1
|
Schäfer AM, Rysz MA, Schädeli J, Hübscher M, Khosravi H, Fehr M, Seibert I, Potterat O, Smieško M, Meyer Zu Schwabedissen HE. St. John's Wort Formulations Induce Rat CYP3A23-3A1 Independent of Their Hyperforin Content. Mol Pharmacol 2023; 105:14-22. [PMID: 37863663 DOI: 10.1124/molpharm.123.000725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/08/2023] [Accepted: 09/21/2023] [Indexed: 10/22/2023] Open
Abstract
The pregnane X receptor (PXR) is a ligand-activated regulator of cytochrome P450 (CYP)3A enzymes. Among the ligands of human PXR is hyperforin, a constituent of St John's wort (SJW) extracts and potent inducer of human CYP3A4. It was the aim of this study to compare the effect of hyperforin and SJW formulations controlled for its content on CYP3A23-3A1 in rats. Hyperiplant was used as it contains a high hyperforin content and Rebalance because it is controlled for a low hyperforin content. In silico analysis revealed a weak hyperforin-rPXR binding affinity, which was further supported in cell-based reporter gene assays showing no hyperforin-mediated reporter activation in presence of rPXR. However, cellular exposure to Hyperiplant and Rebalance transactivated the CYP3A reporter 3.8-fold and 2.8-fold, respectively, and they induced Cyp3a23-3a1 mRNA expression in rat hepatoma cells compared with control 48-fold and 18-fold, respectively. In Wistar rats treated for 10 days with 400 mg/kg of Hyperiplant, we observed 1.8 times the Cyp3a23-3a1 mRNA expression, a 2.6-fold higher CYP3A23-3A1 protein amount, and a 1.6-fold increase in activity compared with controls. For Rebalance we only observed a 1.8-fold hepatic increase of CYP3A23-3A1 protein compared with control animals. Even though there are differing effects on rCyp3a23-3a1/CYP3A23-3A1 in rat liver reflecting the hyperforin content of the SJW extracts, the modulation is most likely not linked to an interaction of hyperforin with rPXR. SIGNIFICANCE STATEMENT: Treatment with St John's wort (SJW) has been reported to affect CYP3A expression and activity in rats. Our comparative study further supports this finding but shows that the pregnane X receptor-ligand hyperforin is not the driving force for changes in rat CYP3A23-3A1 expression and function in vivo and in vitro. Importantly, CYP3A induction mimics findings in humans, but our results suggest that another so far unknown constituent of SJW is responsible for the expression- and function-modifying effects in rat liver.
Collapse
Affiliation(s)
- Anima M Schäfer
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Marta A Rysz
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Julia Schädeli
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Michelle Hübscher
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Haleh Khosravi
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Michelle Fehr
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Isabell Seibert
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Olivier Potterat
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Martin Smieško
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Henriette E Meyer Zu Schwabedissen
- Biopharmacy (A.M.S., M.A.R., J.S., M.H., H.K., M.F., I.S., H.E.M.), Computational Pharmacy (M.S.), and Pharmaceutical Biology (O.P.), Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
2
|
Men S, Wang H. Phenobarbital in Nuclear Receptor Activation: An Update. Drug Metab Dispos 2023; 51:210-218. [PMID: 36351837 PMCID: PMC9900862 DOI: 10.1124/dmd.122.000859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/11/2022] Open
Abstract
Phenobarbital (PB) is a commonly prescribed anti-epileptic drug that can also benefit newborns from hyperbilirubinemia. Being the first drug demonstrating hepatic induction of cytochrome P450 (CYP), PB has since been broadly used as a model compound to study xenobiotic-induced drug metabolism and clearance. Mechanistically, PB-mediated CYP induction is linked to a number of nuclear receptors, such as the constitutive androstane receptor (CAR), pregnane X receptor (PXR), and estrogen receptor α, with CAR being the predominant regulator. Unlike prototypical agonistic ligands, PB-mediated activation of CAR does not involve direct binding with the receptor. Instead, dephosphorylation of threonine 38 in the DNA-binding domain of CAR was delineated as a key signaling event underlying PB-mediated indirect activation of CAR. Further studies revealed that such phosphorylation sites appear to be highly conserved among most human nuclear receptors. Interestingly, while PB is a pan-CAR activator in both animals and humans, PB activates human but not mouse PXR. The species-specific role of PB in gene regulation is a key determinant of its implication in xenobiotic metabolism, drug-drug interactions, energy homeostasis, and cell proliferation. In this review, we summarize the recent progress in our understanding of PB-provoked transactivation of nuclear receptors with a focus on CAR and PXR. SIGNIFICANCE STATEMENT: Extensive studies using PB as a research tool have significantly advanced our understanding of the molecular basis underlying nuclear receptor-mediated drug metabolism, drug-drug interactions, energy homeostasis, and cell proliferation. In particular, CAR has been established as a cell signaling-regulated nuclear receptor in addition to ligand-dependent functionality. This mini-review highlights the mechanisms by which PB transactivates CAR and PXR.
Collapse
Affiliation(s)
- Shuaiqian Men
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (S.M., H.W.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (S.M., H.W.)
| |
Collapse
|
3
|
Dutta M, Lim JJ, Cui JY. Pregnane X Receptor and the Gut-Liver Axis: A Recent Update. Drug Metab Dispos 2022; 50:478-491. [PMID: 34862253 PMCID: PMC11022899 DOI: 10.1124/dmd.121.000415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023] Open
Abstract
It is well-known that the pregnane X receptor (PXR)/Nr1i2 is a critical xenobiotic-sensing nuclear receptor enriched in liver and intestine and is responsible for drug-drug interactions, due to its versatile ligand binding domain (LBD) and target genes involved in xenobiotic biotransformation. PXR can be modulated by various xenobiotics including pharmaceuticals, nutraceuticals, dietary factors, and environmental chemicals. Microbial metabolites such as certain secondary bile acids (BAs) and the tryptophan metabolite indole-3-propionic acid (IPA) are endogenous PXR activators. Gut microbiome is increasingly recognized as an important regulator for host xenobiotic biotransformation and intermediary metabolism. PXR regulates and is regulated by the gut-liver axis. This review summarizes recent research advancements leveraging pharmaco- and toxico-metagenomic approaches that have redefined the previous understanding of PXR. Key topics covered in this review include: (1) genome-wide investigations on novel PXR-target genes, novel PXR-DNA interaction patterns, and novel PXR-targeted intestinal bacteria; (2) key PXR-modulating activators and suppressors of exogenous and endogenous sources; (3) novel bidirectional interactions between PXR and gut microbiome under physiologic, pathophysiological, pharmacological, and toxicological conditions; and (4) modifying factors of PXR-signaling including species and sex differences and time (age, critical windows of exposure, and circadian rhythm). The review also discusses critical knowledge gaps and important future research topics centering around PXR. SIGNIFICANCE STATEMENT: This review summarizes recent research advancements leveraging O'mics approaches that have redefined the previous understanding of the xenobiotic-sensing nuclear receptor pregnane X receptor (PXR). Key topics include: (1) genome-wide investigations on novel PXR-targeted host genes and intestinal bacteria as well as novel PXR-DNA interaction patterns; (2) key PXR modulators including microbial metabolites under physiological, pathophysiological, pharmacological, and toxicological conditions; and (3) modifying factors including species, sex, and time.
Collapse
Affiliation(s)
- Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
4
|
Lin W, Bwayi M, Wu J, Li Y, Chai SC, Huber AD, Chen T. CITCO Directly Binds to and Activates Human Pregnane X Receptor. Mol Pharmacol 2020; 97:180-190. [PMID: 31882411 PMCID: PMC6978709 DOI: 10.1124/mol.119.118513] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
The xenobiotic receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are activated by structurally diverse chemicals to regulate the expression of target genes, and they have overlapping regulation in terms of ligands and target genes. Receptor-selective agonists are, therefore, critical for studying the overlapping function of PXR and CAR. An early effort identified 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO) as a selective human CAR (hCAR) agonist, and this has since been widely used to distinguish the function of hCAR from that of human PXR (hPXR). The selectivity was demonstrated in a green monkey kidney cell line, CV-1, in which CITCO displayed >100-fold selectivity for hCAR over hPXR. However, whether the selectivity observed in CV-1 cells also represented CITCO activity in liver cell models was not hitherto investigated. In this study, we showed that CITCO: 1) binds directly to hPXR; 2) activates hPXR in HepG2 cells, with activation being blocked by an hPXR-specific antagonist, SPA70; 3) does not activate mouse PXR; 4) depends on tryptophan-299 to activate hPXR; 5) recruits steroid receptor coactivator 1 to hPXR; 6) activates hPXR in HepaRG cell lines even when hCAR is knocked out; and 7) activates hPXR in primary human hepatocytes. Together, these data indicate that CITCO binds directly to the hPXR ligand-binding domain to activate hPXR. As CITCO has been widely used, its confirmation as a dual agonist for hCAR and hPXR is important for appropriately interpreting existing data and designing future experiments to understand the regulation of hPXR and hCAR. SIGNIFICANCE STATEMENT: The results of this study demonstrate that 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO) is a dual agonist for human constitutive androstane receptor (hCAR) and human pregnane X receptor (hPXR). As CITCO has been widely used to activate hCAR, and hPXR and hCAR have distinct and overlapping biological functions, these results highlight the value of receptor-selective agonists and the importance of appropriately interpreting data in the context of receptor selectivity of such agonists.
Collapse
Affiliation(s)
- Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Monicah Bwayi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yongtao Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Andrew D Huber
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
5
|
Administration of Vitamin D Metabolites Affects RNA Expression of Xenobiotic Metabolising Enzymes and Function of ABC Transporters in Rats. J CHEM-NY 2019. [DOI: 10.1155/2019/1279036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
From studies on different species and in cell culture systems, it has been suggested that vitamin D metabolites might affect the metabolism and elimination of xenobiotics. Although most studies performed on rodents and cell cultures report an upregulation of respective enzymes and transporters, data from the literature are inconsistent. Especially results obtained with sheep differ from these observations. As vitamin D metabolites are widely used as feed additives or therapeutics in livestock animals, we aimed to assess whether these differences indicate species-specific responses or occurred due to the very high dosages used in the rodent studies. Therefore, we applied treatment protocols to rats that had been used previously in sheep or cattle. Forty-eight female rats were divided into three treatment and corresponding placebo groups: (1) a single intraperitoneal injection of 1,25-(OH)2D3 or placebo 12 h before sacrifice; (2) daily supplementation with 25-OHD3 by oral gavage or placebo for 10 days; and (3) a single intramuscular injection of vitamin D3 10 days before sacrifice. In contrast to a previous study using sheep, treatment of rats with 1,25-dihydroxyvitamin D3 did not result in an upregulation of cytochrome P450 3A isoenzymes (CYP3A), but a decrease was found in hepatic and intestinal expressions. In addition, a downregulation of P-glycoprotein (P-gp) and breast cancer resistance protein was found in the brain. Taken together, the stimulating effects of vitamin D metabolites on the expression of genes involved in the metabolism and elimination of xenobiotics reported previously for rodents and sheep could not be reproduced. In contrast, we even observed a negative impact on the expression of CYP3A enzymes and their most important regulator, the pregnane X receptor. Most interestingly, we could demonstrate an effect of treatment with 25-hydroxyvitamin D3 and vitamin D3 on the functional activity of ileal P-glycoprotein (P-gp) using the Ussing chamber technique.
Collapse
|
6
|
Mackowiak B, Hodge J, Stern S, Wang H. The Roles of Xenobiotic Receptors: Beyond Chemical Disposition. Drug Metab Dispos 2018; 46:1361-1371. [PMID: 29759961 PMCID: PMC6124513 DOI: 10.1124/dmd.118.081042] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/07/2018] [Indexed: 02/06/2023] Open
Abstract
Over the past 20 years, the ability of the xenobiotic receptors to coordinate an array of drug-metabolizing enzymes and transporters in response to endogenous and exogenous stimuli has been extensively characterized and well documented. The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) are the xenobiotic receptors that have received the most attention since they regulate the expression of numerous proteins important to drug metabolism and clearance and formulate a central defensive mechanism to protect the body against xenobiotic challenges. However, accumulating evidence has shown that these xenobiotic sensors also control many cellular processes outside of their traditional realms of xenobiotic metabolism and disposition, including physiologic and/or pathophysiologic responses in energy homeostasis, cell proliferation, inflammation, tissue injury and repair, immune response, and cancer development. This review will highlight recent advances in studying the noncanonical functions of xenobiotic receptors with a particular focus placed on the roles of CAR and PXR in energy homeostasis and cancer development.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Jessica Hodge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| |
Collapse
|
7
|
Wang S, Xie X, Lei T, Zhang K, Lai B, Zhang Z, Guan Y, Mao G, Xiao L, Wang N. Statins Attenuate Activation of the NLRP3 Inflammasome by Oxidized LDL or TNF α in Vascular Endothelial Cells through a PXR-Dependent Mechanism. Mol Pharmacol 2017; 92:256-264. [PMID: 28546421 DOI: 10.1124/mol.116.108100] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 05/22/2017] [Indexed: 12/11/2022] Open
Abstract
Excessive activation of the NLRP3 inflammasome is implicated in cardiovascular diseases. Statins exert an anti-inflammatory effect independent of their cholesterol-lowering effect. This study investigated the potential role of statins in the activation of the NLRP3 inflammasome in endothelial cells (ECs). Western blotting and quantitative reverse-transcription polymerase chain reaction showed that oxidized low-density lipoprotein (ox-LDL) or tumor necrosis factor α (TNFα) activated the NLRP3 inflammasome in ECs. Simvastatin or mevastatin significantly suppressed the effects of ox-LDL or TNFα Promoter reporter assays and small interfering RNA knockdown revealed that statins inhibit ox-LDL-mediated NLRP3 inflammasome activation via the pregnane X receptor (PXR). In addition, PXR agonists (rifampicin and SR12813) or overexpression of a constitutively active PXR markedly suppressed the NLRP3 inflammasome activation. Conversely, PXR knockdown abrogated the suppressive effect of rifampicin on NLRP3 inflammasome activation. Knockdown of lectin-like ox-LDL receptor or overexpression of IκBα-attenuated ox-LDL- or TNFα-triggered activation of the NLRP3 inflammasome. Chromatin immunoprecipitation assays indicated that mevastatin inhibited nuclear factor-κB binding to the promoter regions of the human NLRP3 gene. Collectively, these results demonstrate that the statin activation of PXR inhibits the activation of NLRP3 inflammasome in response to atherogenic stimuli such as ox-LDL and TNFα in ECs, providing a new mechanism for the cardiovascular benefit of statins.
Collapse
MESH Headings
- Cells, Cultured
- Endothelial Cells/metabolism
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Lipoproteins, LDL/antagonists & inhibitors
- Lipoproteins, LDL/pharmacology
- NF-kappa B/physiology
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/physiology
- Pregnane X Receptor
- Promoter Regions, Genetic
- Receptors, Steroid/agonists
- Receptors, Steroid/physiology
- Scavenger Receptors, Class E/physiology
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Shaolan Wang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Xinya Xie
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Ting Lei
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Kang Zhang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Baochang Lai
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Zihui Zhang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Youfei Guan
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Guangmei Mao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Lei Xiao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Nanping Wang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| |
Collapse
|
8
|
Chang JH, Chen J, Liu L, Messick K, Ly J. Rifampin-Mediated Induction of Tamoxifen Metabolism in a Humanized PXR-CAR-CYP3A4/3A7-CYP2D6 Mouse Model. Drug Metab Dispos 2016; 44:1736-1741. [PMID: 27538915 DOI: 10.1124/dmd.116.072132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023] Open
Abstract
Animals are not commonly used to assess drug-drug interactions due to poor clinical translatability arising from species differences that may exist in drug-metabolizing enzymes and transporters, and their regulation pathways. In this study, a transgenic mouse model expressing human pregnane X receptor (PXR), constitutive androstane receptor (CAR), CYP3A4/CYP3A7, and CYP2D6 (Tg-composite) was used to investigate the effect of induction mediated by rifampin on the pharmacokinetics of tamoxifen and its metabolites. In humans, tamoxifen is metabolized primarily by CYP3A4 and CYP2D6, and multiple-day treatment with rifampin decreased tamoxifen exposure by 6.2-fold. Interestingly, exposure of tamoxifen metabolites 4-hydroxytamoxifen (4OHT), N-desmethyltamoxifen (NDM), and endoxifen also decreased. In the Tg-composite model, pretreatment with rifampin decreased tamoxifen area under the time-concentration curve between 0 and 8 hours (AUC0-8) from 0.82 to 0.20 µM*h, whereas AUC0-8 of 4OHT, NDM, and endoxifen decreased by 3.4-, 4.7-, and 1.3-fold, respectively, mirroring the clinic observations. In the humanized PXR-CAR (hPXR-CAR) model, rifampin decreased AUC0-8 of tamoxifen and its metabolites by approximately 2-fold. In contrast, no significant modulation by rifampin was observed in the nonhumanized C57BL/6 (wild-type) animals. In vitro kinetics determined in microsomes prepared from livers of the Tg-composite animals showed that, although Km values were not different between vehicle- and rifampin-treated groups, rifampin increased the Vmax for the CYP3A4-mediated pathways. These data demonstrate that, although the hPXR-CAR model is responsive to rifampin, the extent of the clinical rifampin-tamoxifen interaction is better represented by the Tg-composite model. Consequently, the Tg-composite model may be a suitable tool to examine the extent of rifampin-mediated induction for other compounds whose metabolism is mediated by CYP3A4 and/or CYP2D6.
Collapse
Affiliation(s)
- Jae H Chang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - John Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - Liling Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - Kirsten Messick
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - Justin Ly
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| |
Collapse
|
9
|
Garg A, Zhao A, Erickson SL, Mukherjee S, Lau AJ, Alston L, Chang TKH, Mani S, Hirota SA. Pregnane X Receptor Activation Attenuates Inflammation-Associated Intestinal Epithelial Barrier Dysfunction by Inhibiting Cytokine-Induced Myosin Light-Chain Kinase Expression and c-Jun N-Terminal Kinase 1/2 Activation. J Pharmacol Exp Ther 2016; 359:91-101. [PMID: 27440420 PMCID: PMC5034705 DOI: 10.1124/jpet.116.234096] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
The inflammatory bowel diseases (IBDs) are chronic inflammatory disorders with a complex etiology. IBD is thought to arise in genetically susceptible individuals in the context of aberrant interactions with the intestinal microbiota and other environmental risk factors. Recently, the pregnane X receptor (PXR) was identified as a sensor for microbial metabolites, whose activation can regulate the intestinal epithelial barrier. Mutations in NR1I2, the gene that encodes the PXR, have been linked to IBD, and in animal models, PXR deletion leads to barrier dysfunction. In the current study, we sought to assess the mechanism(s) through which the PXR regulates barrier function during inflammation. In Caco-2 intestinal epithelial cell monolayers, tumor necrosis factor-α/interferon-γ exposure disrupted the barrier and triggered zonula occludens-1 relocalization, increased expression of myosin light-chain kinase (MLCK), and activation of c-Jun N-terminal kinase 1/2 (JNK1/2). Activation of the PXR [rifaximin and [[3,5-Bis(1,1-dimethylethyl)-4-hydroxyphenyl]ethenylidene]bis-phosphonic acid tetraethyl ester (SR12813); 10 μM] protected the barrier, an effect that was associated with attenuated MLCK expression and JNK1/2 activation. In vivo, activation of the PXR [pregnenolone 16α-carbonitrile (PCN)] attenuated barrier disruption induced by toll-like receptor 4 activation in wild-type, but not Pxr-/-, mice. Furthermore, PCN treatment protected the barrier in the dextran-sulfate sodium model of experimental colitis, an effect that was associated with reduced expression of mucosal MLCK and phosphorylated JNK1/2. Together, our data suggest that the PXR regulates the intestinal epithelial barrier during inflammation by modulating cytokine-induced MLCK expression and JNK1/2 activation. Thus, targeting the PXR may prove beneficial for the treatment of inflammation-associated barrier disruption in the context of IBD.
Collapse
Affiliation(s)
- Aditya Garg
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Angela Zhao
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Sarah L Erickson
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Subhajit Mukherjee
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Aik Jiang Lau
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Laurie Alston
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Thomas K H Chang
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Sridhar Mani
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| | - Simon A Hirota
- Department of Physiology and Pharmacology (A.G., A.Z., S.L.E., L.A., S.A.H.), and Department of Microbiology, Immunology and Infectious Diseases (S.A.H.), University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (Su.M., Sr.M.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.J.L., T.K.H.C.)
| |
Collapse
|
10
|
Prough RA, Clark BJ, Klinge CM. Novel mechanisms for DHEA action. J Mol Endocrinol 2016; 56:R139-55. [PMID: 26908835 DOI: 10.1530/jme-16-0013] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/02/2023]
Abstract
Dehydroepiandrosterone (3β-hydroxy-5-androsten-17-one, DHEA), secreted by the adrenal cortex, gastrointestinal tract, gonads, and brain, and its sulfated metabolite DHEA-S are the most abundant endogeneous circulating steroid hormones. DHEA actions are classically associated with age-related changes in cardiovascular tissues, female fertility, metabolism, and neuronal/CNS functions. Early work on DHEA action focused on the metabolism to more potent sex hormones, testosterone and estradiol, and the subsequent effect on the activation of the androgen and estrogen steroid receptors. However, it is now clear that DHEA and DHEA-S act directly as ligands for many hepatic nuclear receptors and G-protein-coupled receptors. In addition, it can function to mediate acute cell signaling pathways. This review summarizes the molecular mechanisms by which DHEA acts in cells and animal models with a focus on the 'novel' and physiological modes of DHEA action.
Collapse
Affiliation(s)
- Russell A Prough
- Department of Biochemistry and Molecular GeneticsCenter for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Barbara J Clark
- Department of Biochemistry and Molecular GeneticsCenter for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular GeneticsCenter for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
11
|
Cherian MT, Lin W, Wu J, Chen T. CINPA1 is an inhibitor of constitutive androstane receptor that does not activate pregnane X receptor. Mol Pharmacol 2015; 87:878-89. [PMID: 25762023 PMCID: PMC4407736 DOI: 10.1124/mol.115.097782] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/11/2015] [Indexed: 11/22/2022] Open
Abstract
Constitutive androstane receptor (CAR) and pregnane X receptor (PXR) are xenobiotic sensors that enhance the detoxification and elimination of xenobiotics and endobiotics by modulating the expression of genes encoding drug-metabolizing enzymes and transporters. Elevated levels of drug-metabolizing enzymes and efflux transporters, resulting from CAR activation in various cancers, promote the elimination of chemotherapeutic agents, leading to reduced therapeutic effectiveness and acquired drug resistance. CAR inhibitors, in combination with existing chemotherapeutics, could therefore be used to attenuate multidrug resistance in cancers. Interestingly, all previously reported CAR inverse-agonists are also activators of PXR, rendering them mechanistically counterproductive in tissues where both these xenobiotic receptors are present and active. We used a directed high-throughput screening approach, followed by subsequent mechanistic studies, to identify novel, potent, and specific small-molecule CAR inhibitors that do not activate PXR. We describe here one such inhibitor, CINPA1 (CAR inhibitor not PXR activator 1), capable of reducing CAR-mediated transcription with an IC50 of ∼70 nM. CINPA1 1) is a specific xenobiotic receptor inhibitor and has no cytotoxic effects up to 30 µM; 2) inhibits CAR-mediated gene expression in primary human hepatocytes, where CAR is endogenously expressed; 3) does not alter the protein levels or subcellular localization of CAR; 4) increases corepressor and reduces coactivator interaction with the CAR ligand-binding domain in mammalian two-hybrid assays; and 5) disrupts CAR binding to the promoter regions of target genes in chromatin immunoprecipitation assays. CINPA1 could be used as a novel molecular tool for understanding CAR function.
Collapse
Affiliation(s)
- Milu T Cherian
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
12
|
Dagnino S, Bellet V, Grimaldi M, Riu A, Aït-Aïssa S, Cavaillès V, Fenet H, Balaguer P. Affinity purification using recombinant PXR as a tool to characterize environmental ligands. ENVIRONMENTAL TOXICOLOGY 2014; 29:207-215. [PMID: 22223399 DOI: 10.1002/tox.20787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 10/27/2011] [Accepted: 11/06/2011] [Indexed: 05/31/2023]
Abstract
Many environmental endocrine disrupting compounds act as ligands for nuclear receptors. The human pregnane X receptor (hPXR), for instance, is activated by a variety of environmental ligands such as steroids, pharmaceutical drugs, pesticides, alkylphenols, polychlorinated biphenyls and polybromo diethylethers. Some of us have previously reported the occurrence of hPXR ligands in environmental samples but failed to identify them. The aim of this study was to test whether a PXR-affinity column, in which recombinant hPXR was immobilized on solid support, could help the purification of these chemicals. Using PXR ligands of different affinity (10 nM < EC50 < 10 μM), we demonstrated that the PXR-affinity preferentially column captured ligands with medium to high affinities (EC50 < 1 μM). Furthermore, by using the PXR-affinity column to analyze an environmental sample containing ERα, AhR, AR, and PXR activities, we show that (i) half of the PXR activity of the sample was due to compounds with medium to high affinity for PXR and (ii) PXR shared ligands with ERα, AR, and AhR. These findings demonstrate that the newly developed PXR-affinity column coupled to reporter cell lines represents a valuable tool for the characterization of the nature of PXR active compounds and should therefore guide and facilitate their further analysis.
Collapse
Affiliation(s)
- Sonia Dagnino
- IRCM, Institut de Recherche en Cancerologie de Montpellier, CRLC Val d'Aurelle Paul Lamarque, INSERM, U896 34298 Montpellier, France; Université Montpellier 1, 34298 Montpellier, France; UMR 5569 Hydrosciences, Université Montpellier I, 34060 Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Rathod V, Jain S, Nandekar P, Sangamwar AT. Human pregnane X receptor: a novel target for anticancer drug development. Drug Discov Today 2014; 19:63-70. [DOI: 10.1016/j.drudis.2013.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/02/2013] [Accepted: 08/15/2013] [Indexed: 02/07/2023]
|
14
|
Lin W, Chen T. A vinblastine fluorescent probe for pregnane X receptor in a time-resolved fluorescence resonance energy transfer assay. Anal Biochem 2013; 443:252-60. [PMID: 24044991 DOI: 10.1016/j.ab.2013.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 12/20/2022]
Abstract
The pregnane X receptor (PXR) regulates the metabolism and excretion of xenobiotics and endobiotics by regulating the expression of drug-metabolizing enzymes and transporters. The unique structure of PXR allows the binding of many drugs and drug leads to it, possibly causing undesired drug-drug interactions. Therefore, it is crucial to evaluate whether lead compounds bind to PXR. Fluorescence-based assays are preferred because of their sensitivity and nonradioactive nature. One fluorescent PXR probe is currently commercially available; however, because its chemical structure is not publicly disclosed, it is not optimal for studying ligand-PXR interactions. Here we report the characterization of BODIPY FL-vinblastine, generated by labeling vinblastine with the fluorophore 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene (BODIPY FL), as a high-affinity ligand for human PXR with a Kd value of 673 nM. We provide evidence that BODIPY FL-vinblastine is a unique chemical entity different from either vinblastine or the fluorophore BODIPY FL in its function as a high-affinity human PXR ligand. We describe a BODIPY FL-vinblastine-based human PXR time-resolved fluorescence resonance energy transfer assay, which was used to successfully test a panel of human PXR ligands. The BODIPY FL-vinblastine-based biochemical assay is suitable for high-throughput screening to evaluate whether lead compounds bind to PXR.
Collapse
Affiliation(s)
- Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
15
|
Ma Y, Sun SX, Cheng XC, Wang SQ, Dong WL, Wang RL, Xu WR. Design and Synthesis of Imidazolidine-2,4-Dione Derivatives as Selective Inhibitors by Targeting Protein Tyrosine Phosphatase-1B Over T-Cell Protein Tyrosine Phosphatase. Chem Biol Drug Des 2013; 82:595-602. [DOI: 10.1111/cbdd.12189] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/22/2013] [Accepted: 07/09/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics); School of Pharmacy; Tianjin Medical University; Tianjin; 300070; China
| | - Su-Xia Sun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics); School of Pharmacy; Tianjin Medical University; Tianjin; 300070; China
| | - Xian-Chao Cheng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics); School of Pharmacy; Tianjin Medical University; Tianjin; 300070; China
| | - Shu-Qing Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics); School of Pharmacy; Tianjin Medical University; Tianjin; 300070; China
| | - Wei-Li Dong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics); School of Pharmacy; Tianjin Medical University; Tianjin; 300070; China
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics); School of Pharmacy; Tianjin Medical University; Tianjin; 300070; China
| | - Wei-Ren Xu
- Tianjin Institute of Pharmaceutical Research (TIPR); Tianjin; 300193; China
| |
Collapse
|
16
|
Modulation of xenobiotic receptors by steroids. Molecules 2013; 18:7389-406. [PMID: 23884115 PMCID: PMC3777271 DOI: 10.3390/molecules18077389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/13/2013] [Accepted: 06/19/2013] [Indexed: 12/13/2022] Open
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors that regulate the expression of their target genes. NRs play important roles in many human diseases, including metabolic diseases and cancer, and are therefore a key class of therapeutic targets. Steroids play important roles in regulating nuclear receptors; in addition to being ligands of steroid receptors, steroids (and their metabolites) also regulate other NRs, such as the pregnane X receptor and constitutive androstane receptor (termed xenobiotic receptors), which participate in steroid metabolism. Xenobiotic receptors have promiscuous ligand-binding properties, and their structurally diverse ligands include steroids and their metabolites. Therefore, steroids, their metabolism and metabolites, xenobiotic receptors, steroid receptors, and the respective signaling pathways they regulate have functional interactions. This review discusses these functional interactions and their implications for activities mediated by steroid receptors and xenobiotic receptors, focusing on steroids that modulate pathways involving the pregnane X receptor and constitutive androstane receptor. The emphasis of the review is on structure-function studies of xenobiotic receptors bound to steroid ligands.
Collapse
|
17
|
Sharma D, Lau AJ, Sherman MA, Chang TK. Agonism of human pregnane X receptor by rilpivirine and etravirine: Comparison with first generation non-nucleoside reverse transcriptase inhibitors. Biochem Pharmacol 2013; 85:1700-11. [DOI: 10.1016/j.bcp.2013.04.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/02/2013] [Accepted: 04/02/2013] [Indexed: 11/24/2022]
|
18
|
Avior Y, Bomze D, Ramon O, Nahmias Y. Flavonoids as dietary regulators of nuclear receptor activity. Food Funct 2013; 4:831-44. [PMID: 23598551 PMCID: PMC3781338 DOI: 10.1039/c3fo60063g] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolic diseases such as obesity, type II diabetes, and dyslipidemia are a rising cause of mortality worldwide. The progression of many metabolic diseases is fundamentally regulated on the transcriptional level by a family of ligand-activated transcription factors, called nuclear receptors, which detect and respond to metabolic changes. Their role in maintaining metabolic homeostasis makes nuclear receptors an important pharmaceutical and dietary target. This review will present the growing evidence that flavonoids, natural secondary plant metabolites, are important regulators of nuclear receptor activity. Structural similarities between flavonoids and cholesterol derivatives combined with the promiscuous nature of most nuclear receptors provide a wealth of possibilities for pharmaceutical and dietary modulation of metabolism. While the challenges of bringing flavonoid-derived therapeutics to the market are significant, we consider this rapidly growing field to be an essential aspect of the functional food initiative and an important mine for pharmaceutical compounds.
Collapse
Affiliation(s)
- Yishai Avior
- School of Computer Science and Engineering, Center for Bioengineering, The Hebrew University of Jerusalem, Edmond J. Safra Campus (Givat Ram), Silberman 3-512, Jerusalem 91904, Israel.
| | - David Bomze
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ory Ramon
- School of Computer Science and Engineering, Center for Bioengineering, The Hebrew University of Jerusalem, Edmond J. Safra Campus (Givat Ram), Silberman 3-512, Jerusalem 91904, Israel.
| | - Yaakov Nahmias
- School of Computer Science and Engineering, Center for Bioengineering, The Hebrew University of Jerusalem, Edmond J. Safra Campus (Givat Ram), Silberman 3-512, Jerusalem 91904, Israel.
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Ghonem NS, Boyer JL. Fibrates as adjuvant therapy for chronic cholestatic liver disease: its time has come. Hepatology 2013; 57:1691-3. [PMID: 23174993 PMCID: PMC4048949 DOI: 10.1002/hep.26155] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2012] [Indexed: 12/07/2022]
|
20
|
Minor role of pregnane-x-receptor for acquired multidrug resistance in head and neck squamous cell carcinoma in vitro. Cancer Chemother Pharmacol 2013; 71:1335-43. [DOI: 10.1007/s00280-013-2133-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/01/2013] [Indexed: 11/26/2022]
|
21
|
Michael Miller KK, Al-Rayyan N, Ivanova MM, Mattingly KA, Ripp SL, Klinge CM, Prough RA. DHEA metabolites activate estrogen receptors alpha and beta. Steroids 2013; 78:15-25. [PMID: 23123738 PMCID: PMC3529809 DOI: 10.1016/j.steroids.2012.10.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/17/2012] [Accepted: 10/17/2012] [Indexed: 11/22/2022]
Abstract
Dehydroepiandrosterone (DHEA) levels were reported to associate with increased breast cancer risk in postmenopausal women, but some carcinogen-induced rat mammary tumor studies question this claim. The purpose of this study was to determine how DHEA and its metabolites affect estrogen receptors α or β (ERα or ERβ)-regulated gene transcription and cell proliferation. In transiently transfected HEK-293 cells, androstenediol, DHEA, and DHEA-S activated ERα. In ERβ transfected HepG2 cells, androstenedione, DHEA, androstenediol, and 7-oxo DHEA stimulated reporter activity. ER antagonists ICI 182,780 (fulvestrant) and 4-hydroxytamoxifen, general P450 inhibitor miconazole, and aromatase inhibitor exemestane inhibited activation by DHEA or metabolites in transfected cells. ERβ-selective antagonist R,R-THC (R,R-cis-diethyl tetrahydrochrysene) inhibited DHEA and DHEA metabolite transcriptional activity in ERβ-transfected cells. Expression of endogenous estrogen-regulated genes: pS2, progesterone receptor, cathepsin D1, and nuclear respiratory factor-1 was increased by DHEA and its metabolites in an ER-subtype, gene, and cell-specific manner. DHEA metabolites, but not DHEA, competed with 17β-estradiol for ERα and ERβ binding and stimulated MCF-7 cell proliferation, demonstrating that DHEA metabolites interact directly with ERα and ERβin vitro, modulating estrogen target genes in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Carolyn M. Klinge
- CORRESPONDING AUTHORS: Russell A. Prough, Ph.D., and Carolyn M. Klinge. Ph.D. Department of Biochemistry & Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40292. Phone: (502) 852-7249 (RAP); 502-852-3668 (CMK); FAX: (502) 852-6222; and
| | - Russell A. Prough
- CORRESPONDING AUTHORS: Russell A. Prough, Ph.D., and Carolyn M. Klinge. Ph.D. Department of Biochemistry & Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40292. Phone: (502) 852-7249 (RAP); 502-852-3668 (CMK); FAX: (502) 852-6222; and
| |
Collapse
|
22
|
Jones RD, Taylor AM, Tong EY, Repa JJ. Carboxylesterases are uniquely expressed among tissues and regulated by nuclear hormone receptors in the mouse. Drug Metab Dispos 2013; 41:40-9. [PMID: 23011759 PMCID: PMC3533427 DOI: 10.1124/dmd.112.048397] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/25/2012] [Indexed: 11/22/2022] Open
Abstract
Carboxylesterases (CES) are a well recognized, yet incompletely characterized family of proteins that catalyze neutral lipid hydrolysis. Some CES have well-defined roles in xenobiotic clearance, pharmacologic prodrug activation, and narcotic detoxification. In addition, emerging evidence suggests other CES may have roles in lipid metabolism. Humans have six CES genes, whereas mice have 20 Ces genes grouped into five isoenzyme classes. Perhaps due to the high sequence similarity shared by the mouse Ces genes, the tissue-specific distribution of expression for these enzymes has not been fully addressed. Therefore, we performed studies to provide a comprehensive tissue distribution analysis of mouse Ces mRNAs. These data demonstrated that while the mouse Ces family 1 is highly expressed in liver and family 2 in intestine, many Ces genes have a wide and unique tissue distribution defined by relative mRNA levels. Furthermore, evaluating Ces gene expression in response to pharmacologic activation of lipid- and xenobiotic-sensing nuclear hormone receptors showed differential regulation. Finally, specific shifts in Ces gene expression were seen in peritoneal macrophages following lipopolysaccharide treatment and in a steatotic liver model induced by high-fat feeding, two model systems relevant to disease. Overall these data show that each mouse Ces gene has its own distinctive tissue expression pattern and suggest that some CES may have tissue-specific roles in lipid metabolism and xenobiotic clearance.
Collapse
Affiliation(s)
- Ryan D. Jones
- Departments of Physiology (R.D.J., A.M.T., E.Y.T., J.J.R.) and Internal Medicine (J.J.R.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Anna M. Taylor
- Departments of Physiology (R.D.J., A.M.T., E.Y.T., J.J.R.) and Internal Medicine (J.J.R.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ernest Y. Tong
- Departments of Physiology (R.D.J., A.M.T., E.Y.T., J.J.R.) and Internal Medicine (J.J.R.), University of Texas Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
23
|
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
|
24
|
Doricakova A, Novotna A, Vrzal R, Pavek P, Dvorak Z. The role of residues T248, Y249 and T422 in the function of human pregnane X receptor. Arch Toxicol 2012; 87:291-301. [PMID: 22976785 DOI: 10.1007/s00204-012-0937-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/28/2012] [Indexed: 10/27/2022]
Abstract
The pregnane X receptor (PXR) is a key xenobiotic receptor that regulates the expression of numerous drug-metabolizing enzymes. Some posttranslational mechanisms modulate its transcriptional activity. Although several kinases have been shown to directly phosphorylate this receptor, little is known about phosphorylation sites of PXR. In the present work, we examined T248, Y249 and T422 putative phosphorylation sites determined based on in silico consensus kinase site prediction analysis. T248 and T422 residues are critical for the interaction of the PXR ligand-binding domain and the activation function-2 (AF2) domain. Site-directed mutagenesis analysis was performed to generate phospho-deficient and phospho-mimetic mutants. We examined transactivation activity of the PXR mutants in gene reporter assays, formation of PXRmutant/RXRα heterodimer, binding of PXR mutants to the CYP3A4 gene response element DR3 and CYP3A4 expression in HepG2 cells after expression of the mutants. We found that T248D mutant activated CYP3A4 transactivation constitutively regardless of the presence or absence of a ligand. Contrary, T248V mutant exhibited low basal and ligand-inducible transactivation capacity as compared to wild-type PXR. Dose-response analysis revealed reduced ligand-dependent transactivation potency of PXR Y249D mutant. Transactivation of the CYP3A4 promoter was abolished with T422A/D mutants. All PXR mutants formed heterodimer with RXRα at a similar level to that observed with wild-type PXR. The ability to bind to DNA in vitro was substantially decreased in case of T248D, T422D and T248V mutants. Our data thus indicate that phosphorylation of T248, Y249 and T422 residues may be critical for the both basal and ligand-activated function of PXR.
Collapse
Affiliation(s)
- Aneta Doricakova
- Department of Cell Biology and Genetic, Palacky University Olomouc, Slechtitelu 11, 783 71 Olomouc, Czech Republic
| | | | | | | | | |
Collapse
|
25
|
QSAR model for human pregnane X receptor (PXR) binding: Screening of environmental chemicals and correlations with genotoxicity, endocrine disruption and teratogenicity. Toxicol Appl Pharmacol 2012; 262:301-9. [DOI: 10.1016/j.taap.2012.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 05/08/2012] [Accepted: 05/13/2012] [Indexed: 02/07/2023]
|
26
|
Jonker JW, Liddle C, Downes M. FXR and PXR: potential therapeutic targets in cholestasis. J Steroid Biochem Mol Biol 2012; 130:147-58. [PMID: 21801835 PMCID: PMC4750880 DOI: 10.1016/j.jsbmb.2011.06.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 05/17/2011] [Accepted: 06/17/2011] [Indexed: 12/17/2022]
Abstract
Cholestatic liver disorders encompass hepatobiliary diseases of diverse etiologies characterized by the accumulation of bile acids, bilirubin and cholesterol as the result of impaired secretion of bile. Members of the nuclear receptor (NR) family of ligand-modulated transcription factors are implicated in the adaptive response to cholestasis. NRs coordinately regulate bile acid and phospholipid transporter genes required for hepatobiliary transport, as well as the phases I and II metabolizing enzymes involved in processing of their substrates. In this review we will focus on FXR and PXR, two members of the NR family whose activities are regulated by bile acids. In addition, we also discuss the potential of pharmacological modulators of these receptors as novel therapies for cholestatic disorders.
Collapse
Affiliation(s)
- Johan W. Jonker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Corresponding author. Tel.: +31 050 361 1261; fax: +31 050 361 1746
| | - Christopher Liddle
- Storr Liver Unit, Westmead Millennium Institute and University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Michael Downes
- Gene Expression Laboratory, The Salk Institute for Biological Studies, Howard Hughes Medical Institute, 10010 Torrey Pines Road, La Jolla, CA 92037, USA
- Corresponding author. Tel.: +1 858 453 4100; fax: +1 858 455 1349
| |
Collapse
|
27
|
Lau AJ, Yang G, Yap CW, Chang TKH. Selective agonism of human pregnane X receptor by individual ginkgolides. Drug Metab Dispos 2012; 40:1113-21. [PMID: 22393123 DOI: 10.1124/dmd.112.045013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ginkgolide A, ginkgolide B, ginkgolide C, and ginkgolide J are structurally related terpene trilactones present in Ginkgo biloba extract. Pregnane X receptor (PXR), glucocorticoid receptor (GR), and constitutive androstane receptor (CAR) regulate the expression of genes involved in diverse biological functions. In the present study, we investigated the effects of individual ginkgolides as single chemical entities on the function of human PXR (hPXR), human GR (hGR), and human CAR (hCAR). In cell-based reporter gene assays, none of the ginkgolides activated hGR or hCAR (wild-type and its SV23, SV24, and SV25 splice variants). Concentration-response experiments showed that ginkgolide A and ginkgolide B activated hPXR and rat PXR to a greater extent than ginkgolide C, whereas ginkgolide J had no effect. As determined by a time-resolved fluorescence resonance energy transfer competitive binding assay, ginkgolide A and ginkgolide B, but not ginkgolide C or ginkgolide J, were shown to bind to the ligand-binding domain of hPXR, consistent with molecular docking data. Compared with tetraethyl 2-(3,5-di-tert-butyl-4-hydroxyphenyl)ethenyl-1,1-bisphosphonate (SR12813) (a known agonist of hPXR), ginkgolide A and ginkgolide B were considerably less potent in binding to hPXR. These two ginkgolides recruited steroid receptor coactivator-1 to hPXR and increased hPXR target gene (CYP3A4) expression, as assessed by a mammalian two-hybrid assay and real-time polymerase chain reaction, respectively. In conclusion, the individual ginkgolides regulate the function of nuclear receptors in a receptor-selective and chemical-dependent manner. This study identifies ginkgolide A and ginkgolide B as naturally occurring agonists of hPXR and provides mechanistic insight into the structure-activity relationship in ligand activation of hPXR.
Collapse
Affiliation(s)
- Aik Jiang Lau
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, BC V6T 1Z3, Canada.
| | | | | | | |
Collapse
|
28
|
Lau AJ, Yang G, Rajaraman G, Baucom CC, Chang TKH. Species-dependent and receptor-selective action of bilobalide on the function of constitutive androstane receptor and pregnane X receptor. Drug Metab Dispos 2012; 40:178-86. [PMID: 22019630 DOI: 10.1124/dmd.111.042879] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Bilobalide is a naturally occurring sesquiterpene trilactone with therapeutic potential in the management of ischemia and neurodegenerative diseases such as Alzheimer's disease. In the present study, we investigated the effect of bilobalide on the activity of rat constitutive androstane receptor (rCAR) and rat pregnane X receptor (rPXR) and compared that with human CAR (hCAR) and human PXR (hPXR). Bilobalide activated rCAR in a luciferase reporter gene assay and increased rCAR target gene expression in cultured rat hepatocytes, as determined by the CYP2B1 mRNA and CYP2B enzyme activity (benzyloxyresorufin O-dealkylation) assays. This increase in hepatocyte CYP2B1 expression by bilobalide was not accompanied by a corresponding increase in rCAR mRNA level. In contrast to the activation of rCAR, the activity of rPXR, hCAR, and hPXR was not influenced by this chemical in cell-based reporter gene assays. Consistent with these results, bilobalide did not alter rPXR, hCAR, or hPXR target gene expression in rat or human hepatocytes, as evaluated by the CYP3A23, CYP2B6, CYP3A4 mRNA assays and the CYP3A (testosterone 6β-hydroxylation) and CYP2B6 (bupropion hydroxylation) enzyme activity assays. Bilobalide was not an antagonist of rPXR, hCAR, or hPXR, as suggested by the finding that it did not attenuate rPXR activation by pregnenolone 16α-carbonitrile, hCAR activation by 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime, or hPXR activation by rifampicin in reporter gene assays. In conclusion, bilobalide is an activator of rCAR, whereas it is not a ligand of rPXR, hCAR, or hPXR. Likewise, it is an inducer of rat CYP2B1, but not of rat CYP3A23, human CYP2B6, or human CYP3A4.
Collapse
Affiliation(s)
- Aik Jiang Lau
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | | | | | | | | |
Collapse
|
29
|
Sekimoto M, Sano S, Hosaka T, Nemoto K, Degawa M. Establishment of a Stable Human Cell Line, HPL-A3, for Use in Reporter Gene Assays of Cytochrome P450 3A Inducers. Biol Pharm Bull 2012; 35:677-85. [DOI: 10.1248/bpb.35.677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masashi Sekimoto
- Department of Molecular Toxicology and Global Center of Excellence Program, School of Pharmaceutical Sciences, University of Shizuoka
| | - Shinsuke Sano
- Department of Molecular Toxicology and Global Center of Excellence Program, School of Pharmaceutical Sciences, University of Shizuoka
| | - Takuomi Hosaka
- Department of Molecular Toxicology and Global Center of Excellence Program, School of Pharmaceutical Sciences, University of Shizuoka
| | - Kiyomitsu Nemoto
- Department of Molecular Toxicology and Global Center of Excellence Program, School of Pharmaceutical Sciences, University of Shizuoka
| | - Masakuni Degawa
- Department of Molecular Toxicology and Global Center of Excellence Program, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
30
|
Shibayama Y, Iwashita Y, Yoshikawa Y, Kondo T, Ikeda R, Takeda Y, Osada T, Sugawara M, Yamada K, Iseki K. Effect of 5-fluorouracil treatment on SN-38 absorption from intestine in rats. Biol Pharm Bull 2011; 34:1418-25. [PMID: 21881227 DOI: 10.1248/bpb.34.1418] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
5-Fluorouracil (5-FU)-based chemotherapies with irinotecan have been applied for the treatment of cancers, and a common dose-limiting toxicity is neutropenia and diarrhea. In this study, we investigated the effect of 5-FU treatment on expression levels of drug transporters for SN-38 transportation and SN-38 absorption from the intestine following 5-FU treatment. Expression levels of several drug transporters and nuclear receptors in rats after 5-FU treatment were evaluated. SN-38 absorption from the intestine was evaluated by SN-38 concentration levels in serum following SN-38 injection into the intestine of 5-FU treated rats. The levels of renal multidrug resistance protein 2 (Mrp2) on day 4 after treatment (400 mg/kg) showed significant upregulation, 359.2 ± 33.2% (mean ± S.E.) of control. Mrp2 levels in the intestine were downregulated to 26.2 ± 8.4% of control. 5-FU treatment (400 mg/kg) also significantly downregurated expression levels of P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp) to 41.2 ± 14.7%, 15.7 ± 4.3% of control, respectively. To evaluate SN-38 absorption from the intestine, SN-38 was loaded in to the intestine on day 4 after 5-FU treatment. Pretreatment with 5-FU significantly increased SN-38 concentration in the blood 30, 60 and 90 min after SN-38 administration. The area under the curve for SN-38 in the 5-FU group was significantly higher than in vehicle groups. 5-FU treatment decreased expression levels of P-glycoprotein and Bcrp in intestine. The present study suggests that combination chemotherapy of 5-FU with irinotecan (CPT-11) may elevate SN-38 absorption from intestine.
Collapse
Affiliation(s)
- Yoshihiko Shibayama
- Education Research Center for Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Hokkaido University, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zafón C. [Evolutionary endocrinology: a pending matter]. ACTA ACUST UNITED AC 2011; 59:62-8. [PMID: 22113050 DOI: 10.1016/j.endonu.2011.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 08/05/2011] [Accepted: 09/01/2011] [Indexed: 12/22/2022]
Abstract
Twenty years have passed since the foundational article of what is now known as evolutionary medicine (EM) was published. This young medical discipline examines, following Darwinian principles, susceptibility to certain diseases and how we react to them. In short, EM analyzes the final cause of the disease from a historical perspective. Over the years, EM has been introduced in various medical areas in very different ways. While it has found a role in some fields such as infectious diseases and oncology, its contribution in other areas has been quite limited. In endocrinology, EM has only gained prominence as a basis for the so-called "diseases of civilization", including diabetes mellitus and obesity. However, many experts suggest that it may have a much higher potential. The aim of this paper is to provide a view about what evolutionary medicine is. Some examples of how EM may contribute to progress of our specialty are also given. There is no doubt that evolution enriches medicine, but medicine also offers knowledge to evolution.
Collapse
Affiliation(s)
- Carles Zafón
- Servicio de Endocrinología y Nutrición, Hospital Universitari Vall d'Hebron, Barcelona, España.
| |
Collapse
|
32
|
Rigalli JP, Ruiz ML, Perdomo VG, Villanueva SSM, Mottino AD, Catania VA. Pregnane X receptor mediates the induction of P-glycoprotein by spironolactone in HepG2 cells. Toxicology 2011; 285:18-24. [DOI: 10.1016/j.tox.2011.03.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 03/22/2011] [Accepted: 03/25/2011] [Indexed: 02/05/2023]
|
33
|
van Waterschoot RAB, Schinkel AH. A critical analysis of the interplay between cytochrome P450 3A and P-glycoprotein: recent insights from knockout and transgenic mice. Pharmacol Rev 2011; 63:390-410. [PMID: 21490128 DOI: 10.1124/pr.110.002584] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
CYP3A is one of the most important drug-metabolizing enzymes, determining the first-pass metabolism, oral bioavailability, and elimination of many drugs. It is also an important determinant of variable drug exposure and is involved in many drug-drug interactions. Recent studies with CYP3A knockout and transgenic mice have yielded a number of key insights that are important to consider during drug discovery and development. For instance, studies with tissue-specific CYP3A-transgenic mice have highlighted the importance of intestinal CYP3A-dependent metabolism. They also revealed that intestinal CYP3A plays an important role in the regulation of various drug-handling systems in the liver. Intestinal CYP3A activity can thus have far-reaching pharmacological effects. Besides CYP3A, the active drug efflux transporter P-glycoprotein also has a strong effect on the pharmacokinetics of numerous drugs. CYP3A and P-glycoprotein have an extensive overlap in their substrate spectrum. It has been hypothesized that for many drugs, the combined activity of CYP3A and P-glycoprotein makes for efficient intestinal first-pass metabolism of orally administered drugs as a result of a potentially synergistic collaboration. However, there is only limited in vitro and in vivo evidence for this hypothesis. There has also been some confusion in the field about what synergy actually means in this case. Our recent studies with Cyp3a/P-glycoprotein combination knockout mice have provided further insights into the CYP3A-P-glycoprotein interplay. We here present our view of the status of the synergy hypothesis and an attempt to clarify the existing confusion about synergy. We hope that this will facilitate further critical testing of the hypothesis and improve communication among researchers. Above all, the recent findings and insights into the interplay between CYP3A and P-glycoprotein may have implications for improving oral drug bioavailability and reducing adverse side effects.
Collapse
Affiliation(s)
- Robert A B van Waterschoot
- Division of Molecular Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
34
|
Staudinger JL, Xu C, Biswas A, Mani S. Post-translational modification of pregnane x receptor. Pharmacol Res 2011; 64:4-10. [PMID: 21397695 DOI: 10.1016/j.phrs.2011.02.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 02/24/2011] [Accepted: 02/27/2011] [Indexed: 01/07/2023]
Abstract
Pregnane x receptor (PXR, NR1I2) was originally characterized as a broad spectrum entero-hepatic xenobiotic 'sensor' and master-regulator of drug inducible gene expression. A compelling description of ligand-mediated gene activation has been unveiled in the last decade that firmly establishes this receptor's central role in the metabolism and transport of xenobiotics in mammals. Interestingly, pharmacotherapy with potent PXR ligands produces several profound side effects including decreased capacities for gluconeogenesis, lipid metabolism, and inflammation; likely due to PXR-mediated repression of gene expression programs underlying these pivotal physiological functions. An integrated model is emerging that reveals a sophisticated interplay between ligand binding and the ubiquitylation, phosphorylation, SUMOylation, and acetylation status of this important nuclear receptor protein. These discoveries point to a key role for the post-translational modification of PXR in the selective suppression of gene expression, and open the door to the study of completely new modes of regulation of the biological activity of PXR.
Collapse
Affiliation(s)
- Jeff L Staudinger
- Pharmacology and Toxicology, University of Kansas, 1251 Wescoe Hall Dr., 5038a Malott Hall, Lawrence, KA 66045, USA.
| | | | | | | |
Collapse
|
35
|
Lau AJ, Yang G, Rajaraman G, Baucom CC, Chang TKH. Differential effect of meclizine on the activity of human pregnane X receptor and constitutive androstane receptor. J Pharmacol Exp Ther 2011; 336:816-26. [PMID: 21131266 DOI: 10.1124/jpet.110.175927] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Conflicting data exist as to whether meclizine is an activator of human pregnane X receptor (hPXR). Therefore, we conducted a detailed, systematic investigation to determine whether meclizine affects hPXR activity by performing a cell-based reporter gene assay, a time-resolved fluorescence resonance energy transfer competitive ligand-binding assay, a mammalian two-hybrid assay to assess coactivator recruitment, and a hPXR target gene expression assay. In pregnane X receptor (PXR)-transfected HepG2 cells, meclizine activated hPXR to a greater extent than rat PXR. It bound to hPXR ligand-binding domain and recruited steroid receptor coactivator-1 to the receptor. Consistent with its hPXR agonism, meclizine increased hPXR target gene expression (CYP3A4) in human hepatocytes. However, it did not increase but decreased testosterone 6β-hydroxylation, suggesting inhibition of CYP3A catalytic activity. Meclizine has also been reported to be an inverse agonist and antagonist of human constitutive androstane receptor (hCAR). Therefore, given that certain tissues (e.g., liver) express both hPXR and hCAR and that various genes are cross-regulated by them, we quantified the expression of a hCAR- and hPXR-regulated gene (CYP2B6) in cultured human hepatocytes treated with meclizine. This drug did not decrease constitutive CYP2B6 mRNA expression or attenuate hCAR agonist-mediated increase in CYP2B6 mRNA and CYP2B6-catalyzed bupropion hydroxylation levels. These observations reflect hPXR agonism and the lack of hCAR inverse agonism and antagonism by meclizine, which were assessed by a hCAR reporter gene assay and mammalian two-hybrid assay. In conclusion, meclizine is a hPXR agonist, and it does not act as a hCAR inverse agonist or antagonist in cultured human hepatocytes.
Collapse
Affiliation(s)
- Aik Jiang Lau
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
36
|
Shukla SJ, Sakamuru S, Huang R, Moeller TA, Shinn P, Vanleer D, Auld DS, Austin CP, Xia M. Identification of clinically used drugs that activate pregnane X receptors. Drug Metab Dispos 2011; 39:151-9. [PMID: 20966043 PMCID: PMC3014269 DOI: 10.1124/dmd.110.035105] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 10/21/2010] [Indexed: 11/22/2022] Open
Abstract
The pregnane X receptor (PXR) binds xenobiotics and regulates the expression of several drug-metabolizing enzymes and transporters. Human PXR (hPXR) activation and CYP3A4 induction can be involved in drug-drug interactions, resulting in reduced efficacy or increased toxicity. However, there are known species-specific differences with regard to PXR activation that should be taken into account when animal PXR data are extrapolated to humans. We profiled 2816 clinically used drugs from the National Institutes of Health Chemical Genomics Center Pharmaceutical Collection for their ability to activate hPXR and rat PXR (rPXR) at the cellular level, induce human CYP3A4 at the cellular level, and bind human PXR at the protein level. From 6 to 11% of drugs were identified as active across the four assays, which included assay-specific and pan-active compounds. The lowest concordance was observed between the hPXR and rPXR assays, and many compounds active in both assays nonetheless demonstrated significant potency differences between species. Analysis based on clustering potency values demonstrated the greatest activity correlation between the hPXR activation and CYP3A4 induction assays. Structure-activity relationship analysis identified chemical scaffolds that were pan-active (e.g., dihydropyridine calcium channel blockers) and others that were uniquely active in individual assays (e.g., steroids and fatty acids). These results provide important information on PXR activation by clinically used drugs, highlight the species specificity of PXR activation by xenobiotics, and provide a means of prioritizing compounds for follow-up studies and optimization efforts.
Collapse
Affiliation(s)
- Sunita J Shukla
- National Institutes of Health Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lau AJ, Yang G, Rajaraman G, Baucom CC, Chang TKH. Human pregnane X receptor agonism by Ginkgo biloba extract: assessment of the role of individual ginkgolides. J Pharmacol Exp Ther 2010; 335:771-80. [PMID: 20739453 DOI: 10.1124/jpet.110.172338] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ginkgo biloba extract activates pregnane X receptor (PXR), but how this occurs is not known. Therefore, we investigated the mechanism of PXR activation by the extract and the role of five individual terpene trilactones in the activation. In a cell-based reporter gene assay, G. biloba extract activated human PXR (hPXR), and at a concentration present in the extract, ginkgolide A, but not ginkgolide B, ginkgolide C, ginkgolide J, or bilobalide was partially responsible for the increase in hPXR activity of the extract. Likewise, in cultured human hepatocytes, only ginkgolide A contributed to the increase in hPXR target gene expression (CYP3A4 mRNA and CYP3A-mediated testosterone 6β-hydroxylation). The extract, but none of the terpene trilactones, bound to hPXR ligand-binding domain, as analyzed by a time-resolved fluorescence resonance energy transfer competitive binding assay. Only the extract and ginkgolide A recruited steroid receptor coactivator-1, as determined by a mammalian two-hybrid assay. Compared with hPXR, rat PXR (rPXR) was activated to a lesser extent by G. biloba extract. Similar to hPXR, only ginkgolide A contributed to rPXR activation by the extract. In contrast to the effect of G. biloba extract on PXR function, it did not affect hPXR expression. Overall, the main conclusions are that G. biloba extract is an hPXR agonist, and among the five terpene trilactones investigated, only ginkgolide A contributes to the actions of the extract. Our findings provide insights into the biological and chemical mechanisms of hPXR activation by G. biloba extract.
Collapse
Affiliation(s)
- Aik Jiang Lau
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
38
|
Cheng J, Shah YM, Ma X, Pang X, Tanaka T, Kodama T, Krausz KW, Gonzalez FJ. Therapeutic role of rifaximin in inflammatory bowel disease: clinical implication of human pregnane X receptor activation. J Pharmacol Exp Ther 2010; 335:32-41. [PMID: 20627999 PMCID: PMC2957776 DOI: 10.1124/jpet.110.170225] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 07/08/2010] [Indexed: 12/13/2022] Open
Abstract
Human pregnane X receptor (PXR) has been implicated in the pathogenesis of inflammatory bowel disease (IBD). Rifaximin, a human PXR activator, is in clinical trials for treatment of IBD and has demonstrated efficacy in Crohn's disease and active ulcerative colitis. In the current study, the protective and therapeutic role of rifaximin in IBD and its respective mechanism were investigated. PXR-humanized (hPXR), wild-type, and Pxr-null mice were treated with rifaximin in the dextran sulfate sodium (DSS)-induced and trinitrobenzene sulfonic acid (TNBS)-induced IBD models to determine the protective function of human PXR activation in IBD. The therapeutic role of rifaximin was further evaluated in DSS-treated hPXR and Pxr-null mice. Results demonstrated that preadministration of rifaximin ameliorated the clinical hallmarks of colitis in DSS- and TNBS-treated hPXR mice as determined by body weight loss and assessment of diarrhea, rectal bleeding, colon length, and histology. In addition, higher survival rates and recovery from colitis symptoms were observed in hPXR mice, but not in Pxr-null mice, when rifaximin was administered after the onset of symptoms. Nuclear factor κB (NF-κB) target genes were markedly down-regulated in hPXR mice by rifaximin treatment. In vitro NF-κB reporter assays demonstrated inhibition of NF-κB activity after rifaximin treatment in colon-derived cell lines expressing hPXR. These findings demonstrated the preventive and therapeutic role of rifaximin on IBD through human PXR-mediated inhibition of the NF-κB signaling cascade, thus suggesting that human PXR may be an effective target for the treatment of IBD.
Collapse
Affiliation(s)
- Jie Cheng
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Evaluation of an in vitro toxicogenetic mouse model for hepatotoxicity. Toxicol Appl Pharmacol 2010; 249:208-16. [PMID: 20869979 DOI: 10.1016/j.taap.2010.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 09/03/2010] [Accepted: 09/16/2010] [Indexed: 01/15/2023]
Abstract
Numerous studies support the fact that a genetically diverse mouse population may be useful as an animal model to understand and predict toxicity in humans. We hypothesized that cultures of hepatocytes obtained from a large panel of inbred mouse strains can produce data indicative of inter-individual differences in in vivo responses to hepato-toxicants. In order to test this hypothesis and establish whether in vitro studies using cultured hepatocytes from genetically distinct mouse strains are feasible, we aimed to determine whether viable cells may be isolated from different mouse inbred strains, evaluate the reproducibility of cell yield, viability and functionality over subsequent isolations, and assess the utility of the model for toxicity screening. Hepatocytes were isolated from 15 strains of mice (A/J, B6C3F1, BALB/cJ, C3H/HeJ, C57BL/6J, CAST/EiJ, DBA/2J, FVB/NJ, BALB/cByJ, AKR/J, MRL/MpJ, NOD/LtJ, NZW/LacJ, PWD/PhJ and WSB/EiJ males) and cultured for up to 7 days in traditional 2-dimensional culture. Cells from B6C3F1, C57BL/6J, and NOD/LtJ strains were treated with acetaminophen, WY-14,643 or rifampin and concentration-response effects on viability and function were established. Our data suggest that high yield and viability can be achieved across a panel of strains. Cell function and expression of key liver-specific genes of hepatocytes isolated from different strains and cultured under standardized conditions are comparable. Strain-specific responses to toxicant exposure have been observed in cultured hepatocytes and these experiments open new opportunities for further developments of in vitro models of hepatotoxicity in a genetically diverse population.
Collapse
|
40
|
Fery Y, Mueller SO, Schrenk D. Development of stably transfected human and rat hepatoma cell lines for the species-specific assessment of xenobiotic response enhancer module (XREM)-dependent induction of drug metabolism. Toxicology 2010; 277:11-9. [PMID: 20832446 DOI: 10.1016/j.tox.2010.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 08/11/2010] [Accepted: 08/13/2010] [Indexed: 11/15/2022]
Abstract
Based on our current knowledge, PXR holds a key position in the induction of a selective battery of enzymes and transporters of drug metabolism. In order to prevent serious adverse drug effects or unpredicted drug-drug interactions (DDI), it is compulsory to investigate the possible inducing potency of drugs under development. Furthermore, analysis of the inducing potency of environmental pollutants and new or manufactured chemicals is part of toxicological risk assessment. In non-transfected human HepG2 and rat H4IIE hepatoma cells, we examined the characteristics of expression of 45 genes involved in drug metabolism. A few gene products such as CYP2B6 or CYP3A4 mRNA were prominent in HepG2 cells while their major rat counterparts were, e.g., CYP2B3 or CYP3A1/3A3. Furthermore, a number of xenobiotic receptors including PXR were expressed in both cell lines. A number of genes were regulated in a cell type and species-specific manner after incubation with the prototypical PXR agonists rifampicin or dexamethasone, respectively. Then, we established cell-based reporter gene assays for screening for PXR-dependent induction of drug metabolism. HepG2 and H4IIE cells were stably transfected with a reporter gene containing PXR responsive elements (XREMs) which mediate the induction of PXR target genes such as CYP3A enzymes. With both stable cell lines the CYP inducers clotrimazole, dexamethasone, omeprazole, phenobarbital, rifampicin, as well as the drug candidate EMD 392949 and the brominated flame retardants hexabromocylododecane (HBCD) and a pentabromodiphenyl ether (pentaBDE) mixture were screened. In the human HepG2-XREM3 and rat H4IIE-XREM3 cells, clotrimazole and HBCD were found as common activators of the human and rat PXR whereas pentaBDE was more effective with the human cell system. Omeprazole and phenobarbital did not induce the rat PXR-dependent reporter gene expression in H4IIE-XREM3 cells, while a moderate increase was found in HepG2-XREM3 cells. EMD 392949 also acted as inducer in human but not in rat cells confirming in vivo observations. In summary, the established PXR-dependent in vitro system allows the simultaneous, fast, and species-specific screening of chemicals, environmental contaminants, food ingredients and drugs for CYP3A induction in cells of human and rat origin.
Collapse
Affiliation(s)
- Yvonne Fery
- Food Chemistry and Toxicology, University of Kaiserslautern, Erwin-Schroedinger-Strasse 52, D-67663 Kaiserslautern, Germany
| | | | | |
Collapse
|
41
|
Hartz AMS, Miller DS, Bauer B. Restoring blood-brain barrier P-glycoprotein reduces brain amyloid-beta in a mouse model of Alzheimer's disease. Mol Pharmacol 2010; 77:715-23. [PMID: 20101004 PMCID: PMC2872973 DOI: 10.1124/mol.109.061754] [Citation(s) in RCA: 255] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 01/25/2010] [Indexed: 12/15/2022] Open
Abstract
Reduced clearance of amyloid-beta (Abeta) from brain partly underlies increased Abeta brain accumulation in Alzheimer's disease (AD). The mechanistic basis for this pathology is unknown, but recent evidence suggests a neurovascular component in AD etiology. We show here that the ATP-driven pump, P-glycoprotein, specifically mediates efflux transport of Abeta from mouse brain capillaries into the vascular space, thus identifying a critical component of the Abeta brain efflux mechanism. We demonstrate in a transgenic mouse model of AD [human amyloid precursor protein (hAPP)-overexpressing mice; Tg2576 strain] that brain capillary P-glycoprotein expression and transport activity are substantially reduced compared with wild-type control mice, suggesting a mechanism by which Abeta accumulates in the brain in AD. It is noteworthy that dosing 12-week-old, asymptomatic hAPP mice over 7 days with pregnenolone-16alpha-carbonitrile to activate the nuclear receptor pregnane X receptor restores P-glycoprotein expression and transport activity in brain capillaries and significantly reduces brain Abeta levels compared with untreated control mice. Thus, targeting intracellular signals that up-regulate blood-brain barrier P-glycoprotein in the early stages of AD has the potential to increase Abeta clearance from the brain and reduce Abeta brain accumulation. This mechanism suggests a new therapeutic strategy in AD.
Collapse
Affiliation(s)
- Anika M S Hartz
- Department of Biochemistry and Molecular Biology, University of Minnesota Medical School, Duluth, MN 55812, USA
| | | | | |
Collapse
|
42
|
Paitz RT, Bowden RM. Progesterone metabolites, "xenobiotic-sensing" nuclear receptors, and the metabolism of maternal steroids. Gen Comp Endocrinol 2010; 166:217-21. [PMID: 19932108 DOI: 10.1016/j.ygcen.2009.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 11/04/2009] [Accepted: 11/12/2009] [Indexed: 12/22/2022]
Abstract
During development, embryos utilize steroid signals to direct sexual differentiation of tissues necessary for reproduction. Disruption of these signals by exogenous substances (both natural and synthetic) frequently produce phenotypic effects that can persist into adulthood and influence reproduction. This paper reviews the evidence that during embryonic development, progesterone metabolites and xenobiotic-sensing nuclear receptors may interact to increase the expression of numerous enzymes responsible for steroid metabolism in oviparous and placental amniotes. In these groups, embryonic development is characterized by (1) elevated progesterone concentrations, (2) 5 beta reduction being the primary metabolic pathway of progesterone, (3) the presence of xenobiotic-sensing nuclear receptors that can bind 5 beta metabolites of progesterone, and (4) increased expression of a suite of enzymes responsible for the metabolism of multiple steroids. We propose that xenobiotic-sensing nuclear receptors initially evolved to buffer the developing embryo from the potentially adverse effects of various maternal steroids on sexual differentiation.
Collapse
Affiliation(s)
- Ryan T Paitz
- School of Biological Sciences, Campus Box 4120, Illinois State University, Normal, IL 61790-4120, USA.
| | | |
Collapse
|
43
|
Weiss J, Haefeli WE. Impact of ATP-binding cassette transporters on human immunodeficiency virus therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 280:219-79. [PMID: 20797684 DOI: 10.1016/s1937-6448(10)80005-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Even though potent antiretrovirals are available against human immunodeficiency virus (HIV)-1 infection, therapy fails in a significant fraction of patients. Among the most relevant reasons for treatment failure are drug toxicity and side effects, but also the development of viral resistance towards the drugs applied. Efflux by ATP-binding cassette (ABC-) transporters represents one major mechanism influencing the pharmacokinetics of antiretroviral drugs and particularly their distribution, thus modifiying the concentration within the infected cells, that is, at the site of action. Moreover, drug-drug interactions may occur at the level of these transporters and modulate their activity or expression thus influencing the efficacy and toxicity of the substrate drugs. This review summarizes current knowledge on the interaction of antiretrovirals used for HIV-1 therapy with ABC-transporters and highlights the impact of ABC-transporters for cellular resistance and therapeutic success. Moreover, the suitability of different cell models for studying the interaction of antiretrovirals with ABC-transporters is discussed.
Collapse
Affiliation(s)
- Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
44
|
Abstract
Due to their well-known differentiation and apoptosis-inducing abilities, retinoic acid (RA) and its analogs have strong anti-cancer efficacy in human cancers. However, in vivo RA is a liver mitogen. While speculation has persisted that RA-mediated signaling is likely involved in hepatocyte proliferation during liver regeneration, direct evidence is still required. Findings in support of this proposition include observations that a release of retinyl palmitate (the precursor of RA) occurs in liver stellate cells following liver injury. Nevertheless, the biological action of this released vitamin A is virtually unknown. More likely is that the released vitamin A is converted to RA, the biological form, and then bound to a specific receptor (retinoid x receptor; RXRα), which is most abundantly expressed in the liver. Considering the mitogenic effects of RA, the RA-activated RXRα would likely then influence hepatocyte proliferation and liver tissue repair. At present, the mechanism by which RA stimulates hepatocyte proliferation is largely unknown. This review summarizes the activation of nuclear receptors (peroxisome proliferator activated receptor-α, pregnane x receptor, constitutive androstane receptor, and farnesoid x receptor) in an RXRα dependent manner to induce hepatocyte proliferation, providing a link between RA and its proliferative role.
Collapse
|
45
|
Yoshizato K, Tateno C. A human hepatocyte-bearing mouse: an animal model to predict drug metabolism and effectiveness in humans. PPAR Res 2009; 2009:476217. [PMID: 19884982 PMCID: PMC2768028 DOI: 10.1155/2009/476217] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 07/13/2009] [Indexed: 01/16/2023] Open
Abstract
Preclinical studies to predict the efficacy and safety of drugs have conventionally been conducted almost exclusively in mice and rats as rodents, despite the differences in drug metabolism between humans and rodents. Furthermore, human (h) viruses such as hepatitis viruses do not infect the rodent liver. A mouse bearing a liver in which the hepatocytes have been largely repopulated with h-hepatocytes would overcome some of these disadvantages. We have established a practical, efficient, and large-scale production system for such mice. Accumulated evidence has demonstrated that these hepatocyte-humanized mice are a useful and reliable animal model, exhibiting h-type responses in a series of in vivo drug processing (adsorption, distribution, metabolism, excretion) experiments and in the infection and propagation of hepatic viruses. In this review, we present the current status of studies on chimeric mice and describe their usefulness in the study of peroxisome proliferator-activated receptors.
Collapse
|
46
|
Chang TKH. Activation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) by herbal medicines. AAPS JOURNAL 2009; 11:590-601. [PMID: 19688601 DOI: 10.1208/s12248-009-9135-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 07/30/2009] [Indexed: 12/14/2022]
Abstract
Pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are transcription factors that control the expression of a broad array of genes involved not only in transcellular transport and biotransformation of many drugs, other xenochemicals, and endogenous substances, such as bile acid, bilirubin, and certain vitamins, but also in various physiological/pathophysiological processes such as lipid metabolism, glucose homeostasis, and inflammation. Ligands of PXR and CAR are chemicals of diverse structures, including naturally occurring compounds present in herbal medicines. The overall aim of this article is to provide an overview of our current understanding of the role of herbal medicines as modulators of PXR and CAR.
Collapse
Affiliation(s)
- Thomas K H Chang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
47
|
Cheng J, Ma X, Krausz KW, Idle JR, Gonzalez FJ. Rifampicin-activated human pregnane X receptor and CYP3A4 induction enhance acetaminophen-induced toxicity. Drug Metab Dispos 2009; 37:1611-21. [PMID: 19460945 PMCID: PMC2712435 DOI: 10.1124/dmd.109.027565] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 05/21/2009] [Indexed: 11/22/2022] Open
Abstract
Acetaminophen (APAP) is safe at therapeutic levels but causes hepatotoxicity via N-acetyl-p-benzoquinone imine-induced oxidative stress upon overdose. To determine the effect of human (h) pregnane X receptor (PXR) activation and CYP3A4 induction on APAP-induced hepatotoxicity, mice humanized for PXR and CYP3A4 (TgCYP3A4/hPXR) were treated with APAP and rifampicin. Human PXR activation and CYP3A4 induction enhanced APAP-induced hepatotoxicity as revealed by hepatic alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities elevated in serum, and hepatic necrosis after coadministration of rifampicin and APAP, compared with APAP administration alone. In contrast, hPXR mice, wild-type mice, and Pxr-null mice exhibited significantly lower ALT/AST levels compared with TgCYP3A4/hPXR mice after APAP administration. Toxicity was coincident with depletion of hepatic glutathione and increased production of hydrogen peroxide, suggesting increased oxidative stress upon hPXR activation. Moreover, mRNA analysis demonstrated that CYP3A4 and other PXR target genes were significantly induced by rifampicin treatment. Urinary metabolomic analysis indicated that cysteine-APAP and its metabolite S-(5-acetylamino-2-hydroxyphenyl)mercaptopyruvic acid were the major contributors to the toxic phenotype. Quantification of plasma APAP metabolites indicated that the APAP dimer formed coincident with increased oxidative stress. In addition, serum metabolomics revealed reduction of lysophosphatidylcholine in the APAP-treated groups. These findings demonstrated that human PXR is involved in regulation of APAP-induced toxicity through CYP3A4-mediated hepatic metabolism of APAP in the presence of PXR ligands.
Collapse
Affiliation(s)
- Jie Cheng
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
48
|
Fery Y, Buschauer I, Salzig C, Lang P, Schrenk D. Technical pentabromodiphenyl ether and hexabromocyclododecane as activators of the pregnane-X-receptor (PXR). Toxicology 2009; 264:45-51. [PMID: 19631710 DOI: 10.1016/j.tox.2009.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 07/06/2009] [Accepted: 07/16/2009] [Indexed: 10/20/2022]
Abstract
Technical pentabrominated diphenyl ether (pentaBDE mix) is a mixture of polybrominated diphenyl ethers (PBDEs) which has been widely used as a flame retardant. Since its ban in several countries it has been replaced by other brominated flame retardants such as hexabromocyclododecane (HBCD). Both certain PBDE congeners and HBCD are present in environmental and human samples reflecting their persistent and bioaccumulative properties. PentaBDE mix and HBCD have recently been found to induce cytochrome P450 (CYP) 3 enzymes in rat liver. In this study we tested both technical pentaBDE mix and HBCD for their potency to induce CYP3A enzymes in rat hepatocytes in primary culture, and in rat H4IIE and human HepG2 hepatoma cells. In rat hepatocytes, HBCD was a more effective CYP3A1 inducer than pentaBDE mix, being less effective, however, than the prototype inducer dexamethasone. In human HepG2 cells, both compounds and the prototype inducer rifampicin were about equally effective. In contrast, in HepG2 cells, HBCD failed to induce luciferin-PFBE dealkylase, a common catalytic activity of a number of CYP3A enzymes, possibly reflecting enzyme inhibition. A significant induction of catalytic activity was observed in rat hepatocytes with both compounds. Analysis of a XREM-driven reporter gene activity in transfected cells confirmed that both compounds act as agonists of the human and rat pregnane-X-receptor, which was detectable in all cell types used.
Collapse
Affiliation(s)
- Yvonne Fery
- Food Chemistry and Toxicology, University of Kaiserslautern, Erwin-Schroedinger-Strasse 52, D-67663 Kaiserslautern, Germany
| | | | | | | | | |
Collapse
|
49
|
Lin YS, Yasuda K, Assem M, Cline C, Barber J, Li CW, Kholodovych V, Ai N, Chen JD, Welsh WJ, Ekins S, Schuetz EG. The major human pregnane X receptor (PXR) splice variant, PXR.2, exhibits significantly diminished ligand-activated transcriptional regulation. Drug Metab Dispos 2009; 37:1295-304. [PMID: 19251824 PMCID: PMC2683691 DOI: 10.1124/dmd.108.025213] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Accepted: 02/23/2009] [Indexed: 02/03/2023] Open
Abstract
The pregnane X receptor (PXR; PXR.1) can be activated by structurally diverse lipophilic ligands. PXR.2, an alternatively spliced form of PXR, lacks 111 nucleotides encoding 37 amino acids in the ligand binding domain. PXR.2 bound a classic CYP3A4 PXR response element (PXRE) in electrophoretic mobility shift assays, but transfected PXR.2 failed to transactivate a CYP3A4-promoter-luciferase reporter plasmid in HepG2 cells treated with various PXR ligands. Cotransfection experiments showed that PXR.2 behaved as a dominant negative, interfering with PXR.1/rifampin activation of CYP3A4-PXRE-LUC. In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. In contrast, PXR.2 stably introduced into the same host cells failed to induce target genes over levels in mock-transfected cells after drug treatment. Our homology modeling suggests that ligands bind PXR.1 more favorably, probably because of the presence of a key disordered loop region, which is missing in PXR.2. Yeast two-hybrid assays revealed that, even in the presence of ligand, the corepressors remain tightly bound to PXR.2, and coactivators are unable to bind at helix 12. In summary, PXR.2 can bind to PXREs but fails to transactivate target genes because ligands do not bind the ligand binding domain of PXR.2 productively, corepressors remain tightly bound, and coactivators are not recruited to PXR.2.
Collapse
Affiliation(s)
- Yvonne S Lin
- Department of Pharmaceutical Sciences, St. Jude Children's Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
di Masi A, De Marinis E, Ascenzi P, Marino M. Nuclear receptors CAR and PXR: Molecular, functional, and biomedical aspects. Mol Aspects Med 2009; 30:297-343. [PMID: 19427329 DOI: 10.1016/j.mam.2009.04.002] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 04/28/2009] [Indexed: 12/31/2022]
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors sharing a common evolutionary history and having similar sequence features at the protein level. Selective ligand(s) for some NRs is not known, therefore these NRs have been named "orphan receptors". Whenever ligands have been recognized for any of the orphan receptor, it has been categorized and grouped as "adopted" orphan receptor. This group includes the constitutive androstane receptor (CAR) and the pregnane X receptor (PXR). They function as sensors of toxic byproducts derived from endogenous metabolites and of exogenous chemicals, in order to enhance their elimination. This unique function of CAR and PXR sets them apart from the steroid hormone receptors. The broad response profile has established that CAR and PXR are xenobiotic sensors that coordinately regulate xenobiotic clearance in the liver and intestine via induction of genes involved in drug and xenobiotic metabolism. In the past few years, research has revealed new and mostly unsuspected roles for CAR and PXR in modulating hormone, lipid, and energy homeostasis as well as cancer and liver steatosis. The purpose of this review is to highlight the structural and molecular bases of CAR and PXR impact on human health, providing information on mechanisms through which diet, chemical exposure, and environment ultimately impact health and disease.
Collapse
Affiliation(s)
- Alessandra di Masi
- Department of Biology, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | | | | | | |
Collapse
|