1
|
Monrose M, Holota H, Martinez G, Damon-Soubeyrand C, Thirouard L, Martinot E, Battistelli E, de Haze A, Bravard S, Tamisier C, Caira F, Coutton C, Barbotin AL, Boursier A, Lakhal L, Beaudoin C, Volle DH. Constitutive Androstane Receptor Regulates Germ Cell Homeostasis, Sperm Quality, and Male Fertility via Akt-Foxo1 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402082. [PMID: 39318179 DOI: 10.1002/advs.202402082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/02/2024] [Indexed: 09/26/2024]
Abstract
Male sexual function can be disrupted by exposure to exogenous compounds that cause testicular physiological alterations. The constitutive androstane receptor (Car) is a receptor for both endobiotics and xenobiotics involved in detoxification. However, its role in male fertility, particularly in regard to the reprotoxic effects of environmental pollutants, remains unclear. This study aims to investigate the role of the Car signaling pathway in male fertility. In vivo, in vitro, and pharmacological approaches are utilized in wild-type and Car-deficient mouse models. The results indicate that Car inhibition impaired male fertility due to altered sperm quality, specifically histone retention, which is correlated with an increased percentage of dying offspring in utero. The data highlighted interactions among Car, Akt, Foxo1, and histone acetylation. This study demonstrates that Car is crucial in germ cell homeostasis and male fertility. Further research on the Car signaling pathway is necessary to reveal unidentified causes of altered fertility and understand the harmful impact of environmental molecules on male fertility and offspring health.
Collapse
Affiliation(s)
- Mélusine Monrose
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Hélène Holota
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Guillaume Martinez
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, F-38000, France
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Grenoble, F-38000, France
| | - Christelle Damon-Soubeyrand
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Plateform Anipath, Clermont-Ferrand, F-63001, France
| | - Laura Thirouard
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Emmanuelle Martinot
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Edwige Battistelli
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Angélique de Haze
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Stéphanie Bravard
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Plateform Anipath, Clermont-Ferrand, F-63001, France
| | - Christelle Tamisier
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Françoise Caira
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Charles Coutton
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, F-38000, France
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Grenoble, F-38000, France
| | - Anne-Laure Barbotin
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, F-59000, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, F-59000, France
| | - Angèle Boursier
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, F-59000, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, F-59000, France
| | - Laila Lakhal
- INRAe UMR1331, ToxAlim, University of Toulouse, Toulouse, F-31027, France
| | - Claude Beaudoin
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - David H Volle
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| |
Collapse
|
2
|
Zhao P, Fan S, Zhou Y, Huang M, Gao Y, Bi H. Constitutive Androstane Receptor and Peroxisome Proliferator-Activated Receptor α Do Not Perform Liquid-Liquid Phase Separation in Cells. J Pharmacol Exp Ther 2024; 390:88-98. [PMID: 38719477 DOI: 10.1124/jpet.124.002174] [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: 02/08/2024] [Accepted: 04/23/2024] [Indexed: 06/23/2024] Open
Abstract
Constitutive androstane receptor (CAR) and peroxisome proliferator-activated receptor α (PPARα) are members of the nuclear receptor superfamily, which regulates various physiologic and pathologic processes. Phase separation is a dynamic biophysical process in which biomacromolecules form liquid-like condensates, which have been identified as contributors to many cellular functions, such as signal transduction and transcription regulation. However, the possibility of phase separation for CAR and PPARα remains unknown. This study explored the potential phase separation of CAR and PPARα The computational analysis utilizing algorithm tools examining the intrinsically disordered regions of CAR and PPARα suggested a limited likelihood of undergoing phase separation. Experimental assays under varying conditions of hyperosmotic stress and agonist treatments confirmed the absence of phase separation for these receptors. Additionally, the optoDroplets assay, which utilizes blue light stimulation to induce condensate formation, showed that there was no condensate formation of the fusion protein of Cry2 with CAR or PPARα Furthermore, phase separation of CAR or PPARα did not occur despite reduced target expression under hyperosmotic stress. In conclusion, these findings revealed that neither the activation of CAR and PPARα nor hyperosmotic stress induces phase separation of CAR and PPARα in cells. SIGNIFICANCE STATEMENT: Constitutive androstane receptor (CAR) and peroxisome proliferator-activated receptor α (PPARα) are key regulators of various functions in the body. This study showed that CAR and PPARα do not exhibit phase separation under hyperosmotic stress or after agonist-induced activation. These findings provide new insights into the CAR and PPARα biology and physiology.
Collapse
Affiliation(s)
- Pengfei Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China (P.Z., S.F., H.B.); Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (P.Z., S.F., Y.Z., M.H., Y.G., H.B.); and The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China (H.B.)
| | - Shicheng Fan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China (P.Z., S.F., H.B.); Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (P.Z., S.F., Y.Z., M.H., Y.G., H.B.); and The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China (H.B.)
| | - Yanying Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China (P.Z., S.F., H.B.); Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (P.Z., S.F., Y.Z., M.H., Y.G., H.B.); and The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China (H.B.)
| | - Min Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China (P.Z., S.F., H.B.); Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (P.Z., S.F., Y.Z., M.H., Y.G., H.B.); and The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China (H.B.)
| | - Yue Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China (P.Z., S.F., H.B.); Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (P.Z., S.F., Y.Z., M.H., Y.G., H.B.); and The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China (H.B.)
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong Provincial Key Laboratory of New Drug Screening and Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China (P.Z., S.F., H.B.); Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (P.Z., S.F., Y.Z., M.H., Y.G., H.B.); and The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China (H.B.)
| |
Collapse
|
3
|
Liu J, Malekoltojari A, Asokakumar A, Chow V, Li L, Li H, Grimaldi M, Dang N, Campbell J, Barrett H, Sun J, Navarre W, Wilson D, Wang H, Mani S, Balaguer P, Anakk S, Peng H, Krause HM. Diindoles produced from commensal microbiota metabolites function as endogenous CAR/Nr1i3 ligands. Nat Commun 2024; 15:2563. [PMID: 38519460 PMCID: PMC10960024 DOI: 10.1038/s41467-024-46559-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 02/27/2024] [Indexed: 03/25/2024] Open
Abstract
Numerous studies have demonstrated the correlation between human gut bacteria and host physiology, mediated primarily via nuclear receptors (NRs). Despite this body of work, the systematic identification and characterization of microbe-derived ligands that regulate NRs remain a considerable challenge. In this study, we discover a series of diindole molecules produced from commensal bacteria metabolites that act as specific agonists for the orphan constitutive androstane receptor (CAR). Using various biophysical analyses we show that their nanomolar affinities are comparable to those of synthetic CAR agonists, and that they can activate both rodent and human CAR orthologues, which established synthetic agonists cannot. We also find that the diindoles, diindolylmethane (DIM) and diindolylethane (DIE) selectively up-regulate bona fide CAR target genes in primary human hepatocytes and mouse liver without causing significant side effects. These findings provide new insights into the complex interplay between the gut microbiome and host physiology, as well as new tools for disease treatment.
Collapse
Affiliation(s)
- Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Ainaz Malekoltojari
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anjana Asokakumar
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Vimanda Chow
- Department of Chemistry, York University, Toronto, ON, M3J 1P3, Canada
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Hao Li
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine; Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, Inserm, U1194, France
| | - Nathanlown Dang
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jhenielle Campbell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Holly Barrett
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Jianxian Sun
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- School of the Environment, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - William Navarre
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Derek Wilson
- Department of Chemistry, York University, Toronto, ON, M3J 1P3, Canada
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Sridhar Mani
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine; Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, Inserm, U1194, France
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada.
- School of the Environment, University of Toronto, Toronto, ON, M5S 3H6, Canada.
| | - Henry M Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Dauwe Y, Mary L, Oliviero F, Grimaldi M, Balaguer P, Gayrard V, Mselli-Lakhal L. Steatosis and Metabolic Disorders Associated with Synergistic Activation of the CAR/RXR Heterodimer by Pesticides. Cells 2023; 12:cells12081201. [PMID: 37190111 DOI: 10.3390/cells12081201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
The nuclear receptor, constitutive androstane receptor (CAR), which forms a heterodimer with the retinoid X receptor (RXR), was initially reported as a transcription factor that regulates hepatic genes involved in detoxication and energy metabolism. Different studies have shown that CAR activation results in metabolic disorders, including non-alcoholic fatty liver disease, by activating lipogenesis in the liver. Our objective was to determine whether synergistic activations of the CAR/RXR heterodimer could occur in vivo as described in vitro by other authors, and to assess the metabolic consequences. For this purpose, six pesticides, ligands of CAR, were selected, and Tri-butyl-tin (TBT) was used as an RXR agonist. In mice, CAR's synergic activation was induced by dieldrin associated with TBT, and combined effects were induced by propiconazole, bifenox, boscalid, and bupirimate. Moreover, a steatosis, characterized by increased triglycerides, was observed when TBT was combined with dieldrin, propiconazole, bifenox, boscalid, and bupirimate. Metabolic disruption appeared in the form of increased cholesterol and lowered free fatty acid plasma levels. An in-depth analysis revealed increased expression of genes involved in lipid synthesis and lipid import. These results contribute to the growing understanding of how environmental contaminants can influence nuclear receptor activity and associated health risks.
Collapse
Affiliation(s)
- Yannick Dauwe
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Lucile Mary
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Fabiana Oliviero
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194-Université Montpellier-Institut régional du Cancer Montpellier, CEDEX 5, F-34298 Montpellier, France
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194-Université Montpellier-Institut régional du Cancer Montpellier, CEDEX 5, F-34298 Montpellier, France
| | - Véronique Gayrard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Laïla Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| |
Collapse
|
5
|
Zhang J, Jia Q, Li Y, He J. The Function of Xenobiotic Receptors in Metabolic Diseases. Drug Metab Dispos 2023; 51:237-248. [PMID: 36414407 DOI: 10.1124/dmd.122.000862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 09/01/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolic diseases are a series of metabolic disorders that include obesity, diabetes, insulin resistance, hypertension, and hyperlipidemia. The increased prevalence of metabolic diseases has resulted in higher mortality and mobility rates over the past decades, and this has led to extensive research focusing on the underlying mechanisms. Xenobiotic receptors (XRs) are a series of xenobiotic-sensing nuclear receptors that regulate their downstream target genes expression, thus defending the body from xenobiotic and endotoxin attacks. XR activation is associated with the development of a number of metabolic diseases such as obesity, nonalcoholic fatty liver disease, type 2 diabetes, and cardiovascular diseases, thus suggesting an important role for XRs in modulating metabolic diseases. However, the regulatory mechanism of XRs in the context of metabolic disorders under different nutrient conditions is complex and remains controversial. This review summarizes the effects of XRs on different metabolic components (cholesterol, lipids, glucose, and bile acids) in different tissues during metabolic diseases. As chronic inflammation plays a critical role in the initiation and progression of metabolic diseases, we also discuss the impact of XRs on inflammation to comprehensively recognize the role of XRs in metabolic diseases. This will provide new ideas for treating metabolic diseases by targeting XRs. SIGNIFICANCE STATEMENT: This review outlines the current understanding of xenobiotic receptors on nutrient metabolism and inflammation during metabolic diseases. This work also highlights the gaps in this field, which can be used to direct the future investigations on metabolic diseases treatment by targeting xenobiotic receptors.
Collapse
Affiliation(s)
- Jinhang Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingyi Jia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Niu Y, Tang S. Circadian clock-mediated nuclear receptors in cancer. J Cell Physiol 2022; 237:4428-4442. [PMID: 36250982 DOI: 10.1002/jcp.30905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/25/2022] [Accepted: 10/03/2022] [Indexed: 11/09/2022]
Abstract
Circadian system coordinates the daily periodicity of physiological and biochemical functions to adapt to environmental changes. Circadian disruption has been identified to increase the risk of cancer and promote cancer progression, but the underlying mechanism remains unclear. And further mechanistic understanding of the crosstalk between clock components and cancer is urgent to achieve clinical anticancer benefits from chronochemotherapy. Recent studies discover that several nuclear receptors regulating circadian clock, also play crucial roles in mediating multiple cancer processes. In this review, we aim to summarize the latest developments of clock-related nuclear receptors in cancer biology and dissect mechanistic insights into how nuclear receptors coordinate with circadian clock to regulate tumorigenesis and cancer treatment. A better understanding of circadian clock-related nuclear receptors in cancer could help prevent tumorigenesis and improve anticancer efficacy.
Collapse
Affiliation(s)
- Ya Niu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shuang Tang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| |
Collapse
|
7
|
Effects of rifampicin on hepatic antioxidant enzymes in PXR and CAR double humanized mice. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00134-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
8
|
Mukha A, Kalkhoven E, van Mil SWC. Splice variants of metabolic nuclear receptors: Relevance for metabolic disease and therapeutic targeting. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166183. [PMID: 34058349 DOI: 10.1016/j.bbadis.2021.166183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022]
Abstract
Metabolic nuclear receptors are ligand-activated transcription factors which control a wide range of metabolic processes and signaling pathways in response to nutrients and xenobiotics. Targeting these NRs is at the forefront of our endeavours to generate novel treatment options for diabetes, metabolic syndrome and fatty liver disease. Numerous splice variants have been described for these metabolic receptors. Structural changes, as a result of alternative splicing, lead to functional differences among NR isoforms, resulting in the regulation of different metabolic pathways by these NR splice variants. In this review, we describe known splice variants of FXR, LXRs, PXR, RXR, LRH-1, CAR and PPARs. We discuss their structure and functions, and elaborate on the regulation of splice variant abundance by nutritional signals. We conclude that NR splice variants pose an intriguing new layer of complexity in metabolic signaling, which needs to be taken into account in the development of treatment strategies for metabolic diseases.
Collapse
Affiliation(s)
- Anna Mukha
- Center for Molecular Medicine, UMC Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, UMC Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Saskia W C van Mil
- Center for Molecular Medicine, UMC Utrecht and Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
9
|
Pham B, Arons AB, Vincent JG, Fernandez EJ, Shen T. Regulatory Mechanics of Constitutive Androstane Receptors: Basal and Ligand-Directed Actions. J Chem Inf Model 2019; 59:5174-5182. [PMID: 31714771 DOI: 10.1021/acs.jcim.9b00695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Constitutive androstane receptor (CAR) is a nuclear hormone receptor that primarily functions in sensing and metabolizing xenobiotics. The basal activity of this receptor is relatively high, and CAR is deemed active in the absence of ligand. The (over)activation can promote drug toxicity and tumor growth. Thus, therapeutic treatments seek inverse agonists to inhibit or modulate CAR activities. To advance our understanding of the regulatory mechanisms of CAR, we used computational and experimental approaches to elucidate three aspects of CAR activation and inactivation: (1) ligand-dependent actions, (2) ligand-orthologue specificity, and (3) constitutive activity. For ligand-dependent actions, we examined the ligand-bound simulations and identified two sets of ligand-induced contacts promoting CAR activation via coactivator binding (H11-H12 contact) or inactivation via corepressor binding (H4-H11 contact). For orthologue specificity, we addressed a puzzling fact that murine CAR (mCAR) and human CAR (hCAR) respond differently to the same ligand (CITCO), despite their high sequence homology. We found that the helix H7 of hCAR is responsible for a stronger binding of the ligand CITCO compared to mCAR, hence a stronger CITCO-induced activation. For basal activity, we reported computer-generated unliganded CAR structures and critical mutagenesis (mCAR's V209A and N333D) results of a cell-based transcription assay. Our results reveal that the basal conformation of CAR shares prominent features with the agonist-bound form, and helix HX has an important contribution to the constitutive activity. These findings altogether can be useful for the understanding of constitutively active receptors and the design of drug molecules targeting them.
Collapse
Affiliation(s)
- Bill Pham
- Department of Biochemistry & Cellular and Molecular Biology , University of Tennessee , Knoxville , Tennessee 37996 , United States
| | - Avery Bancroft Arons
- Department of Biochemistry & Cellular and Molecular Biology , University of Tennessee , Knoxville , Tennessee 37996 , United States
| | - Jeremy G Vincent
- Department of Biochemistry & Cellular and Molecular Biology , University of Tennessee , Knoxville , Tennessee 37996 , United States
| | - Elias J Fernandez
- Department of Biochemistry & Cellular and Molecular Biology , University of Tennessee , Knoxville , Tennessee 37996 , United States
| | - Tongye Shen
- Department of Biochemistry & Cellular and Molecular Biology , University of Tennessee , Knoxville , Tennessee 37996 , United States
| |
Collapse
|
10
|
Matsuzaka Y, Uesawa Y. Prediction Model with High-Performance Constitutive Androstane Receptor (CAR) Using DeepSnap-Deep Learning Approach from the Tox21 10K Compound Library. Int J Mol Sci 2019; 20:ijms20194855. [PMID: 31574921 PMCID: PMC6801383 DOI: 10.3390/ijms20194855] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 12/30/2022] Open
Abstract
The constitutive androstane receptor (CAR) plays pivotal roles in drug-induced liver injury through the transcriptional regulation of drug-metabolizing enzymes and transporters. Thus, identifying regulatory factors for CAR activation is important for understanding its mechanisms. Numerous studies conducted previously on CAR activation and its toxicity focused on in vivo or in vitro analyses, which are expensive, time consuming, and require many animals. We developed a computational model that predicts agonists for the CAR using the Toxicology in the 21st Century 10k library. Additionally, we evaluate the prediction performance of novel deep learning (DL)-based quantitative structure-activity relationship analysis called the DeepSnap-DL approach, which is a procedure of generating an omnidirectional snapshot portraying three-dimensional (3D) structures of chemical compounds. The CAR prediction model, which applies a 3D structure generator tool, called CORINA-generated and -optimized chemical structures, in the DeepSnap-DL demonstrated better performance than the existing methods using molecular descriptors. These results indicate that high performance in the prediction model using the DeepSnap-DL approach may be important to prepare suitable 3D chemical structures as input data and to enable the identification of modulators of the CAR.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Tokyo 204-8588, Japan.
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Tokyo 204-8588, Japan.
| |
Collapse
|
11
|
Liang D, Li L, Lynch C, Diethelm-Varela B, Xia M, Xue F, Wang H. DL5050, a Selective Agonist for the Human Constitutive Androstane Receptor. ACS Med Chem Lett 2019; 10:1039-1044. [PMID: 31312405 DOI: 10.1021/acsmedchemlett.9b00079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/12/2019] [Indexed: 11/30/2022] Open
Abstract
The constitutive androstane receptor (CAR) is a xenobiotic sensor governing the transcription of genes involved in drug disposition, energy homeostasis, and cell proliferation. However, currently available human CAR (hCAR) agonists are nonselective, which commonly activate hCAR along with other nuclear receptors, especially the closely related human pregnane X receptor (hPXR). Using a well-known hCAR agonist CITCO as a template, we report our efforts in the discovery of a potent and highly selective hCAR agonist. Two of the new compounds of the series, 18 and 19 (DL5050), demonstrated excellent potency and selectivity for hCAR over hPXR. DL5050 preferentially induced the expression of CYP2B6 (target of hCAR) over CYP3A4 (target of hPXR) on both the mRNA and protein levels. The selective hCAR agonist DL5050 represents a valuable tool molecule to further define the biological functions of hCAR, and may also be used as a new lead in the discovery of hCAR agonists for various therapeutic applications.
Collapse
Affiliation(s)
- Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Caitlin Lynch
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892-3375, United States
| | - Benjamin Diethelm-Varela
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892-3375, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
12
|
Nuclear Receptor Metabolism of Bile Acids and Xenobiotics: A Coordinated Detoxification System with Impact on Health and Diseases. Int J Mol Sci 2018; 19:ijms19113630. [PMID: 30453651 PMCID: PMC6274770 DOI: 10.3390/ijms19113630] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
Structural and functional studies have provided numerous insights over the past years on how members of the nuclear hormone receptor superfamily tightly regulate the expression of drug-metabolizing enzymes and transporters. Besides the role of the farnesoid X receptor (FXR) in the transcriptional control of bile acid transport and metabolism, this review provides an overview on how this metabolic sensor prevents the accumulation of toxic byproducts derived from endogenous metabolites, as well as of exogenous chemicals, in coordination with the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Decrypting this network should provide cues to better understand how these metabolic nuclear receptors participate in physiologic and pathologic processes with potential validation as therapeutic targets in human disabilities and cancers.
Collapse
|
13
|
Lodato NJ, Melia T, Rampersaud A, Waxman DJ. Sex-Differential Responses of Tumor Promotion-Associated Genes and Dysregulation of Novel Long Noncoding RNAs in Constitutive Androstane Receptor-Activated Mouse Liver. Toxicol Sci 2018; 159:25-41. [PMID: 28903501 DOI: 10.1093/toxsci/kfx114] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Xenobiotic agonists of constitutive androstane receptor (CAR) induce many hepatic drug metabolizing enzymes, but following prolonged exposure, promote hepatocellular carcinoma, most notably in male mouse liver. Here, we used nuclear RNA-seq to characterize global changes in the mouse liver transcriptome following exposure to the CAR-specific agonist ligand 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP), including changes in novel long noncoding RNAs that may contribute to xenobiotic-induced pathophysiology. Protein-coding genes dysregulated by 3 h TCPOBOP exposure were strongly enriched in KEGG pathways of xenobiotic and drug metabolism, with stronger and more extensive gene responses observed in female than male liver. After 27 h TCPOBOP exposure, the number of responsive genes increased >8-fold in males, where the top enriched pathways and their upstream regulators expanded to include factors implicated in cell cycle dysregulation and hepatocellular carcinoma progression (cyclin-D1, oncogenes E2f, Yap, Rb, Myc, and proto-oncogenes β-catenin, FoxM1, FoxO1, all predicted to be activated by TCPOBOP in male but not female liver; and tumor suppressors p21 and p53, both predicted to be inhibited). Upstream regulators uniquely associated with 3 h TCPOBOP-exposed females include TNF/NFkB pathway members, which negatively regulate CAR-dependent proliferative responses and may contribute to the relative resistance of female liver to TCPOBOP-induced tumor promotion. These responses may be modified by the many long noncoding liver RNAs we show are dysregulated by TCPOBOP or pregnane-X-receptor agonist exposure, including lncRNAs proximal to CAR target genes Cyp2b10, Por, and Alas1. These data provide a comprehensive view of the CAR-regulated transcriptome and give insight into the mechanism of sex-biased susceptibility to CAR-dependent mouse liver tumorigenesis.
Collapse
Affiliation(s)
- Nicholas J Lodato
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - Tisha Melia
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - Andy Rampersaud
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
14
|
Buchman CD, Chai SC, Chen T. A current structural perspective on PXR and CAR in drug metabolism. Expert Opin Drug Metab Toxicol 2018; 14:635-647. [PMID: 29757018 DOI: 10.1080/17425255.2018.1476488] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Pregnane X receptor (PXR) and the constitutive androstane receptor (CAR) are two members of the nuclear receptor superfamily that play major roles in the expression of various drug metabolism enzymes and are known for their ligand promiscuity. As with other nuclear receptors, PXR and CAR are each composed of a ligand-binding domain (LBD) and a DNA-binding domain (DBD) connected by a hinge region. Areas covered: This review focuses on the information obtained over the last 15+ years from X-ray crystallography studies of the structure of PXR and CAR. Areas of focus include the mobility of each structure, based on temperature factors (B factors); multimeric interactions; the binding of coregulators and ligands; and how the crystal structures were obtained. The first use of hydrogen-deuterium exchange coupled with mass spectroscopy (HDX-MS) to study compound-protein interactions in the PXR-LBD is also addressed. Expert opinion: X-ray crystallography studies have provided us with an excellent understanding of how the LBDs of each receptor function; however, many questions remain concerning the structure of these receptors. Future research should focus on determining the co-crystal structure of an antagonist bound to PXR and on studying the structural aspects of the full-length CAR and PXR proteins.
Collapse
Affiliation(s)
- Cameron D Buchman
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Sergio C Chai
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Taosheng Chen
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| |
Collapse
|
15
|
How similar is similar enough? A sufficient similarity case study with Ginkgo biloba extract. Food Chem Toxicol 2018; 118:328-339. [PMID: 29752982 DOI: 10.1016/j.fct.2018.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 12/20/2022]
Abstract
Botanical dietary supplements are complex mixtures that can be highly variable in composition and quality, making safety evaluation difficult. A key challenge is determining how diverse products in the marketplace relate to chemically and toxicologically characterized reference samples (i.e., how similar must a product be in order to be well-represented by the tested reference sample?). Ginkgo biloba extract (GBE) was used as a case study to develop and evaluate approaches for determining sufficient similarity. Multiple GBE extracts were evaluated for chemical and biological-response similarity. Chemical similarity was assessed using untargeted and targeted chemistry approaches. Biological similarity was evaluated using in vitro liver models and short-term rodent studies. Statistical and data visualization methods were then used to make decisions about the similarity of products to the reference sample. A majority of the 26 GBE samples tested (62%) were consistently determined to be sufficiently similar to the reference sample, while 27% were different from the reference GBE, and 12% were either similar or different depending on the method used. This case study demonstrated that approaches to evaluate sufficient similarity allow for critical evaluation of complex mixtures so that safety data from the tested reference can be applied to untested materials.
Collapse
|
16
|
The Role of PPAR and Its Cross-Talk with CAR and LXR in Obesity and Atherosclerosis. Int J Mol Sci 2018; 19:ijms19041260. [PMID: 29690611 PMCID: PMC5979375 DOI: 10.3390/ijms19041260] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/13/2018] [Accepted: 04/19/2018] [Indexed: 02/06/2023] Open
Abstract
The prevalence of obesity and atherosclerosis has substantially increased worldwide over the past several decades. Peroxisome proliferator-activated receptors (PPARs), as fatty acids sensors, have been therapeutic targets in several human lipid metabolic diseases, such as obesity, atherosclerosis, diabetes, hyperlipidaemia, and non-alcoholic fatty liver disease. Constitutive androstane receptor (CAR) and liver X receptors (LXRs) were also reported as potential therapeutic targets for the treatment of obesity and atherosclerosis, respectively. Further clarification of the internal relationships between these three lipid metabolic nuclear receptors is necessary to enable drug discovery. In this review, we mainly summarized the cross-talk of PPARs-CAR in obesity and PPARs-LXRs in atherosclerosis.
Collapse
|
17
|
Allinson M, Kameda Y, Kimura K, Allinson G. Occurrence and assessment of the risk of ultraviolet filters and light stabilizers in Victorian estuaries. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:12022-12033. [PMID: 29453716 DOI: 10.1007/s11356-018-1386-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/24/2018] [Indexed: 06/08/2023]
Abstract
This reconnaissance study was undertaken to examine the occurrence of common ultraviolet filters (UVF) and light stabilizers (UVLS), and preservatives in four different estuaries in Port Philip Bay, Victoria, for the first time. In total, 11 UV filters, 10 UV stabilizers, 12 preservatives and a metabolite, and one fragrance were screened in grab samples of water and sediment using a combination of solid phase extraction and gas and liquid chromatography mass spectrometry measurement techniques. In that context, 16 of the UVF and UVLS and 5 of the preservatives screened were observed in water and/or sediment samples. There are no marine water quality guideline values for any of the fragrances, preservatives and UV filters and light stabilizers in Australia's current national water quality guidelines, so potential risk was assessed using the risk quotient (RQ) and toxic unit (TU) concepts. In that context, only two chemicals (OC and EHMC) had both an RQ above 1 and a log10TU above - 3, suggesting that few of the screened chemicals would have posed an individual, short-term risk to organisms in the waters studied at the time of sampling. However, the detection of common UV filters, such as 4MBC, EHMC, OC and the common preservatives 2-PE, MP, and PB in these Victorian estuaries highlights that the existence of personal care products in the environment is not just an issue for more densley populated countries in the northern hemisphere, but also potentially of concern in Australia. And, in that context, more sampling campaigns in Port Philip Bay are of paramount importance to assess the potential risk posed by these compounds to aquatic ecosystems.
Collapse
Affiliation(s)
- Mayumi Allinson
- Centre for Aquatic Pollution Identification and Management (CAPIM), School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yutaka Kameda
- Chiba Institute of Technology, Architecture and Civil Engineering, 2-17-1 Tsudanuma, Narashino, Chiba, 275-0016, Japan
| | - Kumiko Kimura
- Saitama City Institute of Health Science and Research, 7-5-12 Suzuya, Chuo-ku, Saitama, 338-0013, Japan
| | - Graeme Allinson
- Future Farming Systems Research Division, Department of Environment and Primary Industries, DEPI Queenscliff Centre, Queenscliff, Victoria, 3225, Australia.
- School of Science, RMIT University, Melbourne, Victoria, 3001, Australia.
| |
Collapse
|
18
|
Bogen KT. Biphasic hCAR Inhibition-Activation by Two Aminoazo Liver Carcinogens. NUCLEAR RECEPTOR RESEARCH 2018. [DOI: 10.11131/2018/101321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
19
|
|
20
|
Hepatotoxicity of Herbal Supplements Mediated by Modulation of Cytochrome P450. Int J Mol Sci 2017; 18:ijms18112353. [PMID: 29117101 PMCID: PMC5713322 DOI: 10.3390/ijms18112353] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 12/11/2022] Open
Abstract
Herbal supplements are a significant source of drug-drug interactions (DDIs), herb-drug interactions, and hepatotoxicity. Cytochrome P450 (CYP450) enzymes metabolize a large number of FDA-approved pharmaceuticals and herbal supplements. This metabolism of pharmaceuticals and supplements can be augmented by concomitant use of either pharmaceuticals or supplements. The xenobiotic receptors constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) can respond to xenobiotics by increasing the expression of a large number of genes that are involved in the metabolism of xenobiotics, including CYP450s. Conversely, but not exclusively, many xenobiotics can inhibit the activity of CYP450s. Induction of the expression or inhibition of the activity of CYP450s can result in DDIs and toxicity. Currently, the United States (US) Food and Drug Administration does not require the investigation of the interactions of herbal supplements and CYP450s. This review provides a summary of herbal supplements that inhibit CYP450s, induce the expression of CYP450s, and/or whose toxicity is mediated by CYP450s.
Collapse
|
21
|
Kato H, Yamaotsu N, Iwazaki N, Okamura S, Kume T, Hirono S. Precise prediction of activators for the human constitutive androstane receptor using structure-based three-dimensional quantitative structure–activity relationship methods. Drug Metab Pharmacokinet 2017; 32:179-188. [DOI: 10.1016/j.dmpk.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/24/2017] [Accepted: 02/01/2017] [Indexed: 02/07/2023]
|
22
|
Cherian MT, Yang L, Chai SC, Lin W, Chen T. Identification and Characterization of CINPA1 Metabolites Facilitates Structure-Activity Studies of the Constitutive Androstane Receptor. Drug Metab Dispos 2016; 44:1759-1770. [PMID: 27519550 PMCID: PMC5074473 DOI: 10.1124/dmd.116.071993] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/11/2016] [Indexed: 12/19/2022] Open
Abstract
The constitutive androstane receptor (CAR) regulates the expression of genes involved in drug metabolism and other processes. A specific inhibitor of CAR is critical for modulating constitutive CAR activity. We recently described a specific small-molecule inhibitor of CAR, CINPA1 (ethyl (5-(diethylglycyl)-10,11-dihydro-5H-dibenzo[b,f]azepin-3-yl)carbamate), which is capable of reducing CAR-mediated transcription by changing the coregulator recruitment pattern and reducing CAR occupancy at the promoter regions of its target genes. In this study, we showed that CINPA1 is converted to two main metabolites in human liver microsomes. By using cell-based reporter gene and biochemical coregulator recruitment assays, we showed that although metabolite 1 was very weak in inhibiting CAR function and disrupting CAR-coactivator interaction, metabolite 2 was inactive in this regard. Docking studies using the CAR ligand-binding domain structure showed that although CINPA1 and metabolite 1 can bind in the CAR ligand-binding pocket, metabolite 2 may be incapable of the molecular interactions required for binding. These results indicate that the metabolites of CINPA1 may not interfere with the action of CINPA1. We also used in vitro enzyme assays to identify the cytochrome P450 enzymes responsible for metabolizing CINPA1 in human liver microsomes and showed that CINPA1 was first converted to metabolite 1 by CYP3A4 and then further metabolized by CYP2D6 to metabolite 2. Identification and characterization of the metabolites of CINPA1 enabled structure-activity relationship studies of this family of small molecules and provided information to guide in vivo pharmacological studies.
Collapse
Affiliation(s)
- Milu T Cherian
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lei Yang
- 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
| | - Wenwei Lin
- 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
|
23
|
Lack of CAR impacts neuronal function and cerebrovascular integrity in vivo. Exp Neurol 2016; 283:39-48. [PMID: 27240521 DOI: 10.1016/j.expneurol.2016.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/13/2016] [Accepted: 05/12/2016] [Indexed: 12/12/2022]
Abstract
Nuclear receptors (NRs) are a group of transcription factors emerging as players in normal and pathological CNS development. Clinically, an association between the constitutive androstane NR (CAR) and cognitive impairment was proposed, however never experimentally investigated. We wished to test the hypothesis that the impact of CAR on neurophysiology and behavior is underlined by cerebrovascular-neuronal modifications. We have used CAR(-/-) C57BL/6 and wild type mice and performed a battery of behavioral tests (recognition, memory, motor coordination, learning and anxiety) as well as longitudinal video-electroencephalographic recordings (EEG). Brain cell morphology was assessed using 2-photon or electron microscopy and fluorescent immunohistochemistry. We observed recognition memory impairment and increased anxiety-like behavior in CAR(-/-) mice, while locomotor activity was not affected. Concomitantly to memory deficits, EEG monitoring revealed a decrease in 3.5-7Hz waves during the awake/exploration and sleep periods. Behavioral and EEG abnormalities in CAR(-/-) mice mirrored structural changes, including tortuous fronto-parietal penetrating vessels. At the cellular level we found reduced ZO-1, but not CLDN5, tight junction protein expression in cortical and hippocampal isolated microvessel preparations. Interestingly, the neurotoxin kainic acid, when injected peripherally, provoked a rapid onset of generalized convulsions in CAR(-/-) as compared to WT mice, supporting the hypothesis of vascular permeability. The morphological phenotype of CAR(-/-) mice also included some modifications of GFAP/IBA1 glial cells in the parenchymal or adjacent to collagen-IV(+) or FITC(+) microvessels. Neuronal defects were also observed including increased cortical NEUN(+) cell density, hippocampal granule cell dispersion and increased NPY immunoreactivity in the CA1 region in CAR(-/-) mice. The latter may contribute to the in vivo phenotype. Our results indicate that behavioral and electroencephalographic changes in adult CAR(-/-) mice are concomitant to discrete developmental or structural brain defects. The latter could increase the vulnerability to neurotoxins. The possibility that interfering with nuclear receptors during development could contribute to adulthood brain changes is proposed.
Collapse
|