucsan Ave, Hattiesburg, Mississipi 39401
| | - Baltazar Zuniga
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- University of Texas at Austin, 2100 Comal Street, Austin, Texas 78712
| | - Chung Seog Song
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Shoulei Jiang
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Jodie Cropper
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Sulgi Park
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Bandana Chatterjee
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- South Texas Veterans Health Care System, Audie L Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, Texas 78229
| |
Collapse
|
118
|
Developmental toxicity of perfluorononanoic acid in mice. Reprod Toxicol 2015; 51:133-44. [DOI: 10.1016/j.reprotox.2014.12.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 11/22/2014] [Accepted: 12/16/2014] [Indexed: 01/12/2023]
|
119
|
Richter I, Fidler AE. Marine invertebrate xenobiotic-activated nuclear receptors: their application as sensor elements in high-throughput bioassays for marine bioactive compounds. Mar Drugs 2014; 12:5590-618. [PMID: 25421319 PMCID: PMC4245547 DOI: 10.3390/md12115590] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 10/31/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023] Open
Abstract
Developing high-throughput assays to screen marine extracts for bioactive compounds presents both conceptual and technical challenges. One major challenge is to develop assays that have well-grounded ecological and evolutionary rationales. In this review we propose that a specific group of ligand-activated transcription factors are particularly well-suited to act as sensors in such bioassays. More specifically, xenobiotic-activated nuclear receptors (XANRs) regulate transcription of genes involved in xenobiotic detoxification. XANR ligand-binding domains (LBDs) may adaptively evolve to bind those bioactive, and potentially toxic, compounds to which organisms are normally exposed to through their specific diets. A brief overview of the function and taxonomic distribution of both vertebrate and invertebrate XANRs is first provided. Proof-of-concept experiments are then described which confirm that a filter-feeding marine invertebrate XANR LBD is activated by marine bioactive compounds. We speculate that increasing access to marine invertebrate genome sequence data, in combination with the expression of functional recombinant marine invertebrate XANR LBDs, will facilitate the generation of high-throughput bioassays/biosensors of widely differing specificities, but all based on activation of XANR LBDs. Such assays may find application in screening marine extracts for bioactive compounds that could act as drug lead compounds.
Collapse
Affiliation(s)
- Ingrid Richter
- Environmental Technology Group, Cawthron Institute, Private Bag 2, Nelson 7012, New Zealand.
| | - Andrew E Fidler
- Environmental Technology Group, Cawthron Institute, Private Bag 2, Nelson 7012, New Zealand.
| |
Collapse
|
120
|
Li MM, Wang WP, Wu WJ, Huang M, Yu AM. Rapid production of novel pre-microRNA agent hsa-mir-27b in Escherichia coli using recombinant RNA technology for functional studies in mammalian cells. Drug Metab Dispos 2014; 42:1791-5. [PMID: 25161167 PMCID: PMC4201134 DOI: 10.1124/dmd.114.060145] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/25/2014] [Indexed: 01/24/2023] Open
Abstract
Noncoding microRNAs (miRNAs or miRs) have been revealed as critical epigenetic factors in the regulation of various cellular processes, including drug metabolism and disposition. However, research on miRNA functions is limited to the use of synthetic RNA and recombinant DNA agents. Herein, we show that novel pre-miRNA-27b (miR-27b) agents can be biosynthesized in Escherichia coli using recombinant RNA technology, and recombinant transfer RNA (tRNA)/mir-27b chimera was readily purified to a high degree of homogeneity (>95%) using anion-exchange fast protein liquid chromatography. The tRNA-fusion miR-27b was revealed to be processed to mature miRNA miR-27b in human carcinoma LS-180 cells in a dose- and time-dependent manner. Moreover, recombinant tRNA/miR-27b agents were biologically active in reducing the mRNA and protein expression levels of cytochrome P450 3A4 (CYP3A4), which consequently led to lower midazolam 1'-hydroxylase activity. These findings demonstrate that pre-miRNA agents can be produced by recombinant RNA technology for functional studies.
Collapse
Affiliation(s)
- Mei-Mei Li
- Department of Biochemistry and Molecular Medicine, University of California Davis Medical Center, Sacramento, California (M.-M.L., W.-P.W., W.-J.W., A.-M.Y.); Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China (M.-M.L., M.H.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W.)
| | - Wei-Peng Wang
- Department of Biochemistry and Molecular Medicine, University of California Davis Medical Center, Sacramento, California (M.-M.L., W.-P.W., W.-J.W., A.-M.Y.); Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China (M.-M.L., M.H.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W.)
| | - Wen-Juan Wu
- Department of Biochemistry and Molecular Medicine, University of California Davis Medical Center, Sacramento, California (M.-M.L., W.-P.W., W.-J.W., A.-M.Y.); Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China (M.-M.L., M.H.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W.)
| | - Min Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis Medical Center, Sacramento, California (M.-M.L., W.-P.W., W.-J.W., A.-M.Y.); Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China (M.-M.L., M.H.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California Davis Medical Center, Sacramento, California (M.-M.L., W.-P.W., W.-J.W., A.-M.Y.); Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China (M.-M.L., M.H.); and Center of Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China (W.-P.W.)
| |
Collapse
|
121
|
Chen J, Zhao KN, Chen C. The role of CYP3A4 in the biotransformation of bile acids and therapeutic implication for cholestasis. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:7. [PMID: 25332983 DOI: 10.3978/j.issn.2305-5839.2013.03.02] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/11/2013] [Indexed: 12/23/2022]
Abstract
CYP3A4 is a major cytochrome P450. It catalyses a broad range of substrates including xenobiotics such as clinically used drugs and endogenous compounds bile acids. Its function to detoxify bile acids could be used for treating cholestasis, which is a condition characterised by accumulation of bile acids. Although bile acids have important physiological functions, they are very toxic when their concentrations are excessively high. The accumulated bile acids in cholestasis can cause liver and other tissue injuries. Thus, control of the concentrations of bile acids is critical for treatment of cholestasis. CYP3A4 is responsively upregulated in cholestasis mediated by the nuclear receptors farnesol X receptor (FXR) and pregnane X receptor (PXR) as a defence mechanism. However, the regulation of CYP3A4 is complicated by estrogen, which is increased in cholestasis and down regulates CYP3A4 expression. The activity of CYP3A4 is also inhibited by accumulated bile acids due to their property of detergent effect. In some cholestasis cases, genetic polymorphisms of the CYP3A4 and PXR genes may interfere with the adaptive response. Further stimulation of CYP3A4 activity in cholestasis could be an effective approach for treatment of the disease. In this review, we summarise recent progress about the roles of CYP3A4 in the metabolism of bile acids, its regulation and possible implication in the treatment of cholestasis.
Collapse
Affiliation(s)
- Jiezhong Chen
- 1 School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia ; 2 Centre for Kidney Disease-Venomics Research, School of Medicine, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Kong-Nan Zhao
- 1 School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia ; 2 Centre for Kidney Disease-Venomics Research, School of Medicine, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Chen Chen
- 1 School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia ; 2 Centre for Kidney Disease-Venomics Research, School of Medicine, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD 4102, Australia
| |
Collapse
|
122
|
Xu S, Xiao Y, Li L, Yu L, Jiang H, Yu A, Zeng S. Three new shRNA expression vectors targeting the CYP3A4 coding sequence to inhibit its expression. Acta Pharm Sin B 2014; 4:350-7. [PMID: 26579404 PMCID: PMC4629097 DOI: 10.1016/j.apsb.2014.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/24/2014] [Accepted: 08/13/2014] [Indexed: 01/01/2023] Open
Abstract
RNA interference (RNAi) is useful for selective gene silencing. Cytochrome P450 3A4 (CYP3A4), which metabolizes approximately 50% of drugs in clinical use, plays an important role in drug metabolism. In this study, we aimed to develop a short hairpin RNA (shRNA) to modulate CYP3A4 expression. Three new shRNAs (S1, S2 and S3) were designed to target the coding sequence (CDS) of CYP3A4, cloned into a shRNA expression vector, and tested in different cells. The mixture of three shRNAs produced optimal reduction (55%) in CYP3A4 CDS-luciferase activity in both CHL and HEK293 cells. Endogenous CYP3A4 expression in HepG2 cells was decreased about 50% at both mRNA and protein level after transfection of the mixture of three shRNAs. In contrast, CYP3A5 gene expression was not altered by the shRNAs, supporting the selectivity of CYP3A4 shRNAs. In addition, HepG2 cells transfected with CYP3A4 shRNAs were less sensitive to Ginkgolic acids, whose toxic metabolites are produced by CYP3A4. These results demonstrate that vector-based shRNAs could modulate CYP3A4 expression in cells through their actions on CYP3A4 CDS, and CYP3A4 shRNAs may be utilized to define the role of CYP3A4 in drug metabolism and toxicity.
Collapse
Affiliation(s)
- Siyun Xu
- Department of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongsheng Xiao
- Department of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Li
- Department of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lushan Yu
- Department of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huidi Jiang
- Department of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Aiming Yu
- Department of Biochemistry & Molecular Medicine, UC-Davis Medical Center, Sacramento, CA 95817, USA
| | - Su Zeng
- Department of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Corresponding author. Tel./fax: +86 571 88208407.
| |
Collapse
|
123
|
Jaiswal S, Sharma A, Shukla M, Vaghasiya K, Rangaraj N, Lal J. Novel pre-clinical methodologies for pharmacokinetic drug-drug interaction studies: spotlight on "humanized" animal models. Drug Metab Rev 2014; 46:475-93. [PMID: 25270219 DOI: 10.3109/03602532.2014.967866] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Poly-therapy is common due to co-occurrence of several ailments in patients, leading to the elevated possibility of drug-drug interactions (DDI). Pharmacokinetic DDI often accounts for severe adverse drug reactions in patients resulting in withdrawal of drug from the market. Hence, the prediction of DDI is necessary at pre-clinical stage of drug development. Several human tissue and cell line-based in vitro systems are routinely used for screening metabolic and transporter pathways of investigational drugs and for predicting their clinical DDI potentials. However, ample constraints are associated with the in vitro systems and sometimes in vitro-in vivo extrapolation (IVIVE) fail to assess the risk of DDI in clinic. In vitro-in vivo correlation model in animals combined with human in vitro studies may be helpful in better prediction of clinical outcome. Native animal models vary remarkably from humans in drug metabolizing enzymes and transporters, hence, the interpretation of results from animal DDI studies is difficult. With the advent of modern molecular biology and engineering tools, novel pre-clinical animal models, namely, knockout rat/mouse, transgenic rat/mouse with humanized drug metabolizing enzymes and/or transporters and chimeric rat/mouse with humanized liver are developed. These models nearly simulate human-like drug metabolism and help to validate the in vivo relevance of the in vitro human DDI data. This review briefly discusses the application of such novel pre-clinical models for screening various type of DDI along with their advantages and limitations.
Collapse
Affiliation(s)
- Swati Jaiswal
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute , Lucknow , India
| | | | | | | | | | | |
Collapse
|
124
|
Wang YM, Chai SC, Brewer CT, Chen T. Pregnane X receptor and drug-induced liver injury. Expert Opin Drug Metab Toxicol 2014; 10:1521-32. [PMID: 25252616 DOI: 10.1517/17425255.2014.963555] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The liver plays a central role in transforming and clearing foreign substances. The continuous exposure of the liver to xenobiotics sometimes leads to impaired liver function, referred to as drug-induced liver injury (DILI). The pregnane X receptor (PXR) tightly regulates the expression of genes in the hepatic drug-clearance system and its undesired activation plays a role in DILI. AREAS COVERED This review focuses on the recent progress in understanding PXR-mediated DILI and highlights the efforts made to assess and manage PXR-mediated DILI during drug development. EXPERT OPINION Future efforts are needed to further elucidate the mechanisms of PXR-mediated liver injury, including the epigenetic regulation and polymorphisms of PXR. Novel in vitro models containing functional PXR could improve our ability to predict and assess DILI during drug development. PXR inhibitors may provide chemical tools to validate the potential of PXR as a therapeutic target and to develop drugs to be used in the clinic to manage PXR-mediated DILI.
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
|
125
|
LeBaron MJ, Rasoulpour RJ, Gollapudi BB, Sura R, Kan HL, Schisler MR, Pottenger LH, Papineni S, Eisenbrandt DL. Characterization of Nuclear Receptor-Mediated Murine Hepatocarcinogenesis of the Herbicide Pronamide and Its Human Relevance. Toxicol Sci 2014; 142:74-92. [DOI: 10.1093/toxsci/kfu155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
126
|
Gao J, Zhang Y, Yang Y, Yuan C, Qin F, Liu S, Zheng Y, Wang Z. Molecular characterization of PXR and two sulfotransferases and hepatic transcripts of PXR, two sulfotransferases and CYP3A responsive to bisphenol A in rare minnow Gobiocypris rarus. Mol Biol Rep 2014; 41:7153-65. [PMID: 25038724 DOI: 10.1007/s11033-014-3598-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/07/2014] [Indexed: 12/23/2022]
Abstract
Bisphenol A (BPA), a wide distributed endocrine-disrupting chemical, has attracted many attentions. To explore the effect of BPA on hepatic metabolic pathways in Gobiocypris rarus, full-length cDNAs of pregnane X receptor (PXR) and two sulfotransferases (SULT1 ST4 and SULT1 ST6) were firstly isolated and characterized. We detected tissues distribution of PXR, CYP3A, SULT1 ST4 and SULT1 ST6 in adult G. rarus. Then we investigated hepatic transcript profiles of these four genes in adult G. rarus exposed to BPA at concentrations of 5, 15, and 50 µg/L for 14 and 35 days. It demonstrates that these four genes are all highly expressed in liver of both male and female adult G. rarus. In response to BPA, sexual dimorphism of expression patterns for PXR, CYP3A, and SULT1 ST6 shows in G. rarus, which includes increase of mRNA levels in females and decrease of mRNA levels in males in both exposure durations of 14 and 35 days. SULT1 ST6 mRNA demonstrates high responsiveness to BPA in both genders and we recommended SULT1 ST6 as a candidate biomarker for BPA exposure.
Collapse
Affiliation(s)
- Jiancao Gao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, 22 Xinong Road, Yangling, 712100, Shaanxi, China
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Sinz MW, Kim S. Stem cells, immortalized cells and primary cells in ADMET assays. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 3:79-85. [PMID: 24980105 DOI: 10.1016/j.ddtec.2006.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cell-based assays are beginning to replace traditional absorption, distribution, metabolism, elimination and toxicology (ADMET) models employing subcellular fractions in high throughput drug discovery screening and drug development where drugs are characterized and predictions are formulated to forecast in vivo biological outcomes. Significant and continuing advances in stem cell research, new immortalized cell lines and our enhanced ability to predict outcomes from primary cells have increased the ability to employ cell-based assays to study ADMET properties of new drugs.:
Collapse
Affiliation(s)
- Michael W Sinz
- Department of Metabolism and Pharmacokinetics, Pharmaceutical Candidate Optimization, Bristol Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492, USA.
| | - Sean Kim
- Department of Metabolism and Pharmacokinetics, Pharmaceutical Candidate Optimization, Bristol Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492, USA
| |
Collapse
|
128
|
Baghdasaryan A, Chiba P, Trauner M. Clinical application of transcriptional activators of bile salt transporters. Mol Aspects Med 2014; 37:57-76. [PMID: 24333169 PMCID: PMC4045202 DOI: 10.1016/j.mam.2013.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/21/2013] [Accepted: 12/01/2013] [Indexed: 02/07/2023]
Abstract
Hepatobiliary bile salt (BS) transporters are critical determinants of BS homeostasis controlling intracellular concentrations of BSs and their enterohepatic circulation. Genetic or acquired dysfunction of specific transport systems causes intrahepatic and systemic retention of potentially cytotoxic BSs, which, in high concentrations, may disturb integrity of cell membranes and subcellular organelles resulting in cell death, inflammation and fibrosis. Transcriptional regulation of canalicular BS efflux through bile salt export pump (BSEP), basolateral elimination through organic solute transporters alpha and beta (OSTα/OSTβ) as well as inhibition of hepatocellular BS uptake through basolateral Na(+)-taurocholate cotransporting polypeptide (NTCP) represent critical steps in protection from hepatocellular BS overload and can be targeted therapeutically. In this article, we review the potential clinical implications of the major BS transporters BSEP, OSTα/OSTβ and NTCP in the pathogenesis of hereditary and acquired cholestatic syndromes, provide an overview on transcriptional control of these transporters by the key regulatory nuclear receptors and discuss the potential therapeutic role of novel transcriptional activators of BS transporters in cholestasis.
Collapse
Affiliation(s)
- Anna Baghdasaryan
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria; Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Peter Chiba
- Institute of Medical Chemistry, Medical University of Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
129
|
Niu Y, Wang Z, Huang H, Zhong S, Cai W, Xie Y, Shi G. Activated pregnane X receptor inhibits cervical cancer cell proliferation and tumorigenicity by inducing G2/M cell-cycle arrest. Cancer Lett 2014; 347:88-97. [PMID: 24486740 DOI: 10.1016/j.canlet.2014.01.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/02/2014] [Accepted: 01/24/2014] [Indexed: 02/05/2023]
Abstract
Pregnane X receptor (PXR) regulates cell proliferation and carcinogenesis in female reproductive tissue. We showed that PXR was expressed in cervical cells and tissue samples. PXR were lower or greatly diminished in cancer tissues compared to normal control. Functionally, activation of human PXR by rifampicin or ectopic expression of constitutively-activated human VP-PXR inhibited cervical cell proliferation. Constitutively-activated VP-PXR attenuated CaSki and HeLa xenograft tumor growth in nude mice compared with control. The cellular proliferation inhibition of PXR by causing G2/M cell-cycle arrest is involved up-regulation of Cullin1-3, MAD2L1, and down-regulation of ANAPC2 and CDKN1A. Our data suggests that PXR signaling inhibits tumor cell proliferation in vitro and cervical carcinoma growth in vivo.
Collapse
Affiliation(s)
- Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Guangdong, China
| | - Ziliang Wang
- Cancer Research Laboratory, Fudan University Shanghai Cancer Center, Department of Oncology, Fudan University, Shanghai, China
| | - Haihua Huang
- Department of Pathology, Second Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Shuping Zhong
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Wenfeng Cai
- Department of Pharmacology, Shantou University Medical College, Guangdong, China
| | - Yangmin Xie
- Department of Experimental Animal Center, Medical College of Shantou University, Guangdong, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Guangdong, China.
| |
Collapse
|
130
|
Sui Y, Park S, Helsley RN, Sunkara M, Gonzalez FJ, Morris AJ, Zhou C. Bisphenol A increases atherosclerosis in pregnane X receptor-humanized ApoE deficient mice. J Am Heart Assoc 2014; 3:e000492. [PMID: 24755147 PMCID: PMC4187496 DOI: 10.1161/jaha.113.000492] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background Bisphenol A (BPA) is a base chemical used extensively in many consumer products. BPA has recently been associated with increased risk of cardiovascular disease (CVD) in multiple large‐scale human population studies, but the underlying mechanisms remain elusive. We previously reported that BPA activates the pregnane X receptor (PXR), which acts as a xenobiotic sensor to regulate xenobiotic metabolism and has pro‐atherogenic effects in animal models upon activation. Interestingly, BPA is a potent agonist of human PXR but does not activate mouse or rat PXR signaling, which confounds the use of rodent models to evaluate mechanisms of BPA‐mediated CVD risk. This study aimed to investigate the atherogenic mechanism of BPA using a PXR‐humanized mouse model. Methods and Results A PXR‐humanized ApoE deficient (huPXR•ApoE−/−) mouse line was generated that respond to human PXR ligands and feeding studies were performed to determine the effects of BPA exposure on atherosclerosis development. Exposure to BPA significantly increased atherosclerotic lesion area in the aortic root and brachiocephalic artery of huPXR•ApoE−/− mice by 104% (P<0.001) and 120% (P<0.05), respectively. By contrast, BPA did not affect atherosclerosis development in the control littermates without human PXR. BPA exposure did not affect plasma lipid levels but increased CD36 expression and lipid accumulation in macrophages of huPXR•ApoE−/− mice. Conclusion These findings identify a molecular mechanism that could link BPA exposure to increased risk of CVD in exposed individuals. PXR is therefore a relevant target for future risk assessment of BPA and related environmental chemicals in humans.
Collapse
Affiliation(s)
- Yipeng Sui
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY (Y.S., S.H.P., R.N.H., C.Z.)
| | - Se‐Hyung Park
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY (Y.S., S.H.P., R.N.H., C.Z.)
| | - Robert N. Helsley
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY (Y.S., S.H.P., R.N.H., C.Z.)
| | - Manjula Sunkara
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY (M.S., A.J.M.)
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Andrew J. Morris
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY (A.J.M., C.Z.)
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY (M.S., A.J.M.)
| | - Changcheng Zhou
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY (Y.S., S.H.P., R.N.H., C.Z.)
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY (A.J.M., C.Z.)
| |
Collapse
|
131
|
Jiang M, He J, Kucera H, Gaikwad NW, Zhang B, Xu M, O'Doherty RM, Selcer KW, Xie W. Hepatic overexpression of steroid sulfatase ameliorates mouse models of obesity and type 2 diabetes through sex-specific mechanisms. J Biol Chem 2014; 289:8086-97. [PMID: 24497646 DOI: 10.1074/jbc.m113.535914] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The steroid sulfatase (STS)-mediated desulfation is a critical metabolic mechanism that regulates the chemical and functional homeostasis of endogenous and exogenous molecules. In this report, we first showed that the liver expression of Sts was induced in both the high fat diet (HFD) and ob/ob models of obesity and type 2 diabetes and during the fed to fasting transition. In defining the functional relevance of STS induction in metabolic disease, we showed that overexpression of STS in the liver of transgenic mice alleviated HFD and ob/ob models of obesity and type 2 diabetes, including reduced body weight, improved insulin sensitivity, and decreased hepatic steatosis and inflammation. Interestingly, STS exerted its metabolic benefit through sex-specific mechanisms. In female mice, STS may have increased hepatic estrogen activity by converting biologically inactive estrogen sulfates to active estrogens and consequently improved the metabolic functions, whereas ovariectomy abolished this protective effect. In contrast, the metabolic benefit of STS in males may have been accounted for by the male-specific decrease of inflammation in white adipose tissue and skeletal muscle as well as a pattern of skeletal muscle gene expression that favors energy expenditure. The metabolic benefit in male STS transgenic mice was retained after castration. Treatment with the STS substrate estrone sulfate also improved metabolic functions in both the HFD and ob/ob models. Our results have uncovered a novel function of STS in energy metabolism and type 2 diabetes. Liver-specific STS induction or estrogen/estrogen sulfate delivery may represent a novel approach to manage metabolic syndrome.
Collapse
Affiliation(s)
- Mengxi Jiang
- From the Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Spruiell K, Richardson RM, Cullen JM, Awumey EM, Gonzalez FJ, Gyamfi MA. Role of pregnane X receptor in obesity and glucose homeostasis in male mice. J Biol Chem 2013; 289:3244-61. [PMID: 24362030 DOI: 10.1074/jbc.m113.494575] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Clinical obesity is a complex metabolic disorder affecting one in three adults. Recent reports suggest that pregnane X receptor (PXR), a xenobiotic nuclear receptor important for defense against toxic agents and for eliminating drugs and other xenobiotics, may be involved in obesity. Noting differences in ligand specificities between human and mouse PXRs, the role of PXR in high fat diet (HFD)-induced obesity was examined using male PXR-humanized (hPXR) transgenic and PXR-knock-out (PXR-KO) mice in comparison to wild-type (WT) mice. After 16 weeks on either a control diet or HFD, WT mice showed greater weight gain, whereas PXR-KO mice gained less weight due to their resistance to HFD-induced decreases in adipose tissue peroxisome proliferator-activated receptor α and induction of hepatic carnitine palmitoyltransferase 1, suggesting increased energy metabolism. Interestingly, control-fed PXR-KO mice exhibited hepatomegaly, hyperinsulinemia, and hyperleptinemia but hypoadiponectinemia and lower adiponectin receptor R2 mRNA levels relative to WT mice. Evaluation of these biologic indicators in hPXR mice fed a control diet or HFD revealed further differences between the mouse and human receptors. Importantly, although HFD-fed hPXR mice were resistant to HFD-induced obesity, both PXR-KO and hPXR mice exhibited impaired induction of glucokinase involved in glucose utilization and displayed elevated fasting glucose levels and severely impaired glucose tolerance. Moreover, the basal hepatic levels of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase 1 were increased in hPXR mice compared with WT mice. Altogether, although the mouse PXR promotes HFD-induced obesity, the hPXR mouse carries a genetic predisposition for type 2 diabetes and thus provides a model for exploring the role of human PXR in the metabolic syndrome.
Collapse
Affiliation(s)
- Krisstonia Spruiell
- From the Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | | | | | | | | | | |
Collapse
|
133
|
Elcombe CR, Peffer RC, Wolf DC, Bailey J, Bars R, Bell D, Cattley RC, Ferguson SS, Geter D, Goetz A, Goodman JI, Hester S, Jacobs A, Omiecinski CJ, Schoeny R, Xie W, Lake BG. Mode of action and human relevance analysis for nuclear receptor-mediated liver toxicity: A case study with phenobarbital as a model constitutive androstane receptor (CAR) activator. Crit Rev Toxicol 2013; 44:64-82. [PMID: 24180433 DOI: 10.3109/10408444.2013.835786] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The constitutive androstane receptor (CAR) and pregnane X receptor (PXR) are important nuclear receptors involved in the regulation of cellular responses from exposure to many xenobiotics and various physiological processes. Phenobarbital (PB) is a non-genotoxic indirect CAR activator, which induces cytochrome P450 (CYP) and other xenobiotic metabolizing enzymes and is known to produce liver foci/tumors in mice and rats. From literature data, a mode of action (MOA) for PB-induced rodent liver tumor formation was developed. A MOA for PXR activators was not established owing to a lack of suitable data. The key events in the PB-induced liver tumor MOA comprise activation of CAR followed by altered gene expression specific to CAR activation, increased cell proliferation, formation of altered hepatic foci and ultimately the development of liver tumors. Associative events in the MOA include altered epigenetic changes, induction of hepatic CYP2B enzymes, liver hypertrophy and decreased apoptosis; with inhibition of gap junctional intercellular communication being an associative event or modulating factor. The MOA was evaluated using the modified Bradford Hill criteria for causality and other possible MOAs were excluded. While PB produces liver tumors in rodents, important species differences were identified including a lack of cell proliferation in cultured human hepatocytes. The MOA for PB-induced rodent liver tumor formation was considered to be qualitatively not plausible for humans. This conclusion is supported by data from a number of epidemiological studies conducted in human populations chronically exposed to PB in which there is no clear evidence for increased liver tumor risk.
Collapse
|
134
|
Kittayaruksakul S, Zhao W, Xu M, Ren S, Lu J, Wang J, Downes M, Evans RM, Venkataramanan R, Chatsudthipong V, Xie W. Identification of three novel natural product compounds that activate PXR and CAR and inhibit inflammation. Pharm Res 2013; 30:2199-208. [PMID: 23896737 PMCID: PMC3771640 DOI: 10.1007/s11095-013-1101-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE To investigate the effects of three natural product compounds, carapin, santonin and isokobusone, on the activity of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) in induction of drug-metabolizing enzymes and inhibition of inflammation. METHODS The monkey kidney-derived fibroblast (CV-1) cells and human embryonic kidney HEK293 cells were used for transient transfection and luciferase reporter gene assays. Human primary hepatocytes and primary hepatocytes from wild type, PXR-/-, and hPXR transgenic mice were used to study the induction of drug-metabolizing enzymes and the implication of these compounds in inflammation. RESULTS Carapin, santonin and isokobusone activated both PXR and CAR in transient transfection and luciferase reporter gene assays. Mutagenesis studies showed that two amino acid residues, Phe305 of the rodent PXR and Leu308 of the human PXR, are critical for the recognition of these compounds by PXR. Importantly, the activation of PXR and CAR by these compounds induced the expression of drug-metabolizing enzymes in primary human and mouse hepatocytes. Furthermore, activation of PXR by these compounds inhibited the expression of inflammatory mediators in response to lipopolysaccharide (LPS). The effects of these natural compounds on drug metabolism and inflammation were abolished in PXR-/- hepatocytes. CONCLUSIONS Our results show that carapin, santonin and isokobusone activate PXR and CAR and induce drug-metabolizing enzymes. In addition, these compounds inhibited the expression of inflammatory mediators in response to LPS through the activation of PXR.
Collapse
Affiliation(s)
- Suticha Kittayaruksakul
- Department of Physiology, Mahidol University, Bangkok 10400, Thailand
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Meishu Xu
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Songrong Ren
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jing Lu
- Department of Lab Animal Science, Capital Medical University, Beijing 100069, China
| | - Ju Wang
- Department of Lab Animal Science, Capital Medical University, Beijing 100069, China
| | - Michael Downes
- Department of Lab Animal Science, Capital Medical University, Beijing 100069, China
| | - Ronald M. Evans
- Department of Lab Animal Science, Capital Medical University, Beijing 100069, China
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | - Wen Xie
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Gene Expression Laboratory, The Salk Institute for Biological Sciences, La Jolla, CA 92037, USA
| |
Collapse
|
135
|
Scheer N, Snaith M, Wolf CR, Seibler J. Generation and utility of genetically humanized mouse models. Drug Discov Today 2013; 18:1200-11. [PMID: 23872278 DOI: 10.1016/j.drudis.2013.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/20/2013] [Accepted: 07/11/2013] [Indexed: 01/15/2023]
Abstract
Identifying in vivo models that are naturally predictive for particular areas of study in humans can be challenging due to the divergence that has occurred during speciation. One solution to this challenge that is gaining increasing traction is the use of genetic engineering to introduce human genes into mice to generate superior models for predicting human responses. This review describes the state-of-the-art for generating such models, provides an overview of the types of genetically humanized mouse models described to date and their applications in basic research, drug discovery and development and to understand clinical drug toxicity. We discuss limitations and explore promising future directions for the use of genetically humanized mice to further improve translational research.
Collapse
Affiliation(s)
- Nico Scheer
- TaconicArtemis, Neurather Ring 1, Koeln 51063, Germany.
| | | | | | | |
Collapse
|
136
|
Activation of human stearoyl-coenzyme A desaturase 1 contributes to the lipogenic effect of PXR in HepG2 cells. PLoS One 2013; 8:e67959. [PMID: 23874477 PMCID: PMC3706516 DOI: 10.1371/journal.pone.0067959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/23/2013] [Indexed: 11/19/2022] Open
Abstract
The pregnane X receptor (PXR) was previously known as a xenobiotic receptor. Several recent studies suggested that PXR also played an important role in lipid homeostasis but the underlying mechanism remains to be clearly defined. In this study, we found that rifampicin, an agonist of human PXR, induced lipid accumulation in HepG2 cells. Lipid analysis showed the total cholesterol level increased. However, the free cholesterol and triglyceride levels were not changed. Treatment of HepG2 cells with rifampicin induced the expression of the free fatty acid transporter CD36 and ABCG1, as well as several lipogenic enzymes, including stearoyl-CoA desaturase-1 (SCD1), long chain free fatty acid elongase (FAE), and lecithin-cholesterol acyltransferase (LCAT), while the expression of acyl:cholesterol acetyltransferase(ACAT1) was not affected. Moreover, in PXR over-expressing HepG2 cells (HepG2-PXR), the SCD1 expression was significantly higher than in HepG2-Vector cells, even in the absence of rifampicin. Down-regulation of PXR by shRNA abolished the rifampicin-induced SCD1 gene expression in HepG2 cells. Promoter analysis showed that the human SCD1 gene promoter is activated by PXR and a novel DR-7 type PXR response element (PXRE) response element was located at -338 bp of the SCD1 gene promoter. Taken together, these results indicated that PXR activation promoted lipid synthesis in HepG2 cells and SCD1 is a novel PXR target gene.
Collapse
|
137
|
Sekiguchi M, Kobashigawa Y, Moriguchi H, Kawasaki M, Yuda M, Teramura T, Inagaki F. High-Throughput Evaluation Method for Drug Association with Pregnane X Receptor (PXR) Using Differential Scanning Fluorometry. ACTA ACUST UNITED AC 2013; 18:1084-91. [DOI: 10.1177/1087057113491826] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The pregnane xenobiotic receptor (PXR) is a key transcriptional regulator of cytochrome P450 (CYP) 3A, a crucial enzyme in the metabolism and detoxification of xenobiotics and endobiotics. PXR is activated by a wide variety of chemicals and serves as a master regulator of detoxification in mammals. Here, we report a fast evaluation method for PXR-drug interactions using differential scanning fluorometry (DSF). DSF analysis revealed that PXR associates with a fluorescence dye in the native state as well as in the unfolded state, which prevented precise evaluation of any shift in the transition midpoint (Δ Tm) due to association with a drug. Hence, we defined a new parameter, (dF/dT)50, where F is fluorescence intensity and T is temperature, to describe the ligand concentration. (dF/dT)50 exhibited better correlation with EC50 ( r2 = 0.84) than with Δ Tm ( r2 = 0.71). The correlation of Δ Tm measured using differential scanning calorimetry (DSC) with EC50 ( r2 = 0.86) was similar to the above (dF/dT)50 correlation. Therefore, the use of (dF/dT)50 enables DSF to be used for the rapid evaluation of PXR-drug interactions and could provide prescreening to narrow down the collection of candidate ligands that most likely result in transcriptional activation of CYP3A4.
Collapse
Affiliation(s)
- Mitsuhiro Sekiguchi
- Analysis and Pharmacokinetics Research Labs, Department of Drug Discovery, Astellas Pharma Inc., Tsukuba, Japan
| | - Yoshihiro Kobashigawa
- Department of Structural Biology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Analytical and Biophysical Chemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Kumamoto, Japan
| | - Hiroyuki Moriguchi
- Analysis and Pharmacokinetics Research Labs, Department of Drug Discovery, Astellas Pharma Inc., Tsukuba, Japan
| | - Masashi Kawasaki
- Analysis and Pharmacokinetics Research Labs, Department of Drug Discovery, Astellas Pharma Inc., Tsukuba, Japan
| | - Masamichi Yuda
- Analysis and Pharmacokinetics Research Labs, Department of Drug Discovery, Astellas Pharma Inc., Tsukuba, Japan
| | - Toshio Teramura
- Analysis and Pharmacokinetics Research Labs, Department of Drug Discovery, Astellas Pharma Inc., Tsukuba, Japan
| | - Fuyuhiko Inagaki
- Department of Structural Biology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
138
|
He J, Gao J, Xu M, Ren S, Stefanovic-Racic M, O'Doherty RM, Xie W. PXR ablation alleviates diet-induced and genetic obesity and insulin resistance in mice. Diabetes 2013; 62:1876-87. [PMID: 23349477 PMCID: PMC3661619 DOI: 10.2337/db12-1039] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pregnane X receptor (PXR), along with its sister receptor constitutive androstane receptor (CAR), was initially characterized as a xenobiotic receptor that regulates drug metabolism. In this study, we have uncovered an unexpected endobiotic role of PXR in obesity and type 2 diabetes. PXR ablation inhibited high-fat diet (HFD)-induced obesity, hepatic steatosis, and insulin resistance, which were accounted for by increased oxygen consumption, increased mitochondrial β-oxidation, inhibition of hepatic lipogenesis and inflammation, and sensitization of insulin signaling. In an independent model, introducing the PXR(-/-) allele into the ob/ob background also improved body composition and relieved the diabetic phenotype. The ob/ob mice deficient of PXR showed increased oxygen consumption and energy expenditure, as well as inhibition of gluconeogenesis and increased rate of glucose disposal during euglycemic clamp. Mechanistically, the metabolic benefits of PXR ablation were associated with the inhibition of c-Jun NH2-terminal kinase activation and downregulation of lipin-1, a novel PXR target gene. The metabolic benefit of PXR ablation was opposite to the reported prodiabetic effect of CAR ablation. Our results may help to establish PXR as a novel therapeutic target, and PXR antagonists may be used for the prevention and treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Jinhan He
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Gao
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Martin O'Doherty
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Corresponding author: Wen Xie,
| |
Collapse
|
139
|
Gu L, Chen J, Synold TW, Forman BM, Kane SE. Bioimaging real-time PXR-dependent mdr1a gene regulation in mdr1a.fLUC reporter mice. J Pharmacol Exp Ther 2013; 345:438-45. [PMID: 23532932 PMCID: PMC3657110 DOI: 10.1124/jpet.113.203562] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 03/25/2013] [Indexed: 11/22/2022] Open
Abstract
The MDR1 gene encodes P-glycoprotein, a transmembrane drug efflux transporter that confers multidrug resistance in cancer cells and affects drug pharmacokinetics by virtue of its expression in the liver, kidney, and colon. Nuclear receptors human steroid and xenobiotic receptor (SXR) and constitutive androstane receptor (CAR) are possible master regulators of xenobiotic-inducible MDR1 expression in drug processing organs, but the mechanism of MDR1 regulation has yet to be directly demonstrated in vivo. Moreover, it has previously been impossible to determine the sustained or cumulative effect of repeated doses of xenobiotics on in vivo MDR1 expression. We previously reported a mouse model containing firefly luciferase (fLUC) knocked into the mdr1a genomic locus, allowing noninvasive bioimaging of intestinal mdr1a gene expression in live animals. In the current study, we crossed mdr1a.fLUC mice into the pxr knockout (pxr(-/-)) genetic background and injected mice with pregnenolone-16α-carbonitrile (PCN), a strong mouse pregnane X receptor (PXR) ligand, and two therapeutically relevant taxanes, paclitaxel and docetaxel. All three agents induced mdr1a.fLUC expression (bioluminescence), but only PCN and docetaxel appeared to act primarily via PXR. Luminescence returned to baseline by 24-48 hours after drug injection and was reinducible over two additional rounds of drug dosing in pxr(+/+) mice. TCPOBOP, a CAR ligand, modestly induced mdr1a.fLUC in pxr(+/+) and pxr(-/-) strains, consistent with CAR's minor role in mdr1a regulation. Collectively, these results demonstrate that the mdr1a.fLUC bioimaging model can capture changes in mdr1 gene expression under conditions of repeated xenobiotic treatment in vivo and that it can be used to probe the mechanism of gene regulation in response to different xenobiotic agents.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/drug effects
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Constitutive Androstane Receptor
- Docetaxel
- Fatty Acids, Monounsaturated/pharmacology
- Gene Expression/drug effects
- Genes, Reporter/drug effects
- Genes, Reporter/genetics
- Humans
- Image Processing, Computer-Assisted
- Intestinal Mucosa/metabolism
- Ligands
- Luciferases/genetics
- Luminescent Proteins/biosynthesis
- Mice
- Mice, Knockout
- Paclitaxel/pharmacology
- Pregnane X Receptor
- Pyridines/pharmacology
- Quaternary Ammonium Compounds/pharmacology
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Steroid/drug effects
- Receptors, Steroid/genetics
- Taxoids/pharmacology
- Xenobiotics/pharmacology
Collapse
Affiliation(s)
- Long Gu
- Department of Cancer Biology, Beckman Research Institute at City of Hope, Duarte, California, USA
| | | | | | | | | |
Collapse
|
140
|
Kodama S, Negishi M. PXR cross-talks with internal and external signals in physiological and pathophysiological responses. Drug Metab Rev 2013; 45:300-10. [DOI: 10.3109/03602532.2013.795585] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
141
|
Imai Y, Youn MY, Inoue K, Takada I, Kouzmenko A, Kato S. Nuclear receptors in bone physiology and diseases. Physiol Rev 2013; 93:481-523. [PMID: 23589826 PMCID: PMC3768103 DOI: 10.1152/physrev.00008.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During the last decade, our view on the skeleton as a mere solid physical support structure has been transformed, as bone emerged as a dynamic, constantly remodeling tissue with systemic regulatory functions including those of an endocrine organ. Reflecting this remarkable functional complexity, distinct classes of humoral and intracellular regulatory factors have been shown to control vital processes in the bone. Among these regulators, nuclear receptors (NRs) play fundamental roles in bone development, growth, and maintenance. NRs are DNA-binding transcription factors that act as intracellular transducers of the respective ligand signaling pathways through modulation of expression of specific sets of cognate target genes. Aberrant NR signaling caused by receptor or ligand deficiency may profoundly affect bone health and compromise skeletal functions. Ligand dependency of NR action underlies a major strategy of therapeutic intervention to correct aberrant NR signaling, and significant efforts have been made to design novel synthetic NR ligands with enhanced beneficial properties and reduced potential negative side effects. As an example, estrogen deficiency causes bone loss and leads to development of osteoporosis, the most prevalent skeletal disorder in postmenopausal women. Since administration of natural estrogens for the treatment of osteoporosis often associates with undesirable side effects, several synthetic estrogen receptor ligands have been developed with higher therapeutic efficacy and specificity. This review presents current progress in our understanding of the roles of various nuclear receptor-mediated signaling pathways in bone physiology and disease, and in development of advanced NR ligands for treatment of common skeletal disorders.
Collapse
Affiliation(s)
- Yuuki Imai
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
142
|
Papageorgiou I, Grepper S, Unadkat JD. Induction of hepatic CYP3A enzymes by pregnancy-related hormones: studies in human hepatocytes and hepatic cell lines. Drug Metab Dispos 2013; 41:281-90. [PMID: 23223499 DOI: 10.1124/dmd.112.049015] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
CYP3A activity is induced by approximately 2-fold during the third trimester of human pregnancy. Placental growth hormone (PGH), estrogens (primarily 17β-estradiol), cortisol, and progesterone have the potential to modulate CYP3A activity. Therefore, we determined whether the elevated plasma concentrations of these hormones during pregnancy induce hepatic CYP3A expression. We incubated sandwich-cultured human hepatocytes (SCHH) from premenopausal female donors (n = 2) with the physiologic (unbound, 1× total) and the 10× total third trimester hormone plasma concentrations (individually and in combination) and determined their effect on CYP3A activity and the transcripts of CYP3A4, CYP3A5, and the respective hormone receptors (growth hormone receptor, glucocorticoid receptor, and estrogen receptor alpha). Of all the hormones, cortisol was the most potent inducer of CYP3A activity and CYP3A4, CYP3A5 mRNA expression. The combination of PGH/growth hormone and cortisol induced CYP3A activity and expression significantly more than did cortisol alone. When incubated with the unbound or total plasma concentration of all the hormones, CYP3A activity in SCHH was induced to an extent comparable to that observed in vivo during the third trimester. These hormones had only a modest effect on the mRNA expression of the hormone receptors. The pattern of induction observed in SCHH was reproduced in HepaRG cells but not in HuH7/HepG2 cells. SCHH or HepaRG cells could be used to determine the mechanistic basis of CYP3A induction during pregnancy and to predict the magnitude of induction likely to be observed during the first and second trimesters, when phenotyping studies to measure in vivo CYP3A activity are logistically difficult to perform.
Collapse
Affiliation(s)
- Ioannis Papageorgiou
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195-7610, USA
| | | | | |
Collapse
|
143
|
Abstract
The cytosolic sulfotransferases (SULTs) are a multigene family of enzymes that catalyze the transfer of a sulfonate group from the physiologic sulfate donor, 3'-phosphoadenosine-5'-phosphosulfate, to a nucleophilic substrate to generate a polar product that is more amenable to elimination from the body. As catalysts of both xenobiotic and endogenous metabolism, the SULTs are major points of contact between the external and physiological environments, and modulation of SULT-catalyzed metabolism can not only affect xenobiotic disposition, but it can also alter endogenous metabolic processes. Therefore, it is not surprising that SULT expression is regulated by numerous members of the nuclear receptor (NR) superfamily that function as sensors of xenobiotics as well as endogenous molecules, such as fatty acids, bile acids, and oxysterols. These NRs include the peroxisome proliferator-activated receptors, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, liver X receptors, farnesoid X receptor, retinoid-related orphan receptors, and estrogen-related receptors. This review summarizes current information about NR regulation of SULT expression. Because species differences in SULT subfamily composition and tissue-, sex-, development-, and inducer-dependent regulation are prominent, these differences will be emphasized throughout the review. In addition, because of the central role of the SULTs in cellular physiology, the effect of NR-mediated SULT regulation on physiological and pathophysiological processes will be discussed. Gaps in current knowledge that require further investigation are also highlighted.
Collapse
Affiliation(s)
- Melissa Runge-Morris
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan 48201, USA.
| | | | | |
Collapse
|
144
|
Chai X, Zeng S, Xie W. Nuclear receptors PXR and CAR: implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin Drug Metab Toxicol 2013; 9:253-66. [PMID: 23327618 DOI: 10.1517/17425255.2013.754010] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION 'Orphan' nuclear receptors belong to the nuclear receptor (NR) superfamily of transcriptional factors. Binding of ligands to these receptors results in the recruitment of the co-activators, thereby regulating the expression of cognate target genes. AREAS COVERED This review discusses the transcriptional regulation of P450 genes by two major xenobiotic nuclear receptors, pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Additional PXR and CAR target genes include those encoded for UDP-glucuronosyltransferases, glutathione S-transferases, sulfotransferases and drug transporters. The authors discuss the involvement of PXR and CAR in endobiotic metabolism. They also review the polymorphisms of PXR and CAR. EXPERT OPINION PXR and CAR are both xenobiotic and endobiotic receptors. A remarkably diverse set of chemicals can activate PXR and CAR. There is significant cross-talk among xenobiotic receptors. Future studies are needed to focus on the polymorphisms of the nuclear receptors and the complex regulatory networks among nuclear receptors. Considerations should be given while designing PXR- or CAR-targeting pharmaceutics to avoid adverse drug effects. In the meantime, due to the diverse functions of PXR and CAR, agonists or antagonists for these receptors may have therapeutic potentials in managing certain diseases and enhancing therapeutic indexes.
Collapse
Affiliation(s)
- Xiaojuan Chai
- Zhejiang University, College of Pharmaceutical Sciences, Department of Pharmaceutical Analysis and Drug Metabolism, Hangzhou 310058, China
| | | | | |
Collapse
|
145
|
Zhou T, Cong S, Sun S, Sun H, Zou R, Wang S, Wang C, Jiao J, Goto K, Nawata H, Yanase T, Zhao Y. Identification of endocrine disrupting chemicals activating SXR-mediated transactivation of CYP3A and CYP7A1. Mol Cell Endocrinol 2013; 365:36-43. [PMID: 22975079 DOI: 10.1016/j.mce.2012.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 09/03/2012] [Accepted: 09/03/2012] [Indexed: 10/27/2022]
Abstract
Endocrine disrupting chemicals (EDCs) have emerged as a major public health issue because of their potentially disruptive effects on physiological hormonal actions. SXR (steroid xenobiotic receptor), also known as NR1I2, regulates CYP3A expression in response to exogenous chemicals, such as EDCs, after binding to SXRE (SXR response element). In our study, luciferase assay showed that 14 out of 55 EDCs could enhance SXR-mediated rat or human CYP3A gene transcription nearly evenly, and could also activate rat CYP7A1 gene transcription by cross-interaction of SXR and LXRE (LXRα response element). SXR diffused in the nucleus without ligand, whereas intranuclear foci of liganded SXR were produced. Furthermore, endogenous mRNA expression of CYP3A4 gene was enhanced by the 14 positive EDCs. Our results suggested a probable mechanism of EDCs disrupting the steroid or xenobiotic metabolism homeostasis via SXR.
Collapse
Affiliation(s)
- Tingting Zhou
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110001, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Yue X, Utsunomiya H, Akahira JI, Suzuki F, Ito K, Nagase S, Sasano H, Yaegashi N. Expression of steroid and xenobiotic receptor in uterine carcinosarcoma, leiomyosarcoma and endometrial stromal sarcoma. Oncol Lett 2012; 5:835-839. [PMID: 23443531 PMCID: PMC3576214 DOI: 10.3892/ol.2012.1094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/24/2012] [Indexed: 11/16/2022] Open
Abstract
We analyzed the expression of the steroid and xenobiotic receptor (SXR) in human uterine sarcomas and evaluated its clinical significance. Forty-seven cases with archival specimens were examined for SXR expression using immunohistochemistry. All cases were scored using a semi-quantitative histological scoring (HSCORE) method. Specimens with a HSCORE >40 were regarded as SXR-positive. Various clinicopathological variables, including the expression status of estrogen receptor (ER)-α, progesterone receptor (PR) and Ki67 (MIB-1) were examined. The mean SXR HSCOREs of carcinosarcoma (CS) and leiomyosarcoma (LMS) were 9.13 and 23.6, respectively, and SXR-positive rates were 3 out of 24 (12.5%) and 4 out of 17 (23.5%), respectively. SXR was not detected in endometrial stromal sarcoma (ESS). In CS cases, significant differences were detected between the expression of SXR and age and disease stages. There was no significant correlation between SXR-positive status and either disease-free survival or overall survival. Our results support an association between SXR and malignant behavior. Our results show that overexpression of SXR may represent a useful marker to identify patients with advanced-stage CS. In addition, our results showed that SXR may aid in the diagnosis of uterine sarcomas.
Collapse
Affiliation(s)
- Xiaoni Yue
- Departments of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University, Sendai 980-8574, Japan ; ; Department of Obstetrics and Gynecology, Fudan University, Shanghai 042465, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Ishizawa M, Ogura M, Kato S, Makishima M. Impairment of bilirubin clearance and intestinal interleukin-6 expression in bile duct-ligated vitamin D receptor null mice. PLoS One 2012; 7:e51664. [PMID: 23240054 PMCID: PMC3519857 DOI: 10.1371/journal.pone.0051664] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/06/2012] [Indexed: 01/06/2023] Open
Abstract
The vitamin D receptor (VDR) mediates the physiological and pharmacological actions of 1α,25-dihydroxyvitamin D3 in bone and calcium metabolism, cellular growth and differentiation, and immunity. VDR also responds to secondary bile acids and belongs to the NR1I subfamily of the nuclear receptor superfamily, which regulates expression of xenobiotic metabolism genes. When compared to knockout mouse investigations of the other NR1I nuclear receptors, pregnane X receptor and constitutive androstane receptor, an understanding of the role of VDR in xenobiotic metabolism remains limited. We examined the effect of VDR deletion in a mouse model of cholestasis. We performed bile duct ligation (BDL) on VDR-null mice and compared blood biochemistry, mRNA expression of genes involved in bile acid and bilirubin metabolism, cytokine production, and expression of inflammatory regulators with those of wild-type mice. VDR-null mice had elevated plasma conjugated bilirubin levels three days after BDL compared with wild-type mice. Urine bilirubin levels and renal mRNA and/or protein expression of multidrug resistance-associated proteins 2 and 4 were decreased in VDR-null mice, suggesting impaired excretion of conjugated bilirubin into urine. While VDR-null kidney showed mRNA expression of interleukin-6 (IL-6) after BDL and VDR-null macrophages had higher IL-6 protein levels after lipopolysaccharide stimulation, the induction of intestinal Il6 mRNA expression and plasma IL-6 protein levels after BDL was impaired in VDR-null mice. Immunoblotting analysis showed that expression of an immune regulator, IκBα, was elevated in the jejunum of VDR-null mice, a possible mechanism for the attenuated induction of Il6 expression in the intestine after BDL. Increased expression of IκBα may be a consequence of compensatory mechanisms for VDR deletion. These results reveal a role of VDR in bilirubin clearance during cholestasis. VDR is also suggested to contribute to tissue-selective immune regulation.
Collapse
Affiliation(s)
- Michiyasu Ishizawa
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | - Michitaka Ogura
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | - Shigeaki Kato
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
148
|
Abstract
The nuclear receptors pregnane X receptor, constitutive androstane receptor, and peroxisome proliferator-activated receptor alpha have important endogenous functions and are also involved in the induction of drug-metabolizing enzymes and transporters in response to exogenous xenobiotics. Though not belonging to the same protein family, the Per-Sim-ARNT domain receptor aryl hydrocarbon receptor functionally overlaps with the three nuclear receptors in many aspects and is therefore included in this review. Significant species differences in ligand affinity and biological responses as a result of activation of these receptors have been described. Several xenobiotic receptor humanized mice have been created to overcome these species differences and to provide in vivo models that are more predictive for human responses. This review provides an overview of the different xenobiotic receptor humanized mouse models described to date and will summarize how these models can be applied in basic research and improve drug discovery and development. Some of the key applications in the evaluation of drug induction, drug-drug interactions, nongenotoxic carcinogenicity, other toxicity, or efficacy studies are described. We also discuss relevant considerations in the interpretation of such data and potential future directions for the use of xenobiotic receptor humanized mice.
Collapse
Affiliation(s)
- Nico Scheer
- TaconicArtemis GmbH, Neurather Ring 1, Koeln, Germany.
| | | |
Collapse
|
149
|
Poulton EJ, Levy L, Lampe JW, Shen DD, Tracy J, Shuhart MC, Thummel KE, Eaton DL. Sulforaphane is not an effective antagonist of the human pregnane X-receptor in vivo. Toxicol Appl Pharmacol 2012; 266:122-31. [PMID: 23153560 DOI: 10.1016/j.taap.2012.10.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 09/28/2012] [Accepted: 10/15/2012] [Indexed: 12/20/2022]
Abstract
Sulforaphane (SFN), is an effective in vitro antagonist of ligand activation of the human pregnane and xenobiotic receptor (PXR). PXR mediated CYP3A4 up-regulation is implicated in adverse drug-drug interactions making identification of small molecule antagonists a desirable therapeutic goal. SFN is not an antagonist to mouse or rat PXR in vitro; thus, normal rodent species are not suitable as in vivo models for human response. To evaluate whether SFN can effectively antagonize ligand activation of human PXR in vivo, a three-armed, randomized, crossover trial was conducted with 24 healthy adults. The potent PXR ligand - rifampicin (300mg/d) was given alone for 7days in arm 1, or in daily combination with 450μmol SFN (Broccoli Sprout extract) in arm 2; SFN was given alone in arm 3. Midazolam as an in vivo phenotype marker of CYP3A was administered before and after each treatment arm. Rifampicin alone decreased midazolam AUC by 70%, indicative of the expected increase in CYP3A4 activity. Co-treatment with SFN did not reduce CYP3A4 induction. Treatment with SFN alone also did not affect CYP3A4 activity in the cohort as a whole, although in the subset with the highest basal CYP3A4 activity there was a statistically significant increase in midazolam AUC (i.e., decrease in CYP3A4 activity). A parallel study in humanized PXR mice yielded similar results. The parallel effects of SFN between humanized PXR mice and human subjects demonstrate the predictive value of humanized mouse models in situations where species differences in ligand-receptor interactions preclude the use of a native mouse model for studying human ligand-receptor pharmacology.
Collapse
Affiliation(s)
- Emma Jane Poulton
- Center for Ecogenetics and Environmental Health, University of Washington, WA 98105-6099, USA
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Kazuki Y, Kobayashi K, Aueviriyavit S, Oshima T, Kuroiwa Y, Tsukazaki Y, Senda N, Kawakami H, Ohtsuki S, Abe S, Takiguchi M, Hoshiya H, Kajitani N, Takehara S, Kubo K, Terasaki T, Chiba K, Tomizuka K, Oshimura M. Trans-chromosomic mice containing a human CYP3A cluster for prediction of xenobiotic metabolism in humans. Hum Mol Genet 2012; 22:578-92. [PMID: 23125282 DOI: 10.1093/hmg/dds468] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human CYP3A is the most abundant P450 isozyme present in the human liver and small intestine, and metabolizes around 50% of medical drugs on the market. The human CYP3A subfamily comprises four members (CYP3A4, CYP3A5, CYP3A7, CYP3A43) encoded on human chromosome 7. However, transgenic mouse lines carrying the entire human CYP3A cluster have not been constructed because of limitations in conventional cloning techniques. Here, we show that the introduction of a human artificial chromosome (HAC) containing the entire genomic human CYP3A locus recapitulates tissue- and stage-specific expression of human CYP3A genes and xenobiotic metabolism in mice. About 700 kb of the entire CYP3A genomic segment was cloned into a HAC (CYP3A-HAC), and trans-chromosomic (Tc) mice carrying a single copy of germline-transmittable CYP3A-HAC were generated via a chromosome-engineering technique. The tissue- and stage-specific expression profiles of CYP3A genes were consistent with those seen in humans. We further generated mice carrying the CYP3A-HAC in the background homozygous for targeted deletion of most endogenous Cyp3a genes. In this mouse strain with 'fully humanized' CYP3A genes, the kinetics of triazolam metabolism, CYP3A-mediated mechanism-based inactivation effects and formation of fetal-specific metabolites of dehydroepiandrosterone observed in humans were well reproduced. Thus, these mice are likely to be valuable in evaluating novel drugs metabolized by CYP3A enzymes and in studying the regulation of human CYP3A gene expression. Furthermore, this system can also be used for generating Tc mice carrying other human metabolic genes.
Collapse
Affiliation(s)
- Yasuhiro Kazuki
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|