1
|
Yano J, Kawamoto K, Shimotsuma Y, Matsunaga K, Abe J, Fukunaga S, Osimitz TG, Lake BG, Asano H. Mode of action analysis for rat thyroid gland follicular cell tumor formation by MGK-264 and human relevance. Regul Toxicol Pharmacol 2025; 160:105834. [PMID: 40315979 DOI: 10.1016/j.yrtph.2025.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/06/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
MGK-264 (N-(2-ethylhexyl)-5-norborene-2.3-dicarboximide or N-octyl bicycloheptene dicarboximide), an insecticidal synergist, produced thyroid gland follicular cell (TFC) tumors in male Sprague-Dawley (SD) rats in a carcinogenicity study. The purpose of this study was to evaluate the possible mode of action (MoA) for TFC tumor induction by MGK-264 and its relevance to humans. In short-term in vivo studies, the treatment of male SD rats with MGK-264 resulted in induction of hepatic UDPglucuronosyltransferase (UGT) activity towards thyroxine (T4) as substrate (UGT activity), a decrease in serum T4 levels, an increase in serum thyroid stimulating hormone levels, and TFC hypertrophy at MGK-264 dose levels where TFC tumors were noted in the carcinogenicity study. Other possible MoAs such as genotoxicity, thyroperoxidase inhibition, and sodium/iodide symporter inhibition were excluded. Therefore, it is reasonable to conclude that MGK-264 has mitogenic activity on TFCs via induction of hepatic UGT activity followed by perturbation of the hypothalamus-pituitary-thyroid axis, similar to other hepatic xenobiotic enzyme inducers like phenobarbital. Literature data demonstrates that there are marked species differences between rats and humans in the effects of hepatic xenobiotic enzyme inducers on thyroid hormones and the thyroid gland. Overall, the proposed MoA for MGK-264-induced rat TFC tumor formation is considered quantitatively not plausible for humans.
Collapse
Affiliation(s)
- Junji Yano
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan.
| | - Kensuke Kawamoto
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan
| | - Yukako Shimotsuma
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan
| | - Kohei Matsunaga
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan
| | - Jun Abe
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan
| | - Satoki Fukunaga
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan
| | | | - Brian G Lake
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom
| | - Hiroyuki Asano
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-8558, Japan
| |
Collapse
|
2
|
Yang F, Wu L, Xu W, Wu Y, Zhu S, Zhang Y, Chong Y, Peng L. Sodium taurocholate co-transporting polypeptide deficiency attenuates acetaminophen-induced hepatotoxicity via regulating expression of drug metabolism enzymes in mice. Toxicol Appl Pharmacol 2025; 497:117266. [PMID: 39947258 DOI: 10.1016/j.taap.2025.117266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Acetaminophen (APAP) overdose can induce liver injury and is generally accompanied by disruption of bile acid homeostasis. Physiologically, sodium taurocholate co-transporting polypeptide (NTCP) participates in the uptake of bile acids from portal blood into hepatocytes to maintain enterohepatic recirculation but its role in APAP-induced hepatotoxicity is unclear. Wild-type (WT) C57BL/6J and NTCP knockout (KO) mice were injected with 400mg/kg APAP and liver injury was evaluated by serum biochemical markers and histologic evaluation. RNA-seq analysis was performed to evaluate the liver gene expression profiles in APAP-treated mice. Compared with WT mice, the exposure to APAP overdose caused liver dysfunction, oxidative stress, inflammation and cell death, which were ameliorated by NTCP deficiency. APAP detoxification, metabolism, and elimination were significantly accelerated by the upregulation of UDP-glucuronosyltransferase (Ugt1a1, Ugt1a6 and Ugt1a9), sulfotransferase (Sult1a1 and Sult2a1) and bile acid efflux transporters (Abcc2/3/4) in NTCP KO mice compared with WT mice. Interestingly, APAP-induced hepatotoxicity was ameliorated using Irbesartan and Ezetimibe (NTCP inhibitors). In conclusion, NTCP deficiency attenuates APAP-induced hepatotoxicity by enhancing the metabolism and elimination of APAP. NTCP inhibitors protect against APAP-induced hepatotoxicity and thus are a potential therapeutic option.
Collapse
Affiliation(s)
- Fangji Yang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Department of Critical Care Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lina Wu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenxiong Xu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuankai Wu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shu Zhu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuzhen Zhang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yutian Chong
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Key Laboratory of Liver Disease of Guangdong Province, Guangzhou, China.
| | - Liang Peng
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Key Laboratory of Liver Disease of Guangdong Province, Guangzhou, China.
| |
Collapse
|
3
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
4
|
Baze A, Wiss L, Horbal L, Biemel K, Asselin L, Richert L. Comparison of in vitro thyroxine (T4) metabolism between Wistar rat and human hepatocyte cultures. Toxicol In Vitro 2024; 96:105763. [PMID: 38142784 DOI: 10.1016/j.tiv.2023.105763] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/24/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
In vitro assays remain relatively new in exploring human relevance of liver, in particular nuclear receptor-mediated perturbations of the hypothalamus-pituitary-thyroid axis seen in rodents, mainly in the rat. Consistent with in vivo data, we confirm that thyroid hormone thyroxine metabolism was 9 times higher in primary rat hepatocytes (PRH) than in primary human hepatocytes (PHH) cultured in a 2D sandwich (2Dsw) configuration. In addition, thyroxine glucuronide (T4-G) was by far the major metabolite formed in both species (99.1% in PRH and 69.7% in PHH) followed by thyroxine sulfate (T4-S, 0.7% in PRH and 18.1% in PHH) and triiodothyronine/reverse triiodothyronine (T3/rT3, 0.2% in PRH and 12.2% in PHH). After a 7-day daily exposure to orphan receptor-mediated liver inducers, T4 metabolism was strongly increased in PRH, almost exclusively through increased T4-G formation. These results were consistent with the inductions of glucuronosyltransferase Ugt2b1 and canalicular transporter Mrp2. PHH also responded to activation of the three nuclear receptors, with mainly induction of glucuronosyltransferase UGT1A1 and canalicular transporter MRP2. Despite this, T4 disappearance rate and secreted T4 metabolites were only slightly increased in PHH. Overall, our data highlight that cryopreserved hepatocytes in 2Dsw culture allowing long-term exposure and species comparison are of major interest in improving liver-mediated human safety assessment.
Collapse
Affiliation(s)
- Audrey Baze
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Lucille Wiss
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Liliia Horbal
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrüken, Germany
| | - Klaus Biemel
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrüken, Germany
| | - Laure Asselin
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Lysiane Richert
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France; Zylan SAS, 8 rue de la Haute Corniche, 67210 Obernai, France.
| |
Collapse
|
5
|
Piell KM, Petri BJ, Xu J, Cai L, Rai SN, Li M, Wilkey DW, Merchant ML, Cave MC, Klinge CM. Chronic Aroclor 1260 exposure alters the mouse liver proteome, selenoproteins, and metals in steatotic liver disease. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104430. [PMID: 38552755 PMCID: PMC11044900 DOI: 10.1016/j.etap.2024.104430] [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: 01/09/2024] [Revised: 03/19/2024] [Accepted: 03/23/2024] [Indexed: 04/09/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to increase due in part to the obesity epidemic and to environmental exposures to metabolism disrupting chemicals. A single gavage exposure of male mice to Aroclor 1260 (Ar1260), an environmentally relevant mixture of non-dioxin-like polychlorinated biphenyls (PCBs), resulted in steatohepatitis and altered RNA modifications in selenocysteine tRNA 34 weeks post-exposure. Unbiased approaches identified the liver proteome, selenoproteins, and levels of 25 metals. Ar1260 altered the abundance of 128 proteins. Enrichment analysis of the liver Ar1260 proteome included glutathione metabolism and translation of selenoproteins. Hepatic glutathione peroxidase 4 (GPX4) and Selenoprotein O (SELENOO) were increased and Selenoprotein F (SELENOF), Selenoprotein S (SELENOS), Selenium binding protein 2 (SELENBP2) were decreased with Ar1260 exposure. Increased copper, selenium (Se), and zinc and reduced iron levels were detected. These data demonstrate that Ar1260 exposure alters the (seleno)proteome, Se, and metals in MASLD-associated pathways.
Collapse
Affiliation(s)
- Kellianne M Piell
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Belinda J Petri
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40292, USA; Kentucky IDeA Networks of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Jason Xu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40292, USA; Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA; University of Louisville Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville, Louisville, KY 40292, USA
| | - Shesh N Rai
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ming Li
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Daniel W Wilkey
- University of Louisville Hepatobiology and Toxicology Center; University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Michael L Merchant
- University of Louisville Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville, Louisville, KY 40292, USA; Division of Nephrology & Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA; University of Louisville Hepatobiology and Toxicology Center; University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Matthew C Cave
- University of Louisville Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville, Louisville, KY 40292, USA; University of Louisville Hepatobiology and Toxicology Center; University of Louisville School of Medicine, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA; The University of Louisville Superfund Research Center, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Carolyn M Klinge
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40292, USA; University of Louisville Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
6
|
Kurosawa K, Nakano M, Yokoseki I, Nagaoka M, Takemoto S, Sakai Y, Kobayashi K, Kazuki Y, Fukami T, Nakajima M. ncBAF enhances PXR-mediated transcriptional activation in the human and mouse liver. Biochem Pharmacol 2023; 215:115733. [PMID: 37543347 DOI: 10.1016/j.bcp.2023.115733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/25/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Pregnane X receptor (PXR) is one of the key regulators of drug metabolism, gluconeogenesis, and lipid synthesis in the human liver. Activation of PXR by drugs such as rifampicin, simvastatin, and efavirenz causes adverse reactions such as drug-drug interaction, hyperglycemia, and dyslipidemia. The inhibition of PXR activation has merit in preventing such adverse events. Here, we demonstrated that bromodomain containing protein 9 (BRD9), a component of non-canonical brahma-related gene 1-associated factor (ncBAF), one of the chromatin remodelers, interacts with PXR. Rifampicin-mediated induction of CYP3A4 expression was attenuated by iBRD9, an inhibitor of BRD9, in human primary hepatocytes and CYP3A/PXR-humanized mice, indicating that BRD9 enhances the transcriptional activation of PXR in vitro and in vivo. Chromatin immunoprecipitation assay reveled that iBRD9 treatment resulted in attenuation of the rifampicin-mediated binding of PXR to the CYP3A4 promoter region, suggesting that ncBAF functions to facilitate the binding of PXR to its response elements. Efavirenz-induced hepatic lipid accumulation was attenuated by iBRD9 in C57BL/6J mice, suggesting that the inhibition of BRD9 would be useful to reduce the risk of efavirenz-induced hepatic steatosis. Collectively, we found that inhibitors of BRD9, a component of ncBAF that plays a role in assisting transactivation by PXR, would be useful to reduce the risk of PXR-mediated adverse reactions.
Collapse
Affiliation(s)
- Kiamu Kurosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| | - Itsuki Yokoseki
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Seiya Takemoto
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kaoru Kobayashi
- Laboratory of Biopharmaceutics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Yasuhiro Kazuki
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan; Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute (WPI-NanoLSI) Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
7
|
Riddick DS. Fifty Years of Aryl Hydrocarbon Receptor Research as Reflected in the Pages of Drug Metabolism and Disposition. Drug Metab Dispos 2023; 51:657-671. [PMID: 36653119 DOI: 10.1124/dmd.122.001009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
The induction of multiple drug-metabolizing enzymes by halogenated and polycyclic aromatic hydrocarbon toxicants is mediated by the aryl hydrocarbon receptor (AHR). This fascinating receptor also has natural dietary and endogenous ligands, and much is now appreciated about the AHR's developmental and physiologic roles, as well as its importance in cancer and other diseases. The past several years has witnessed increasing emphasis on understanding the multifaceted roles of the AHR in the immune system. Most would agree that the "discovery" of the AHR occurred in 1976, with the report of specific binding of a high affinity radioligand in mouse liver, just three years after the launch of the journal Drug Metabolism and Disposition (DMD) in 1973. Over the ensuing 50 years, the AHR and DMD have led parallel and often intersecting lives. The overall goal of this mini-review is to provide a decade-by-decade overview of major historical landmark discoveries in the AHR field and to highlight the numerous contributions made by publications appearing in the pages of DMD. It is hoped that this historical tour might inspire current and future research in the AHR field. SIGNIFICANCE STATEMENT: With the launch of Drug Metabolism and Disposition (DMD) in 1973 and the discovery of the aryl hydrocarbon receptor (AHR) in 1976, the journal and the receptor have led parallel and often intersecting lives over the past 50 years. Tracing the history of the AHR can reveal how knowledge in the field has evolved to the present and highlight the important contributions made by discoveries reported in DMD. This may inspire additional DMD papers reporting future AHR landmark discoveries.
Collapse
Affiliation(s)
- David S Riddick
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Miners JO, Polasek TM, Hulin JA, Rowland A, Meech R. Drug-drug interactions that alter the exposure of glucuronidated drugs: Scope, UDP-glucuronosyltransferase (UGT) enzyme selectivity, mechanisms (inhibition and induction), and clinical significance. Pharmacol Ther 2023:108459. [PMID: 37263383 DOI: 10.1016/j.pharmthera.2023.108459] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Drug-drug interactions (DDIs) arising from the perturbation of drug metabolising enzyme activities represent both a clinical problem and a potential economic loss for the pharmaceutical industry. DDIs involving glucuronidated drugs have historically attracted little attention and there is a perception that interactions are of minor clinical relevance. This review critically examines the scope and aetiology of DDIs that result in altered exposure of glucuronidated drugs. Interaction mechanisms, namely inhibition and induction of UDP-glucuronosyltransferase (UGT) enzymes and the potential interplay with drug transporters, are reviewed in detail, as is the clinical significance of known DDIs. Altered victim drug exposure arising from modulation of UGT enzyme activities is relatively common and, notably, the incidence and importance of UGT induction as a DDI mechanism is greater than generally believed. Numerous DDIs are clinically relevant, resulting in either loss of efficacy or an increased risk of adverse effects, necessitating dose individualisation. Several generalisations relating to the likelihood of DDIs can be drawn from the known substrate and inhibitor selectivities of UGT enzymes, highlighting the importance of comprehensive reaction phenotyping studies at an early stage of drug development. Further, rigorous assessment of the DDI liability of new chemical entities that undergo glucuronidation to a significant extent has been recommended recently by regulatory guidance. Although evidence-based approaches exist for the in vitro characterisation of UGT enzyme inhibition and induction, the availability of drugs considered appropriate for use as 'probe' substrates in clinical DDI studies is limited and this should be research priority.
Collapse
Affiliation(s)
- John O Miners
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Thomas M Polasek
- Certara, Princeton, NJ, USA; Centre for Medicines Use and Safety, Monash University, Melbourne, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Andrew Rowland
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
9
|
Motohira K, Yohannes YB, Ikenaka Y, Eguchi A, Nakayama SM, Wepener V, Smit NJ, VAN Vuren JH, Ishizuka M. Investigation of dichlorodiphenyltrichloroethane (DDT) on xenobiotic enzyme disruption and metabolomic bile acid biosynthesis in DDT-sprayed areas using wild rats. J Vet Med Sci 2023; 85:236-243. [PMID: 36596564 PMCID: PMC10017292 DOI: 10.1292/jvms.22-0490] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Dichlorodiphenyltrichloroethane (DDT) is an organochlorine insecticide used worldwide. Several studies have reported the toxic effects of DDT and its metabolites on steroid hormone biosynthesis; however, its environmental effects are not well understood. This study examined wild rats collected in DDT-sprayed areas of South Africa and quantified plasma metabolites using liquid chromatography quadrupole time-of-flight mass spectrometry (LC-Q-TOF-MS). Fold change analysis of the metabolome revealed the effect of DDT on bile acid biosynthesis. Gene expression of the related enzyme in rat liver samples was also quantified. Significant association was found between DDT and gene expression levels related to constitutive androstane receptor mediated enzymes, such as Cyp2b1 in rat livers. However, our results could not fully demonstrate that enzymes related to bile acid biosynthesis were strongly affected by DDT. The correlation between DDT concentration and gene expression involved in steroid hormone synthesis in testis was also evaluated; however, no significant correlation was found. The disturbance of metabolic enzymes occurred in rat liver in the target area. Our results suggest that DDT exposure affects gene expression in wild rats living in DDT-sprayed areas. Therefore, there is a need for DDT toxicity evaluation in mammals living in DDT-sprayed areas. We could not find an effective biomarker that could reflect the mechanism of DDT exposure; however, this approach can provide new insights for future research to evaluate DDT effects in sprayed areas.
Collapse
Affiliation(s)
- Kodai Motohira
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Laboratory of Toxicology, Hokkaido University, Hokkaido, Japan
| | - Yared Beyene Yohannes
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Laboratory of Toxicology, Hokkaido University, Hokkaido, Japan.,Department of Chemistry, College of Natural and Computational Science, University of Gondar, Gondar, Ethiopia
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Laboratory of Toxicology, Hokkaido University, Hokkaido, Japan.,Water Research Group, Unit for Environmental Sciences and Management, Potchefstroom Campus, North-West University, Potchefstroom, South Africa.,Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan.,One Health Research Center, Hokkaido University, Hokkaido, Japan
| | - Akifumi Eguchi
- Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Shouta Mm Nakayama
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Laboratory of Toxicology, Hokkaido University, Hokkaido, Japan.,Biomedical Sciences Department, School of Veterinary Medicine, The University of Zambia, Lusaka, Zambia
| | - Victor Wepener
- Water Research Group, Unit for Environmental Sciences and Management, Potchefstroom Campus, North-West University, Potchefstroom, South Africa
| | - Nico J Smit
- Water Research Group, Unit for Environmental Sciences and Management, Potchefstroom Campus, North-West University, Potchefstroom, South Africa
| | - Johan Hj VAN Vuren
- Water Research Group, Unit for Environmental Sciences and Management, Potchefstroom Campus, North-West University, Potchefstroom, South Africa
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Laboratory of Toxicology, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
10
|
Metabolic Profiling and Investigation of the Modulatory Effect of Fagonia cretica L. Aerial Parts on Hepatic CYP3A4 and UGT2B7 Enzymes in Streptozotocin-Induced Diabetic Model. Antioxidants (Basel) 2023; 12:antiox12010119. [PMID: 36670981 PMCID: PMC9854966 DOI: 10.3390/antiox12010119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Drug-metabolizing enzymes are either boosted or suppressed by diabetes mellitus. This research was designed to explore Fagonia cretica L. aerial parts' impact on CYP3A4 and UGT2B7 activity and their mRNA expression in diabetic rats. Fagonia cretica (F. cretica) dried powder was sequentially extracted with n-hexane, chloroform, ethyl acetate, methanol, and water. The methanol extract and aqueous fraction presented the most significant potential to decrease the concentration of alpha-hydroxyl midazolam, with 176.0 ± 0.85 mg/Kg and 182.9 ± 0.99 mg/Kg, respectively, compared to the streptozotocin (STZ)-induced diabetic group, reflecting the inhibition in CYP3A4 activity. The fold change in mRNA expression of CYP3A4 was decreased significantly by the methanol extract, and the aqueous fraction of F. cretica estimated by 0.15 ± 0.002 and 0.16 ± 0.001, respectively, compared with the diabetic group. Morphine metabolism was significantly increased in rats treated with F. cretica methanol extract and its aqueous fraction, displaying 93.4 ± 0.96 mg/Kg and 96.4 ± 1.27 mg/Kg, respectively, compared with the metabolism of morphine in the diabetic group, which highlights the induction of UGT2B7 activity. The fold change in mRNA expression of UGT2B7 was significantly increased by the methanol extract and the aqueous fraction, estimated at 8.14 ± 0.26 and 7.17 ± 0.23 respectively, compared to the diabetic group. Phytochemical analysis was performed using high-performance liquid chromatography (HPLC), where the methanol extract showed more flavonoids and phenolic compounds compared to the aqueous fraction of F. cretica. The obtained results were further consolidated by molecular docking studies, where quercetin showed the best fitting within the active pocket of CYP3A4, followed by gallic acid, displaying free binding energies (∆G) of -30.83 and -23.12 kcal/mol, respectively. Thus, F. cretica could serve as a complementary medicine with standard anti-diabetic therapy that can modulate the activity of the drug-metabolizing enzymes.
Collapse
|
11
|
Vansell NR. Mechanisms by Which Inducers of Drug Metabolizing Enzymes Alter Thyroid Hormones in Rats. Drug Metab Dispos 2022; 50:508-517. [PMID: 35046065 DOI: 10.1124/dmd.121.000498] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/23/2021] [Indexed: 02/13/2025] Open
Abstract
Increased disposition of thyroid hormones is a way that xenobiotics may alter thyroid homeostasis and, in rats, produce thyroid follicular adenoma/carcinoma. This capacity is historically attributed to induction of thyroxine (T4) glucuronidation by UDP-glycosyltransferase (UGT) enzymes, and cytochrome P450 induction is often a surrogate. However, gaps exist in correlating the effectiveness of certain chemical inducers at increasing T4 glucuronidation with decreases in systemic T4 and resulting increases in thyroid-stimulating hormone. With the identification of other key inducible drug processing genes and proteins involved in hepatic disposition of thyroid hormones, including uptake (e.g., organic anion transporter polypeptides) and efflux (e.g., multidrug resistance proteins) transporters, data exist that support transporters as additional target sites of induction. These data are reviewed herein and indicate an increase in hepatic uptake of thyroid hormones, as well as increased biliary excretion of iodothyronine conjugates, represent critical activities that differentiate inducer effectiveness in disrupting thyroid hormones in rats. Increased membrane transport of thyroid hormones, likely in conjunction with induced glucuronidation of thyroid hormone (triiodothyronine more relevant than T4), provide a better indication of thyroid disrupting potential than consideration of UGT induction alone. Because coordinate regulation of these targets is inconsistent among inducers belonging to various classes and among species, and there are disparities between in vitro assays and in vivo responses, further work is required to identify specific and relevant inducible thyroid hormone uptake transporters. Data from Mrp2-null animals have contributed key information, yet the contributions of efflux transport (canalicular and basolateral) to the mechanism of individual, effective inducers also require further study. SIGNIFICANCE STATEMENT: Key advances in understanding the target sites for altered disposition of thyroid hormones have occurred in the last 2 decades to better inform potential sites of action of inducing chemicals. Ultimately, the knowledge of inducible thyroid hormone transport into and out of liver, beyond induction of glucuronidation, should be considered and applied to screening and risk assessment paradigms when assessing an inducer's potential to alter thyroid homeostasis in nonclinical species and humans.
Collapse
|
12
|
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: 20] [Impact Index Per Article: 6.7] [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
|
13
|
Cao ZM, Qiang J, Zhu JH, Li HX, Tao YF, He J, Xu P, Dong ZJ. Transcriptional inhibition of steroidogenic factor 1 in vivo in Oreochromis niloticus increased weight and suppressed gonad development. Gene 2022; 809:146023. [PMID: 34673205 DOI: 10.1016/j.gene.2021.146023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/09/2021] [Accepted: 10/14/2021] [Indexed: 11/16/2022]
Abstract
Steroidogenic factor 1 (sf1) (officially designated as nuclear receptor subfamily 5 group A member 1 [NR5A1]) is an important regulator of gonad development. Previous studies on sf1 in fish have been limited to cloning and in vitro expression experiments. In this study, we used antisense RNA to down-regulate sf1 transcription and sf1 protein expression. Down-regulation of sf1 resulted in an increase in body weight and inhibition of gonadal development in both males and females with the consequent lower gonadosomatic index compared to fish in the control group. Hematoxylin-eosin staining of the gonads of fish with down-regulated sf1 revealed fewer seminiferous tubules and sperm in the testis of males. In addition, the oocytes were mainly stage II and many of them were atretic follicle. We conducted comparative transcriptome and proteome analyses between the sf1-down-regulated group and the control group. These analyses revealed multiple gene-protein pairs and pathways involved in regulating the observed changes, including 44 and 74 differently expressed genes and proteins in males and females, respectively. The results indicated that dysfunctional retinal metabolism and fatty acid metabolism could be causes of the observed weight gain and gonad abnormalities in sf1-down-regulated fish. These findings demonstrate the feasibility of using antisense RNA for gene editing in fish. This methodology allows the study gene function in species less amenable to gene editing as for example aquaculture species with long life cycles.
Collapse
Affiliation(s)
- Zhe-Ming Cao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China
| | - Jun Qiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Jun-Hao Zhu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China
| | - Hong-Xia Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China
| | - Yi-Fan Tao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China
| | - Jie He
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China.
| | - Zai-Jie Dong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, Jiangsu, China.
| |
Collapse
|
14
|
Lv Y, Luo YY, Ren HW, Li CJ, Xiang ZX, Luan ZL. The role of pregnane X receptor (PXR) in substance metabolism. Front Endocrinol (Lausanne) 2022; 13:959902. [PMID: 36111293 PMCID: PMC9469194 DOI: 10.3389/fendo.2022.959902] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
As a member of the nuclear receptor (NR) superfamily, pregnane X receptor (PXR; NR1I2) is a ligand-activated transcription factor that plays a crucial role in the metabolism of xenobiotics and endobiotics in mammals. The tissue distribution of PXR is parallel to its function with high expression in the liver and small intestine and moderate expression in the kidney, stomach, skin, and blood-brain barrier, which are organs and tissues in frequent contact with xenobiotics. PXR was first recognized as an exogenous substance receptor regulating metabolizing enzymes and transporters and functioning in detoxification and drug metabolism in the liver. However, further research revealed that PXR acts as an equally important endogenous substance receptor in the metabolism and homeostasis of endogenous substances. In this review, we summarized the functions of PXR in metabolism of different substances such as glucose, lipid, bile acid, vitamin, minerals, and endocrines, and also included insights of the application of PXR ligands (drugs) in specific diseases.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian Medical University, Dalian, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian Medical University, Dalian, China
- *Correspondence: Zhi-Lin Luan,
| |
Collapse
|
15
|
Parmentier C, Baze A, Untrau M, Kampkoetter A, Lasserre D, Richert L. Evaluation of human relevance of Nicofluprole-induced rat thyroid disruption. Toxicol Appl Pharmacol 2021; 435:115831. [PMID: 34922950 DOI: 10.1016/j.taap.2021.115831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 10/19/2022]
Abstract
Nicofluprole is a novel insecticide of the phenylpyrazole class conferring selective antagonistic activity on insect GABA receptors. After repeated daily dietary administration to Wistar rats for 28/90 days, Nicofluprole induced increases in thyroid (and liver) weight, associated with histopathology changes. Nicofluprole did not inhibit thyroid peroxydase nor sodium/iodide symporter, two key players in the biosynthesis of thyroid hormones, indicating the absence of a direct thyroid effect. The results seen in rats suggested a mode of action of Nicofluprole driven by the molecular initiating event of CAR/PXR nuclear receptor activation in livers, with key events of increases in liver weight and hypertrophy, decreasing circulatory thyroid hormones, a compensatory increase in TSH release and follicular cell hypertrophy. To explore the relevance of these changes to humans, well established in vitro rat and human sandwich-cultured hepatocytes were exposed to Nicofluprole up to 7 days. A concentration-dependent CYP3A induction (PXR-activation), an increase in T4-glucuronoconjugation accompanied by UGT1A/2B inductions was observed in rat but not in human hepatocytes. The inductions seen with Nicofluprole in rat (in vivo and in vitro in hepatocytes) that were absent in human hepatocytes represent another example of species-selectivity of nuclear CAR/PXR receptor activators. Importantly, the different pattern observed in rat and human models demonstrate that Nicofluprole-related thyroid effects observed in the rat are with no human relevance.
Collapse
Affiliation(s)
- Céline Parmentier
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| | - Audrey Baze
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| | - Meiggie Untrau
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Andreas Kampkoetter
- Bayer Animal Health GmbH, An Elanco Animal Health Company, 50 Alfred-Nobel-Strasse, 40789 Monheim, Germany.
| | - Dominique Lasserre
- Bayer S.A.S. Bayer CropScience, 355 rue Dostoïevski, F-06560 Sophia Antipolis, France.
| | - Lysiane Richert
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| |
Collapse
|
16
|
Landerer S, Kalthoff S, Strassburg CP. UDP-glucuronosyltransferases mediate coffee-associated reduction of liver fibrosis in bile duct ligated humanized transgenic UGT1A mice. Hepatobiliary Surg Nutr 2021; 10:766-781. [PMID: 35004944 DOI: 10.21037/hbsn-20-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Coffee consumption has been shown to reduce the risk of liver fibrosis and is capable of inducing human UDP-glucuronosyltransferase (UGT) 1A genes. UGT1A enzymes act as indirect antioxidants catalyzing the elimination of reactive metabolites, which in turn are potent initiators of profibrotic mechanisms. The aim of this study was to analyze the role of UGT1A genes as effectors of the protective properties of coffee in bile duct ligation (BDL) induced liver fibrosis. METHODS Fourteen days BDL with and without coffee pre- and co-treatment was performed in htgUGT1A-WT and htgUGT1A-SNP mice. Hepatic UGT1A mRNA expression levels, serum bilirubin and aminotransferase activities were determined. Liver fibrosis was assessed by collagen deposition, computational analysis of Sirius red tissue staining and expression of profibrotic marker genes. Oxidative stress was measured by hepatic peroxidase concentrations and immunofluorescence staining. RESULTS UGT1A transcription was differentially activated in the livers of htgUGT1A-WT mice after BDL, in contrast to a reduced or absent induction in the presence of SNPs. Co-treated (coffee + BDL) htgUGT1A-WT-mice showed significantly increased UGT1A expression and protein levels and a considerably higher induction compared to water drinking WT mice (BDL), whereas in co-treated htgUGT1A-SNP mice absolute expression levels remained below those observed in htgUGT1A-WT mice. Collagen deposition, oxidative stress and the expression of profibrotic markers inversely correlated with UGT1A expression levels in htgUGT1A-WT and SNP mice after BDL and coffee + BDL co-treatment. CONCLUSIONS Coffee exerts hepatoprotective and antioxidative effects via activation of UGT1A enzymes. Attenuated hepatic fibrosis as a result of coffee-mediated UGT1A induction during cholestasis was detected, while the protective action of coffee was lower in a common low-function UGT1A SNP haplotype present in 10% of the Caucasian population. This study suggests that coffee consumption might constitute a potential strategy to support the conventional treatment of cholestasis-related liver diseases.
Collapse
Affiliation(s)
- Steffen Landerer
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, Bonn, Germany
| | - Sandra Kalthoff
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, Bonn, Germany
| | - Christian P Strassburg
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, Bonn, Germany
| |
Collapse
|
17
|
Suzuki N, Iwamura Y, Nakai T, Kato K, Otsuki A, Uruno A, Saigusa D, Taguchi K, Suzuki M, Shimizu R, Yumoto A, Okada R, Shirakawa M, Shiba D, Takahashi S, Suzuki T, Yamamoto M. Gene expression changes related to bone mineralization, blood pressure and lipid metabolism in mouse kidneys after space travel. Kidney Int 2021; 101:92-105. [PMID: 34767829 DOI: 10.1016/j.kint.2021.09.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022]
Abstract
Space travel burdens health by imposing considerable environmental stress associated with radioactivity and microgravity. In particular, gravity change predominantly impacts blood pressure and bone homeostasis, both of which are controlled mainly by the kidneys. Nuclear factor erythroid-2-related transcription factor 2 (Nrf2) plays essential roles in protecting the kidneys from various environmental stresses and injuries. To elucidate the effects of space travel on mammals in preparation for the upcoming space era, our study investigated the contribution of Nrf2 to kidney function in mice two days after their return from a 31-day stay in the International Space Station using Nrf2 knockout mice. Meaningfully, expression levels of genes regulating bone mineralization, blood pressure and lipid metabolism were found to be significantly altered in the kidneys after space travel in an Nrf2-independent manner. In particular, uridine diphosphate-glucuronosyltransferase 1A (Ugt1a) isoform genes were found to be expressed in an Nrf2-dependent manner and induced exclusively in the kidneys after return to Earth. Since spaceflight elevated the concentrations of fatty acids in the mouse plasma, we suggest that Ugt1a isoform expression in the kidneys was induced to promote glucuronidation of excessively accumulated lipids and excrete them into urine after the return from space. Thus, the kidneys were proven to play central roles in adaptation to gravity changes caused by going to and returning from space by controlling blood pressure and bone mineralization. Additionally, kidney Ugt1a isoform induction after space travel implies a significant role of the kidneys for space travelers in the excretion of excessive lipids.
Collapse
Affiliation(s)
- Norio Suzuki
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yuma Iwamura
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Nakai
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koichiro Kato
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihito Otsuki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Keiko Taguchi
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Mikiko Suzuki
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan; Center for Radioisotope Sciences, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ritsuko Shimizu
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akane Yumoto
- Japanese Experiment Module (JEM) Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Japan
| | - Risa Okada
- Japanese Experiment Module (JEM) Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Japan
| | - Masaki Shirakawa
- Japanese Experiment Module (JEM) Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Japan
| | - Dai Shiba
- Japanese Experiment Module (JEM) Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takafumi Suzuki
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
18
|
Kawase A, Mukai H, Tateishi S, Kuroda S, Kazaoka A, Satoh R, Shimada H, Sugiura R, Iwaki M. Protein Kinase N Family Negatively Regulates Constitutive Androstane Receptor-Mediated Transcriptional Induction of Cytochrome P450 2b10 in the Livers of Mice. J Pharmacol Exp Ther 2021; 379:53-63. [PMID: 34312179 DOI: 10.1124/jpet.121.000790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/22/2021] [Indexed: 01/04/2023] Open
Abstract
In receptor-type transcription factors-mediated cytochrome P450 (P450) induction, few studies have attempted to clarify the roles of protein kinase N (PKN) in the transcriptional regulation of P450s. This study aimed to examine the involvement of PKN in the transcriptional regulation of P450s by receptor-type transcription factors, including the aryl hydrocarbon receptor, constitutive androstane receptor (CAR), and pregnane X receptor. The mRNA and protein levels and metabolic activity of P450s in the livers of wild-type (WT) and double-mutant (D) mice harboring both PKN1 kinase-negative knock-in and PKN3 knockout mutations [PKN1 T778A/T778A; PKN3 -/-] were determined after treatment with activators for receptor-type transcription factors. mRNA and protein levels and metabolic activity of CYP2B10 were significantly higher in D mice treated with the CAR activator phenobarbital (PB) but not with 1,4-bis((3,5-dichloropyridin-2-yl)oxy)benzene compared with WT mice. We examined the CAR-dependent pathway regulated by PKN after PB treatment because the extent of CYP2B10 induction in WT and D mice was notably different in response to treatment with different CAR activators. The mRNA levels of Cyp2b10 in primary hepatocytes from WT and D mice treated with PB alone or in combination with Src kinase inhibitor 1 (SKI-1) or U0126 (a mitogen-activated protein kinase inhibitor) were evaluated. Treatment of hepatocytes from D mice with the combination of PB with U0126 but not SKI-1 significantly increased the mRNA levels of Cyp2b10 compared with those from the corresponding WT mice. These findings suggest that PKN may have inhibitory effects on the Src-receptor for activated C kinase 1 (RACK1) pathway in the CAR-mediated induction of Cyp2b10 in mice livers. SIGNIFICANCE STATEMENT: This is the first report of involvement of PKN in the transcriptional regulation of P450s. The elucidation of mechanisms responsible for induction of P450s could help optimize the pharmacotherapy and improve drug development. We examined whether the mRNA and protein levels and activities of P450s were altered in double-mutant mice harboring both PKN1 kinase-negative knock-in and PKN3 knockout mutations. PKN1/3 negatively regulates CAR-mediated induction of Cyp2b10 through phosphorylation of a signaling molecule in the Src-RACK1 pathway.
Collapse
Affiliation(s)
- Atsushi Kawase
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Hideyuki Mukai
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Shunsuke Tateishi
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Shintaro Kuroda
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Akira Kazaoka
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Ryosuke Satoh
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Hiroaki Shimada
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Reiko Sugiura
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| | - Masahiro Iwaki
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka, Japan (A.Kaw., S.T., S.K., A.Kaz., H.S., M.I.); Biosignal Research Center, Kobe University, Hyogo, Japan (H.M.); Department of Clinical Laboratory, Kitano Hospital, Osaka, Japan (H.M.); Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka, Japan (R.Sa., R.Su.); Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan (R.Su., M.I.); and Antiaging Center, Kindai University, Osaka, Japan (R.Su., M.I.)
| |
Collapse
|
19
|
Disease-drug and drug-drug interaction in COVID-19: Risk and assessment. Biomed Pharmacother 2021; 139:111642. [PMID: 33940506 PMCID: PMC8078916 DOI: 10.1016/j.biopha.2021.111642] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is announced as a global pandemic in 2020. Its mortality and morbidity rate are rapidly increasing, with limited medications. The emergent outbreak of COVID-19 prompted by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) keeps spreading. In this infection, a patient's immune response plays pivotal role in the pathogenesis. This inflammatory factor was shown by its mediators that, in severe cases, reach the cytokine at peaks. Hyperinflammatory state may sparks significant imbalances in transporters and drug metabolic machinery, and subsequent alteration of drug pharmacokinetics may result in unexpected therapeutic response. The present scenario has accounted for the requirement for therapeutic opportunities to relive and overcome this pandemic. Despite the diminishing developments of COVID-19, there is no drug still approved to have significant effects with no side effect on the treatment for COVID-19 patients. Based on the evidence, many antiviral and anti-inflammatory drugs have been authorized by the Food and Drug Administration (FDA) to treat the COVID-19 patients even though not knowing the possible drug-drug interactions (DDI). Remdesivir, favipiravir, and molnupiravir are deemed the most hopeful antiviral agents by improving infected patient’s health. Dexamethasone is the first known steroid medicine that saved the lives of seriously ill patients. Some oligopeptides and proteins have also been using. The current review summarizes medication updates to treat COVID-19 patients in an inflammatory state and their interaction with drug transporters and drug-metabolizing enzymes. It gives an opinion on the potential DDI that may permit the individualization of these drugs, thereby enhancing the safety and efficacy.
Collapse
|
20
|
Short-term standard alcohol consumption enhances platelet response to clopidogrel through inhibition of Nrf2/Ces1 pathway and induction of Cyp2c in mice. Life Sci 2021; 279:119268. [PMID: 33626394 DOI: 10.1016/j.lfs.2021.119268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/22/2022]
Abstract
AIMS Drinking alcohol is prevalent worldwide; however, it is unknown whether alcohol could affect the antiplatelet effects of clopidogrel in patients when taking both concomitantly. This study was designed to investigate the influence of short-term standard alcohol consumption on the metabolic activation of and platelet response to clopidogrel in mice as well as the mechanisms involved. MAIN METHODS Male C57BL/6J mice were administered with normal saline (vehicle control) or alcohol at 2 g/kg/day for 7 days, and then gavaged with vehicle control or a single dose of clopidogrel at 10 mg/kg. Inhibition of ADP-induced platelet aggregation and activation by clopidogrel, plasma concentrations of clopidogrel and its active metabolite H4, and changes in mRNA and protein expression of genes related to clopidogrel metabolism and its regulation were measured in mice pretreated with or without alcohol. KEY FINDINGS Compared with vehicle control, alcohol pretreatment significantly reduced hydrolysis of clopidogrel as a result of significant down-regulation of Nrf2-mediated Ces1 expression (responsible for the formation of clopidogrel carboxylate), increased metabolic activation of clopidogrel due to significant up-regulation of Cyp2c (for the formation of active thiol metabolite H4), and consequently enhanced inhibition of ADP-induced platelet aggregation and activation by clopidogrel. SIGNIFICANCE Short-term standard alcohol consumption would significantly enhance suppression of ADP-induced platelet aggregation and activation by clopidogrel through significant inhibition of Nrf2/Ces1 signaling pathway and induction of Cyp2c, suggesting that alcohol may interact with drugs that are predominantly metabolized by CES1 or CYP2C in patient care, including clopidogrel.
Collapse
|
21
|
Creeden JF, Gordon DM, Stec DE, Hinds TD. Bilirubin as a metabolic hormone: the physiological relevance of low levels. Am J Physiol Endocrinol Metab 2021; 320:E191-E207. [PMID: 33284088 PMCID: PMC8260361 DOI: 10.1152/ajpendo.00405.2020] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recent research on bilirubin, a historically well-known waste product of heme catabolism, suggests an entirely new function as a metabolic hormone that drives gene transcription by nuclear receptors. Studies are now revealing that low plasma bilirubin levels, defined as "hypobilirubinemia," are a possible new pathology analogous to the other end of the spectrum of extreme hyperbilirubinemia seen in patients with jaundice and liver dysfunction. Hypobilirubinemia is most commonly seen in patients with metabolic dysfunction, which may lead to cardiovascular complications and possibly stroke. We address the clinical significance of low bilirubin levels. A better understanding of bilirubin's hormonal function may explain why hypobilirubinemia might be deleterious. We present mechanisms by which bilirubin may be protective at mildly elevated levels and research directions that could generate treatment possibilities for patients with hypobilirubinemia, such as targeting of pathways that regulate its production or turnover or the newly designed bilirubin nanoparticles. Our review here calls for a shift in the perspective of an old molecule that could benefit millions of patients with hypobilirubinemia.
Collapse
Affiliation(s)
- Justin F Creeden
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Darren M Gordon
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - David E Stec
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
22
|
Jiao T, Yao X, Zhao Y, Zhou Y, Gao Y, Fan S, Chen P, Li X, Jiang Y, Yang X, Gonzalez FJ, Huang M, Bi H. Dexamethasone-Induced Liver Enlargement Is Related to PXR/YAP Activation and Lipid Accumulation but Not Hepatocyte Proliferation. Drug Metab Dispos 2020; 48:830-839. [PMID: 32561593 PMCID: PMC7497622 DOI: 10.1124/dmd.120.000061] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Dexamethasone (Dex), a widely prescribed anti-inflammatory drug, was reported to induce liver enlargement (hepatomegaly) in clinical practice and in animal models. However, the underlying mechanisms are not elucidated. Dex is a known activator of pregnane X receptor (PXR). Yes-associated protein (YAP) has been implicated in chemically induced liver enlargement. Here, the roles of PXR and YAP pathways were investigated in Dex-induced hepatomegaly. Upregulation of PXR downstream proteins, including cytochrome P450 (CYP) 3A11, 2B10, and organic anion transporter polypeptide 2 (OATP2), indicated PXR signaling was activated after high dose of Dex (50 mg/kg, i.p.), and Dex at 100 μM activated PXR in the dual-luciferase reporter gene assay. Dex also increased the expression of total YAP, nuclear YAP, and YAP downstream proteins, including connective tissue growth factor and cysteine-rich angiogenic inducer 61, indicating activation of the YAP pathway. Furthermore, nuclear translocation of YAP was promoted by activation of PXR. However, hepatocyte proliferation was inhibited with significant decrease in the expression of proliferation-related proteins cyclin D1 and proliferating cell nuclear antigen as well as other regulatory factors, such as forkhead box protein M1, c-MYC, and epidermal growth factor receptor. The inhibitory effect of Dex on hepatocyte proliferation was likely due to its anti-inflammation effect of suppression of inflammation factors. β-catenin staining revealed enlarged hepatocytes, which were mostly attributable to the accumulation of lipids, such as triglycerides. In summary, high-dose Dex increased liver size accompanied by enlarged hepatocytes, and this was due to the activation of PXR/YAP and their effects on lipid accumulation but not hepatocyte proliferation. These findings provide new insights for understanding the mechanism of Dex-induced hepatomegaly. SIGNIFICANCE STATEMENT: This study identified the roles of pregnane X receptor (PXR) and yes-associated protein (YAP) pathways in dexamethasone (Dex)-induced hepatomegaly. Dex induced PXR/YAP activation, enlarged hepatocytes, and promoted liver enlargement with lipid accumulation, such as triglycerides. However, hepatocyte proliferation was inhibited by the anti-inflammatory effect of Dex. These findings provide new insights for understanding the mechanism of Dex-induced hepatomegaly.
Collapse
Affiliation(s)
- Tingying Jiao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Xinpeng Yao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yingyuan Zhao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yanying Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yue Gao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Shicheng Fan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Panpan Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Xuan Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yiming Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Xiao Yang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Frank J Gonzalez
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| |
Collapse
|
23
|
Efavirenz-Based Antiretroviral Therapy Reduces Artemether-Lumefantrine Exposure for Malaria Treatment in HIV-Infected Pregnant Women. J Acquir Immune Defic Syndr 2020; 83:140-147. [PMID: 31929402 DOI: 10.1097/qai.0000000000002237] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The choice of malaria treatment for HIV-infected pregnant women receiving efavirenz-based antiretroviral therapy must consider the potential impact of drug interactions on antimalarial exposure and clinical response. The aim of this study was to investigate the effects of efavirenz on artemether-lumefantrine (AL) because no studies have isolated the impact of efavirenz for HIV-infected pregnant women. METHODS A prospective clinical pharmacokinetic (PK) study compared HIV-infected, efavirenz-treated pregnant women with HIV-uninfected pregnant women in Tororo, Uganda. All women received the standard 6-dose AL treatment regimen for Plasmodium falciparum malaria with intensive PK samples collected over 21 days and 42-days of clinical follow-up. PK exposure parameters were calculated for artemether, its active metabolite dihydroartemisinin (DHA), and lumefantrine to determine the impact of efavirenz. RESULTS Nine HIV-infected and 30 HIV-uninfected pregnant women completed intensive PK evaluations. Relative to controls, concomitant efavirenz therapy lowered the 8-hour artemether concentration by 76% (P = 0.013), DHA peak concentration by 46% (P = 0.033), and day 7 and 14 lumefantrine concentration by 61% and 81% (P = 0.046 and 0.023), respectively. In addition, there were nonsignificant reductions in DHA area under the concentration-time curve0-8hr (35%, P = 0.057) and lumefantrine area under the concentration-time curve0-∞ (34%, P = 0.063) with efavirenz therapy. CONCLUSIONS Pregnant HIV-infected women receiving efavirenz-based antiretroviral therapy during malaria treatment with AL showed reduced exposure to both the artemisinin and lumefantrine. These data suggest that malaria and HIV coinfected pregnant women may require adjustments in AL dosage or treatment duration to achieve exposure comparable with HIV-uninfected pregnant women.
Collapse
|
24
|
Rekka EA, Kourounakis PN, Pantelidou M. Xenobiotic Metabolising Enzymes: Impact on Pathologic Conditions, Drug Interactions and Drug Design. Curr Top Med Chem 2019; 19:276-291. [DOI: 10.2174/1568026619666190129122727] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 12/21/2022]
Abstract
Background:
The biotransformation of xenobiotics is a homeostatic defensive response of the
body against bioactive invaders. Xenobiotic metabolizing enzymes, important for the metabolism,
elimination and detoxification of exogenous agents, are found in most tissues and organs and are distinguished
into phase I and phase II enzymes, as well as phase III transporters. The cytochrome P450 superfamily
of enzymes plays a major role in the biotransformation of most xenobiotics as well as in the
metabolism of important endogenous substrates such as steroids and fatty acids. The activity and the
potential toxicity of numerous drugs are strongly influenced by their biotransformation, mainly accomplished
by the cytochrome P450 enzymes, one of the most versatile enzyme systems.
Objective:
In this review, considering the importance of drug metabolising enzymes in health and disease,
some of our previous research results are presented, which, combined with newer findings, may
assist in the elucidation of xenobiotic metabolism and in the development of more efficient drugs.
Conclusion:
Study of drug metabolism is of major importance for the development of drugs and provides
insight into the control of human health. This review is an effort towards this direction and may
find useful applications in related medical interventions or help in the development of more efficient
drugs.
Collapse
Affiliation(s)
- Eleni A. Rekka
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, Thessaloniki- 54124, Greece
| | - Panos N. Kourounakis
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, Thessaloniki- 54124, Greece
| | - Maria Pantelidou
- Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia 1036, Cyprus
| |
Collapse
|
25
|
Chen M, Guo L, Dong D, Yu F, Zhang T, Wu B. The nuclear receptor Shp regulates morphine withdrawal syndrome via modulation of Ugt2b expression in mice. Biochem Pharmacol 2019; 161:163-172. [DOI: 10.1016/j.bcp.2019.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 01/25/2019] [Indexed: 11/30/2022]
|
26
|
Abstract
PURPOSE OF REVIEW Mucosal immune cells in the intestinal tract are continuously exposed to a barrage of both foreign and endogenously generated metabolites, termed xenobiotics, and endobiotics, respectively. This review summarizes recent insights into the mechanisms by which xenobiotics and endobiotics regulate intestinal immunity and inflammation. RECENT FINDINGS The community of enteric microbes (i.e., microbiota) has profound impacts on the development and function of the mucosal immune system. The composition and function of gut microbiota is dynamically regulated by diet, and this interplay dictates which and how many immunomodulatory xenobiotics are present in the intestine. Microbiota also regulate the concentration and composition of circulating bile acids, an abundant class of liver-derived endobiotics with pleotropic immunoregulatory activities. A growing body of literature is emerging that sheds new light on the mechanisms by which xenobiotics and endobiotics interact with germline-encoded receptors and transporters to shape mucosal immune function. SUMMARY The complex and dynamic interplay among xenobiotics, endobiotics, and the mucosal immune system is a new frontier in mucosal immunology that is proving fruitful for the discovery of novel and pharmacologically accessible mechanisms with relevance to human inflammatory diseases.
Collapse
|
27
|
Wang X, Wang F, Lu Z, Jin X, Zhang Y. Semi-quantitative profiling of bile acids in serum and liver reveals the dosage-related effects of dexamethasone on bile acid metabolism in mice. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1095:65-74. [DOI: 10.1016/j.jchromb.2018.07.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 07/06/2018] [Accepted: 07/15/2018] [Indexed: 12/11/2022]
|
28
|
Bartsch R, Brinkmann B, Jahnke G, Laube B, Lohmann R, Michaelsen S, Neumann I, Greim H. Human relevance of follicular thyroid tumors in rodents caused by non-genotoxic substances. Regul Toxicol Pharmacol 2018; 98:199-208. [PMID: 30076866 DOI: 10.1016/j.yrtph.2018.07.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 12/31/2022]
Abstract
Chronic stimulation of the thyroid gland of rodents by TSH leads to thyroid follicular hyperplasia and subsequently to thyroid follicular adenomas and carcinomas. However, the interpretations of rodent thyroid tumors are contradictory. The U.S. Food and Drug Administration (FDA) concluded that findings with drugs that lead to increased levels of thyroid-stimulating hormone (TSH) in rats are not relevant to humans, whereas the U.S. Environmental Protection Agency (US EPA) concluded that chemicals that produce rodent thyroid tumors may pose a carcinogenic hazard for humans although the thyroid of rodents appears to be more sensitive to a carcinogenic stimulus than that of humans. Meanwhile, based on the CLP Criteria of the European Chemicals Agency (ECHA), rodent thyroid tumors caused by the induction of uridine-diphosphate-glucuronosyl transferases (UDGT) were assessed as not relevant to humans. To clarify these discrepant positions, the function and regulation of the thyroid gland are described and the types of thyroid tumors and the causes of their development in humans and animals are examined. Based on these data and the evidence that so far, except radiation, no chemical is known to increase the incidence of thyroid tumors in humans, it is concluded that rodent thyroid tumors resulting from continuous stimulation of the thyroid gland by increased TSH levels are not relevant to humans. Consequently, compounds that induce such tumors do not warrant classification as carcinogenic.
Collapse
Affiliation(s)
- Ruediger Bartsch
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Britta Brinkmann
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Gunnar Jahnke
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Britta Laube
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Ruth Lohmann
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Sandra Michaelsen
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Ingrid Neumann
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Helmut Greim
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany.
| |
Collapse
|
29
|
Morales-Prieto N, Ruiz-Laguna J, Sheehan D, Abril N. Transcriptome signatures of p,p´-DDE-induced liver damage in Mus spretus mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:150-167. [PMID: 29554563 DOI: 10.1016/j.envpol.2018.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/19/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023]
Abstract
The use of DDT (1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane) in some countries, although regulated, is contributing to an increased worldwide risk of exposure to this organochlorine pesticide or its derivative p,p'-DDE [1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene]. Many studies have associated p,p'-DDE exposure to type 2 diabetes, obesity and alterations of the reproductive system, but their molecular mechanisms of toxicity remain poorly understood. We have addressed this issue by using commercial microarrays based on probes for the entire Mus musculus genome to determine the hepatic transcriptional signatures of p,p'-DDE in the phylogenetically close mouse species Mus spretus. High-stringency hybridization conditions and analysis assured reliable results, which were also verified, in part, by qRT-PCR, immunoblotting and/or enzymatic activity. Our data linked 198 deregulated genes to mitochondrial dysfunction and perturbations of central signaling pathways (kinases, lipids, and retinoic acid) leading to enhanced lipogenesis and aerobic glycolysis, inflammation, cell proliferation and testosterone catabolism and excretion. Alterations of transcript levels of genes encoding enzymes involved in testosterone catabolism and excretion would explain the relationships established between p,p´-DDE exposure and reproductive disorders, obesity and diabetes. Further studies will help to fully understand the molecular basis of p,p´-DDE molecular toxicity in liver and reproductive organs, to identify effective exposure biomarkers and perhaps to design efficient p,p'-DDE exposure counteractive strategies.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Julia Ruiz-Laguna
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - David Sheehan
- College of Arts and Science, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| |
Collapse
|
30
|
Cai SA, Hou N, Zhao GJ, Liu XW, He YY, Liu HL, Hua YQ, Li LR, Huang Y, Ou CW, Luo CF, Chen MS. Nrf2 Is a Key Regulator on Puerarin Preventing Cardiac Fibrosis and Upregulating Metabolic Enzymes UGT1A1 in Rats. Front Pharmacol 2018; 9:540. [PMID: 29928229 PMCID: PMC5997811 DOI: 10.3389/fphar.2018.00540] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/04/2018] [Indexed: 01/25/2023] Open
Abstract
Puerarin is an isoflavone isolated from Radix puerariae. Emerging evidence shown that puerarin possesses therapeutic benefits that aid in the prevention of cardiovascular diseases. In this study, we evaluated the effects of puerarin on oxidative stress and cardiac fibrosis induced by abdominal aortic banding (AB) and angiotensin II (AngII). We also investigated the mechanisms underlying this phenomenon. The results of histopathological analysis, as well as measurements of collagen expression and cardiac fibroblast proliferation indicated that puerarin administration significantly inhibited cardiac fibrosis induced by AB and AngII. These effects of puerarin may reflect activation of Nrf2/ROS pathway. This hypothesis is supported by observed decreases of reactive oxygen species (ROS), decreases Keap 1, increases Nrf2 expression and nuclear translocation, and decreases of collagen expressions in cardiac fibroblasts treated with a combination of puerarin and AngII. Inhibition of Nrf2 with specific Nrf2 siRNA or Nrf2 inhibitor brusatol attenuated anti-fibrotic and anti-oxidant effects of puerarin. The metabolic effects of puerarin were mediated by Nrf2 through upregulation of UDP-glucuronosyltransferase (UGT) 1A1. The Nrf2 agonist tBHQ upregulated protein expression of UGT1A1 over time in cardiac fibroblasts. Treatment with Nrf2 siRNA or brusatol dramatically decreased UGT1A1 expression in puerarin-treated fibroblasts. The results of chromatin immunoprecipitation–qPCR further confirmed that puerarin significantly increased binding of Nrf2 to the promoter region of Ugt1a1. Western blot analysis showed that puerarin significantly inhibited AngII-induced phosphorylation of p38-MAPK. A specific inhibitor of p38-MAPK, SB203580, decreased collagen expression, and ROS generation induced by AngII in cardiac fibroblast. Together, these results suggest that puerarin prevents cardiac fibrosis via activation of Nrf2 and inactivation of p38-MAPK. Nrf2 is the key regulator of anti-fibrotic effects and upregulates metabolic enzymes UGT1A1. Autoregulatory circuits between puerarin and Nrf2-regulated UGT1A1 attenuates side effects associated with treatment, but it does not weaken puerarin’s pharmacological effects.
Collapse
Affiliation(s)
- Shao-Ai Cai
- Department of Cardiology, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ning Hou
- School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Gan-Jian Zhao
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xia-Wen Liu
- School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ying-Yan He
- Department of Cardiology, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-Lin Liu
- Department of Cardiology, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yong-Quan Hua
- Department of Cardiology, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Li-Rong Li
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yin Huang
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cai-Wen Ou
- Department of Cardiology, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cheng-Feng Luo
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min-Sheng Chen
- Department of Cardiology, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Yanaka A. Role of NRF2 in protection of the gastrointestinal tract against oxidative stress. J Clin Biochem Nutr 2018; 63:18-25. [PMID: 30087539 PMCID: PMC6064821 DOI: 10.3164/jcbn.17-139] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/21/2018] [Indexed: 12/17/2022] Open
Abstract
The gastrointestinal tract is exposed to a variety of noxious factors, such as Helicobacter pylori, nonsteroidal anti-inflammatory drugs, gastric acid, ischemia-reperfusion, and mental stresses. Theses stressors generate free radicals within gastrointestinal tissues, causing organ injury and functional disturbance. Although the gastrointestinal tract can withstand such oxidative stresses to some extent by enhancing its antioxidant system via nuclear factor erythroid 2-related factor 2-Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1-mediated pathways, acute or chronic exposure to oxidative stress can cause several gastrointestinal tract disorders, such as inflammation, ulcers, cancers, and various functional disturbances. Recent studies have demonstrated that some natural compounds and drugs can upregulate the nuclear factor erythroid 2-related factor 2-mediated antioxidant system, ameliorating or preventing these disorders. Although these compounds may be useful as chemopreventive agents, sufficient evidence for their clinical efficacy has not yet been provided. In addition, it is important to note that excessive nuclear factor erythroid 2-related factor 2 stimulation can be harmful to human health, especially from the standpoint of tumor biology.
Collapse
Affiliation(s)
- Akinori Yanaka
- Hitachi Medical Education and Research Center, University of Tsukuba Hospital, Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| |
Collapse
|
32
|
Yang N, Li S, Yan C, Sun R, He J, Xie Y, Peng Y, Wang G, Aa J. Inhibitory Effects of Endogenous Linoleic Acid and Glutaric Acid on the Renal Glucuronidation of Berberrubine in Mice and on Recombinant Human UGT1A7, 1A8, and 1A9. Mol Pharmacol 2018; 93:216-227. [PMID: 29351921 DOI: 10.1124/mol.117.110668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/08/2018] [Indexed: 12/22/2022] Open
Abstract
Berberrubine (BRB) has a strong lipid-lowering effect and can be extensively metabolized into berberrubine-9-O-β-d-glucuronide (BRBG) in vivo. Recently, pharmacokinetics studies showed that the production of BRBG was significantly decreased in the urine of mice fed with a high-fat diet (HFD), indicating a decreased glucuronidation capacity. Based on the UDP-glucuronosyltransferase (UGT) isoform identification, hepatic and renal microsomal incubation, glucuronidation was examined to suggest the metabolism of BRB in liver and kidneys. The results showed that the renal UGT activity for metabolizing BRB markedly decreased, which may be highly related to the decreased expression and activity of renal Ugt1a7c. Surprisingly, in vitro studies revealed neither BRB nor BRBG inhibited the renal UGT activity. By employing an integrated strategy of metabolomics and pharmacokinetics, we identified and confirmed for the first time the inhibitory effect of some potential endogenous molecules on the renal glucuronidation of C57BL/6J mice, such as glutaric acid (GA) and linoleic acid (LA). By employing recombinant human UGTs, we found that GA and LA efficiently affect the activity of recombinant human UGT1A7, 1A9, and 1A8 at their normal or abnormal physiologic levels in vivo. GA (2 mM) markedly inhibited the activity of UGT1A7 by 89.4% and UGT1A9 by 32.8%. The inhibition rates reached 99.3% for UGT1A9, 48.3% for UGT1A7, and 46.8% for UGT1A8 with LA at 200 μM. It has been suggested that the endogenous molecules have the potential to affect the efficiency of glucuronidation, which might be a key factor contributing to individual differences in drug metabolism.
Collapse
Affiliation(s)
- Na Yang
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Sijia Li
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Caixia Yan
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Runbin Sun
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jun He
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yuan Xie
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ying Peng
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Guangji Wang
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jiye Aa
- Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
33
|
Induction of xenobiotic-metabolizing enzymes in hepatocytes by beta-naphthoflavone: Time-dependent changes in activities, protein and mRNA levels. ACTA PHARMACEUTICA 2018; 68:75-85. [PMID: 29453911 DOI: 10.2478/acph-2018-0005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/02/2017] [Indexed: 12/11/2022]
Abstract
In the present study, time-dependency of the induction effect of a selective inducer on the activity, protein and mRNA levels of cytochromes P450 1A1/2 (CYP1A1/2), NAD(P)H:quinone oxidoreductase 1 (NQO1) and glutathione S-transferases (GSTA), in primary culture of rat hepatocytes was tested and evaluated. To show the differences in responses of tested enzymes, the common aryl hydrocarbon receptor (AhR) ligand agonist, beta-naphthoflavone (BNF), was used. Induction of CYP1A1/2 by BNF was detected at all time intervals and at all levels (i.e., mRNA, protein, enzyme activity). Different responses of NQO1 and GSTA upon BNF treatment were observed. Our results demonstrate that the responses of different xenobiotic-metabolizing enzymes to the inducer vary in time and depend on the measured parameter. For these reasons, an induction study featuring only one-time interval treatment and/ or one parameter testing could produce misleading information.
Collapse
|
34
|
Dou L, Poitevin S, Sallée M, Addi T, Gondouin B, McKay N, Denison MS, Jourde-Chiche N, Duval-Sabatier A, Cerini C, Brunet P, Dignat-George F, Burtey S. Aryl hydrocarbon receptor is activated in patients and mice with chronic kidney disease. Kidney Int 2018; 93:986-999. [PMID: 29395338 DOI: 10.1016/j.kint.2017.11.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 10/18/2017] [Accepted: 11/09/2017] [Indexed: 11/25/2022]
Abstract
Patients with chronic kidney disease (CKD) are exposed to uremic toxins and have an increased risk of cardiovascular disease. Some uremic toxins, like indoxyl sulfate, are agonists of the transcription factor aryl hydrocarbon receptor (AHR). These toxins induce a vascular procoagulant phenotype. Here we investigated AHR activation in patients with CKD and in a murine model of CKD. We performed a prospective study in 116 patients with CKD stage 3 to 5D and measured the AHR-Activating Potential of serum by bioassay. Compared to sera from healthy controls, sera from CKD patients displayed a strong AHR-Activating Potential; strongly correlated with eGFR and with the indoxyl sulfate concentration. The expression of the AHR target genes Cyp1A1 and AHRR was up-regulated in whole blood from patients with CKD. Survival analyses revealed that cardiovascular events were more frequent in CKD patients with an AHR-Activating Potential above the median. In mice with 5/6 nephrectomy, there was an increased serum AHR-Activating Potential, and an induction of Cyp1a1 mRNA in the aorta and heart, absent in AhR-/- CKD mice. After serial indoxyl sulfate injections, we observed an increase in serum AHR-AP and in expression of Cyp1a1 mRNA in aorta and heart in WT mice, but not in AhR-/- mice. Thus, the AHR pathway is activated both in patients and mice with CKD. Hence, AHR activation could be a key mechanism involved in the deleterious cardiovascular effects observed in CKD.
Collapse
Affiliation(s)
- Laetitia Dou
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France
| | - Stéphane Poitevin
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France
| | - Marion Sallée
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France; Centre de Néphrologie et Transplantation Rénale, AP-HM, Marseille, France
| | - Tawfik Addi
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France
| | - Bertrand Gondouin
- Centre de Néphrologie et Transplantation Rénale, AP-HM, Marseille, France
| | - Nathalie McKay
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France
| | - Michael S Denison
- Department of Environmental Toxicology, University of California, Davis, California, USA
| | - Noémie Jourde-Chiche
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France; Centre de Néphrologie et Transplantation Rénale, AP-HM, Marseille, France
| | - Ariane Duval-Sabatier
- Centre de Néphrologie et Transplantation Rénale, AP-HM, Marseille, France; Association des dialysés Provence-Corse, Marseille, France
| | - Claire Cerini
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France
| | - Philippe Brunet
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France; Centre de Néphrologie et Transplantation Rénale, AP-HM, Marseille, France
| | | | - Stéphane Burtey
- Aix-Marseille University, INSERM, UMR-S 1076, VRCM, Marseille, France; Centre de Néphrologie et Transplantation Rénale, AP-HM, Marseille, France.
| |
Collapse
|
35
|
Lněničková K, Dymáková A, Szotáková B, Boušová I. Sulforaphane Alters β-Naphthoflavone-Induced Changes in Activity and Expression of Drug-Metabolizing Enzymes in Rat Hepatocytes. Molecules 2017; 22:molecules22111983. [PMID: 29144397 PMCID: PMC6150368 DOI: 10.3390/molecules22111983] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022] Open
Abstract
Sulforaphane (SFN), an isothiocyanate found in cruciferous vegetables, exerts many beneficial effects on human health such as antioxidant, anti-inflammatory, and anticancer effects. The effect of SFN alone on drug-metabolizing enzymes (DMEs) has been investigated in numerous in vitro and in vivo models, but little is known about the effect of SFN in combination with cytochrome P450 (CYP) inducer. The aim of our study was to evaluate the effect of SFN on the activity and gene expression of selected DMEs in primary cultures of rat hepatocytes treated or non-treated with β-naphthoflavone (BNF), the model CYP1A inducer. In our study, SFN alone did not significantly alter the activity and expression of the studied DMEs, except for the glutathione S-transferase (GSTA1) mRNA level, which was significantly enhanced. Co-treatment of hepatocytes with SFN and BNF led to a substantial increase in sulfotransferase, aldoketoreductase 1C, carbonylreductase 1 and NAD(P)H:quinone oxidoreductase 1 activity and a marked decrease in cytochrome P450 (CYP) Cyp1a1, Cyp2b and Cyp3a4 expression in comparison to the treatment with BNF alone. Sulforaphane is able to modulate the activity and/or expression of DMEs, thus shifting the balance of carcinogen metabolism toward deactivation, which could represent an important mechanism of its chemopreventive activity.
Collapse
Affiliation(s)
- Kateřina Lněničková
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Heyrovského 1203, 50005 Hradec Králové, Czech Republic.
| | - Andrea Dymáková
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Heyrovského 1203, 50005 Hradec Králové, Czech Republic.
| | - Barbora Szotáková
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Heyrovského 1203, 50005 Hradec Králové, Czech Republic.
| | - Iva Boušová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Heyrovského 1203, 50005 Hradec Králové, Czech Republic.
| |
Collapse
|
36
|
Bockor L, Bortolussi G, Vodret S, Iaconcig A, Jašprová J, Zelenka J, Vitek L, Tiribelli C, Muro AF. Modulation of bilirubin neurotoxicity by the Abcb1 transporter in the Ugt1-/- lethal mouse model of neonatal hyperbilirubinemia. Hum Mol Genet 2017; 26:145-157. [PMID: 28025333 DOI: 10.1093/hmg/ddw375] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/27/2016] [Indexed: 01/20/2023] Open
Abstract
Moderate neonatal jaundice is the most common clinical condition during newborn life. However, a combination of factors may result in acute hyperbilirubinemia, placing infants at risk of developing bilirubin encephalopathy and death by kernicterus. While most risk factors are known, the mechanisms acting to reduce susceptibility to bilirubin neurotoxicity remain unclear. The presence of modifier genes modulating the risk of developing bilirubin-induced brain damage is increasingly being recognised. The Abcb1 and Abcc1 members of the ABC family of transporters have been suggested to have an active role in exporting unconjugated bilirubin from the central nervous system into plasma. However, their role in reducing the risk of developing neurological damage and death during neonatal development is still unknown.To this end, we mated Abcb1a/b-/- and Abcc1-/- strains with Ugt1-/- mice, which develop severe neonatal hyperbilirubinemia. While about 60% of Ugt1-/- mice survived after temporary phototherapy, all Abcb1a/b-/-/Ugt1-/- mice died before postnatal day 21, showing higher cerebellar levels of unconjugated bilirubin. Interestingly, Abcc1 role appeared to be less important.In the cerebellum of Ugt1-/- mice, hyperbilirubinemia induced the expression of Car and Pxr nuclear receptors, known regulators of genes involved in the genotoxic response.We demonstrated a critical role of Abcb1 in protecting the cerebellum from bilirubin toxicity during neonatal development, the most clinically relevant phase for human babies, providing further understanding of the mechanisms regulating bilirubin neurotoxicity in vivo. Pharmacological treatments aimed to increase Abcb1 and Abcc1 expression, could represent a therapeutic option to reduce the risk of bilirubin neurotoxicity.
Collapse
Affiliation(s)
- Luka Bockor
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Iaconcig
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Jana Jašprová
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 3, 166 28 Prague, Czech Republic
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Claudio Tiribelli
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza Trieste, Italy and
| | - Andrés F Muro
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| |
Collapse
|
37
|
Saez-Lopez C, Brianso-Llort L, Torres-Torronteras J, Simó R, Hammond GL, Selva DM. Resveratrol Increases Hepatic SHBG Expression through Human Constitutive Androstane Receptor: a new Contribution to the French Paradox. Sci Rep 2017; 7:12284. [PMID: 28947831 PMCID: PMC5612985 DOI: 10.1038/s41598-017-12509-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 09/12/2017] [Indexed: 01/08/2023] Open
Abstract
Sex hormone-binding globulin (SHBG) carries sex steroids in blood regulating their bioavailability. Red wine consumption increases plasma SHBG levels, and we have discovered that resveratrol, a polyphenol enriched in red wine, acts specifically through the human constitutive androstane receptor (CAR), a drug/xenobiotic detoxification gene regulator, to increase hepatic SHBG production. Chromatin immunoprecipitation and luciferase reporter gene assays show that human CAR binds to a typical direct repeat 1 nuclear hormone receptor-binding element in the human SHBG proximal promoter. Resveratrol also increased hepatic SHBG production in humanized SHBG/CAR transgenic mice. Moreover, SHBG expression correlated significantly with CAR mRNA levels in human liver biopsies. We conclude that the beneficial effects of red wine on the metabolic syndrome and it associated co-morbidities, including cardiovascular disease and type 2 diabetes, may be mediated in part by resveratrol acting via CAR to increase plasma SHBG levels.
Collapse
Affiliation(s)
- Cristina Saez-Lopez
- Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona and Biomedical Network Research Centre on Diabetes and Metabolic Diseases (CIBERDEM, ISCIII), Barcelona, Spain
| | - Laura Brianso-Llort
- Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona and Biomedical Network Research Centre on Diabetes and Metabolic Diseases (CIBERDEM, ISCIII), Barcelona, Spain
| | - J Torres-Torronteras
- Research Group on Neuromuscular and Mitochondrial Diseases, Vall Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona and Biomedical Network Research Centre on Rare Diseases (CIBERER, ISCIII), Barcelona, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona and Biomedical Network Research Centre on Diabetes and Metabolic Diseases (CIBERDEM, ISCIII), Barcelona, Spain.
| | - Geoffrey L Hammond
- Cellular & Physiological Sciences, University of British Columbia, Vancouver, Canada.
| | - David M Selva
- Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona and Biomedical Network Research Centre on Diabetes and Metabolic Diseases (CIBERDEM, ISCIII), Barcelona, Spain.
| |
Collapse
|
38
|
Blunder S, Kõks S, Kõks G, Reimann E, Hackl H, Gruber R, Moosbrugger-Martinz V, Schmuth M, Dubrac S. Enhanced Expression of Genes Related to Xenobiotic Metabolism in the Skin of Patients with Atopic Dermatitis but Not with Ichthyosis Vulgaris. J Invest Dermatol 2017; 138:98-108. [PMID: 28899689 DOI: 10.1016/j.jid.2017.08.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/15/2022]
Abstract
Previous transcriptome analyses underscored the importance of immunological and skin barrier abnormalities in atopic dermatitis (AD). We sought to identify pathogenic pathways involved in AD by comparing the transcriptomes of AD patients stratified for filaggrin (FLG)-null mutations to those of both healthy donors and patients with ichthyosis vulgaris. We applied RNA sequencing to analyze the whole transcriptome of nonlesional skin. We found that 607 genes (476 up-regulated and 131 down-regulated by >2-fold) and 193 genes (172 up-regulated and 21 down-regulated by >2-fold) were differentially expressed when all AD or ichthyosis vulgaris patients were compared with healthy donors, respectively. Expression of genes involved in RNA/protein turnover and adenosine triphosphate synthesis, as well as genes involved in cell death, response to oxidative stress, DNA damage/repair, and autophagy, were significantly enriched in AD skin and, to a lesser extent, in ichthyosis vulgaris skin. FLG-null mutations appear to hardly interfere with current observations. Genes related to xenobiotic metabolism were up-regulated in AD skin only, as were genes related to arachidonic, linoleic, and α-linolenic acid metabolism. Thus, this work newly links AD pathogenesis to aberrant expression of genes related to xenobiotic metabolism.
Collapse
Affiliation(s)
- Stefan Blunder
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sulev Kõks
- Department of Pathophysiology, Center of Translational Medicine, University of Tartu, Tartu, Estonia
| | - Gea Kõks
- Department of Pathophysiology, Center of Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ene Reimann
- Department of Pathophysiology, Center of Translational Medicine, University of Tartu, Tartu, Estonia
| | - Hubert Hackl
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria; Department of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Moosbrugger-Martinz
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Schmuth
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
39
|
Kalthoff S, Landerer S, Reich J, Strassburg CP. Protective effects of coffee against oxidative stress induced by the tobacco carcinogen benzo[α]pyrene. Free Radic Biol Med 2017; 108:66-76. [PMID: 28300668 DOI: 10.1016/j.freeradbiomed.2017.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 01/18/2023]
Abstract
AIMS Coffee consumption has been epidemiologically associated with a lower risk for liver cirrhosis and cancer. UDP-glucuronosyltransferases (UGT1A) catalyze the detoxification of reactive metabolites thereby acting as indirect antioxidants. Aim of the study was to examine UGT1A regulation in response to Benzo[α]pyrene (BaP) to elucidate the potentially protective effects of coffee on BaP-induced oxidative stress and toxicity. RESULTS In cell culture (HepG2, KYSE70 cells) and in htgUGT1A-WT mice, UGT1A transcription was activated by BaP, while it was reduced or absent htgUGT1A-SNP (containing 10 commonly occurring UGT1A-SNPs) mice. siRNA-mediated knockdown identified aryl hydrocarbon receptor (AhR) and nuclear factor erythroid2-related factor-2 (Nrf2) as mediators of BaP-induced UGT1A upregulation. Exposure to coffee led to a reduction of BaP-induced production of reactive oxygen species in vitro and in htgUGT1A-WT and -SNP mice. After UGT1A silencing by UGT1A-specific siRNA in cell culture, the coffee-mediated reduction of ROS production was significantly impaired compared to UGT1A expressing cells. CONCLUSION A common UGT1A haplotype, prevalent in 9% (homozygous) of the White population, significantly impairs the expression of UGT1A enzymes in response to the putative tobacco carcinogen BaP and is likely to represent a significant risk factor for reduced detoxification and increased genotoxicity. Coffee was demonstrated to inhibit BaP-induced production of oxidative stress by UGT1A activation, and is therefore an attractive candidate for chemoprotection in risk groups for HCC or other tumors.
Collapse
Affiliation(s)
- Sandra Kalthoff
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany.
| | - Steffen Landerer
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany.
| | - Julia Reich
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany.
| | | |
Collapse
|
40
|
Yang N, Sun R, Liao X, Aa J, Wang G. UDP-glucuronosyltransferases (UGTs) and their related metabolic cross-talk with internal homeostasis: A systematic review of UGT isoforms for precision medicine. Pharmacol Res 2017; 121:169-183. [PMID: 28479371 DOI: 10.1016/j.phrs.2017.05.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) are the primary phase II enzymes catalyzing the conjugation of glucuronic acid to the xenobiotics with polar groups for facilitating their clearance. The UGTs belong to a superfamily that consists of diverse isoforms possessing distinct but overlapping metabolic activity. The abnormality or deficiency of UGTs in vivo is highly associated with some diseases, efficacy and toxicity of drugs, and precisely therapeutic personality. Despite the great effects and fruitful results achieved, to date, the expression and functions of individual UGTs have not been well clarified, the inconsistency of UGTs is often observed in human and experimental animals, and the complex regulation factors affecting UGTs have not been systematically summarized. This article gives an overview of updated reports on UGTs involving the various regulatory factors in terms of the genetic, environmental, pathological, and physiological effects on the functioning of individual UGTs, in turn, the dysfunction of UGTs induced disease risk and endo- or xenobiotic metabolism-related toxicity. The complex cross-talk effect of UGTs with internal homeostasis is systematically summarized and discussed in detail, which would be of great importance for personalized precision medicine.
Collapse
Affiliation(s)
- Na Yang
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Runbin Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoying Liao
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
41
|
Kurita A, Miyauchi Y, Ikushiro S, Mackenzie PI, Yamada H, Ishii Y. Comprehensive Characterization of Mouse UDP-Glucuronosyltransferase (Ugt) Belonging to the Ugt2b Subfamily: Identification of Ugt2b36 as the Predominant Isoform Involved in Morphine Glucuronidation. J Pharmacol Exp Ther 2017; 361:199-208. [PMID: 28228532 DOI: 10.1124/jpet.117.240382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/14/2017] [Indexed: 12/18/2022] Open
Abstract
UDP-Glucuronosyltransferases (UGTs) are classified into three subfamilies in mice: Ugt1a, 2b, and 2a. In the Ugt1a subfamily, Ugt1a1 and 1a6 appear to correspond to human UGT1A1 and 1A6 The mouse is an important animal for its use in investigations, but the substrate specificities of Ugt isoforms belonging to the 2b subfamily in mice remain largely unknown. To address this issue, we characterized the substrate specificity of all isoforms of the Ugt2b subfamily expressed in the mouse liver. The cDNAs of Ugt1a1, Ugt2a3, and all the Ugt2b isoforms expressed in the liver were reverse-transcribed from the total RNA of male FVB-mouse livers and then amplified. A baculovirus-Sf9 cell system for expressing each Ugt was established. Of all the Ugts examined, Ugt2b34, 2b36, and 2b37 exhibited the ability to glucuronidate morphine with Ugt2b36, the most active in this regard. Ugt1a1, but also Ugt2b34, 2b36, and 2b37 to a lesser extent, preferentially catalyzed the glucuronidation of 17β-estradiol on the 3-hydroxyl group (E3G). With these isoforms, E3G formation by Ugt1a1 was efficient; however, Ugt2b5 exhibited a preference for the 17β-hydroxyl group (E17G). Ugt2b1 and Ugt2a3 formed comparable levels of E3G and E17G. Ugt2b1 and 2b5 were the only isoforms involved in chloramphenicol glucuronidation. As Ugt2b36 is highly expressed in the liver, it is most likely that Ugt2b36 is a major morphine Ugt in mouse liver. Regarding E3G formation, Ugt1a1, like the human homolog, seems to play an important role in the liver.
Collapse
Affiliation(s)
- Ayumi Kurita
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (A.K., Y.M., H.Y., Y.I.), Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan (S.I.), and Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA, Australia (P.I.M.)
| | - Yuu Miyauchi
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (A.K., Y.M., H.Y., Y.I.), Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan (S.I.), and Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA, Australia (P.I.M.)
| | - Shin'ichi Ikushiro
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (A.K., Y.M., H.Y., Y.I.), Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan (S.I.), and Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA, Australia (P.I.M.)
| | - Peter I Mackenzie
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (A.K., Y.M., H.Y., Y.I.), Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan (S.I.), and Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA, Australia (P.I.M.)
| | - Hideyuki Yamada
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (A.K., Y.M., H.Y., Y.I.), Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan (S.I.), and Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA, Australia (P.I.M.)
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (A.K., Y.M., H.Y., Y.I.), Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan (S.I.), and Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, SA, Australia (P.I.M.)
| |
Collapse
|
42
|
Jilek JL, Tian Y, Yu AM. Effects of MicroRNA-34a on the Pharmacokinetics of Cytochrome P450 Probe Drugs in Mice. Drug Metab Dispos 2017; 45:512-522. [PMID: 28254952 PMCID: PMC5399649 DOI: 10.1124/dmd.116.074344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 03/01/2017] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs or miRs), including miR-34a, have been shown to regulate nuclear receptor, drug-metabolizing enzyme, and transporter gene expression in various cell model systems. However, to what degree miRNAs affect pharmacokinetics (PK) at the systemic level remains unknown. In addition, miR-34a replacement therapy represents a new cancer treatment strategy, although it is unknown whether miR-34a therapeutic agents could elicit any drug-drug interactions. To address this question, we refined a practical single-mouse PK approach and investigated the effects of a bioengineered miR-34a agent on the PK of several cytochrome P450 probe drugs (midazolam, dextromethorphan, phenacetin, diclofenac, and chlorzoxazone) administered as a cocktail. This approach involves manual serial blood microsampling from a single mouse and requires a sensitive liquid chromatography-tandem mass spectrometry assay, which was able to illustrate the sharp changes in midazolam PK by ketoconazole and pregnenolone 16α-carbonitrile as well as phenacetin PK by α-naphthoflavone and 3-methylcholanthrene. Surprisingly, 3-methylcholanthrene also decreased systemic exposure to midazolam, whereas both pregnenolone 16α-carbonitrile and 3-methylcholanthrene largely reduced the exposure to dextromethorphan, diclofenac, and chlorzoxazone. Finally, the biologic miR-34a agent had no significant effects on the PK of cocktail drugs but caused a marginal (45%-48%) increase in systemic exposure to midazolam, phenacetin, and dextromethorphan in mice. In vitro validation of these data suggested that miR-34a slightly attenuated intrinsic clearance of dextromethorphan. These findings from single-mouse PK and corresponding mouse liver microsome models suggest that miR-34a might have minor or no effects on the PK of coadministered cytochrome P450-metabolized drugs.
Collapse
Affiliation(s)
- Joseph L Jilek
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California (J.L.J., Y.T., A.-M.Y.); and Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China (Y.T.)
| | - Ye Tian
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California (J.L.J., Y.T., A.-M.Y.); and Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China (Y.T.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California (J.L.J., Y.T., A.-M.Y.); and Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China (Y.T.)
| |
Collapse
|
43
|
Hydroethanolic extract of Baccharis trimera ameliorates alcoholic fatty liver disease in mice. Chem Biol Interact 2016; 260:22-32. [DOI: 10.1016/j.cbi.2016.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/09/2016] [Accepted: 10/03/2016] [Indexed: 12/20/2022]
|
44
|
Narayanapillai SC, Lin SH, Leitzman P, Upadhyaya P, Baglole CJ, Xing C. Dihydromethysticin (DHM) Blocks Tobacco Carcinogen 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-Induced O 6-Methylguanine in a Manner Independent of the Aryl Hydrocarbon Receptor (AhR) Pathway in C57BL/6 Female Mice. Chem Res Toxicol 2016; 29:1828-1834. [PMID: 27728767 PMCID: PMC6532060 DOI: 10.1021/acs.chemrestox.6b00203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is a key carcinogen responsible for tobacco smoke-induced lung carcinogenesis. Among the types of DNA damage caused by NNK and its metabolite, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), O6-methylguanine (O6-mG) is likely the most carcinogen in A/J mice. Results of our previous studies showed that levels of O6-mG and other types of NNAL-derived DNA damage were preferentially reduced in the lung of female A/J mice upon dietary treatment with dihydromethysticin (DHM), a promising lung cancer chemopreventive agent from kava. Such a differential blockage may be mediated via an increased level of NNAL glucuronidation, thereby leading to its detoxification. The potential of the aryl hydrocarbon receptor (AhR) as an upstream target of DHM mediating these events was evaluated herein using Ahr+/- and Ahr-/- C57BL/6 female mice because DHM was reported as an AhR agonist. DHM (0.05, 0.2, and 1.0 mg/g of diet) and dihydrokavain (DHK, an inactive analogue, 1.0 mg/g of diet) were given to mice for 7 days, followed by a single intraperitoneal dose of NNK at 100 mg/kg of body weight. The effects of DHM on the amount of O6-mG in the lung, on the urinary ratio of glucuronidated NNAL (NNAL-Gluc) and free NNAL, and on CYP1A1/2 activity in the liver microsomes were analyzed. As observed in A/J mice, DHM treatment significantly and dose-dependently reduced the level of O6-mG in the target lung tissue, but there were no significant differences in O6-mG reduction between mice from Ahr+/- and Ahr-/- backgrounds. Similarly, in both strains, DHM at 1 mg/g of diet significantly increased the urinary ratio of NNAL-Gluc to free NNAL and CYP1A1/2 enzymatic activity in liver with no changes detected at lower DHM dosages. Because none of these effects of DHM were dependent on Ahr status, AhR clearly is not the upstream target for DHM.
Collapse
Affiliation(s)
- Sreekanth C. Narayanapillai
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Shang-Hsuan Lin
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Pablo Leitzman
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Pramod Upadhyaya
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carolyn J. Baglole
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
45
|
Vrzal R, Illes P, Dvorak Z. Transplant drugs affect the expression of phase II and antioxidant enzymes in human carcinoma cells HepG2 but not in primary cultures of human hepatocytes: In vitro comparative study. Pharmacol Rep 2016; 68:1008-14. [DOI: 10.1016/j.pharep.2016.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/02/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
|
46
|
Shi R, Wu J, Meng C, Ma B, Wang T, Li Y, Ma Y. Cyp3a11-mediated testosterone-6β-hydroxylation decreased, while UGT1a9-mediated propofol O-glucuronidation increased, in mice with diabetes mellitus. Biopharm Drug Dispos 2016; 37:433-443. [PMID: 27514509 DOI: 10.1002/bdd.2027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 08/04/2016] [Accepted: 08/07/2016] [Indexed: 12/20/2022]
Abstract
The db/db mouse is one of the most popular animal models for type 2 diabetes mellitus, but changes in the activities of important P450s and UGTs are still not completely clear. This study was designed to investigate the alterations of major hepatic cytochrome P450s and UDP-glucuronyltransferase enzymes in db/db mice. Mouse liver microsomes (MLMs) were obtained from male db/db mice and their wild type littermates. After incubation of the substrates separately with MLMs, the samples were pooled and analysed by high-throughput liquid chromatography-tandem mass spectrometry system for the simultaneous study of nine phase I metabolic reactions and three glucuronidation conjugation reactions to determine the activity of the metabolic enzymes. Compared with normal controls, the Clint estimate for testosterone-6β-hydroxylation was lower (46%) (p < 0.05), while the Vmax and Clint estimates for propofol O-glucuronidation were 5-fold higher (p < 0.01) in the liver microsomes from db/db mice. There was no significant difference in phase I metabolic reactions of phenacetin-O-deethylation, coumarin-7-hydroxylation, bupropion-hydroxylation, omeprazole-5-hydroxylation, dextromethorphan-O-demethylation, tolbutamide-4-hydroxylation, chlorzoxazone-6-hydroxylation and midazolam-1-hydroxylation and in glucuronidation reactions of estradiol 3-O-glucuronidation, and 3-azido-3-deoxythymidine glucuronidation. The data suggest that, in db/db mice, the activity of Cyp3a11, catalysing testosterone-6β-hydroxylation, decreased, while the activity of UGT1a9, catalysing propofol O-glucuronidation, increased. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rong Shi
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiasheng Wu
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cong Meng
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bingliang Ma
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianming Wang
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueming Ma
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
47
|
Park S, Cheng SL, Cui JY. Characterizing drug-metabolizing enzymes and transporters that are bona fide CAR-target genes in mouse intestine. Acta Pharm Sin B 2016; 6:475-491. [PMID: 27709017 PMCID: PMC5045557 DOI: 10.1016/j.apsb.2016.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 04/29/2016] [Accepted: 05/20/2016] [Indexed: 12/26/2022] Open
Abstract
Intestine is responsible for the biotransformation of many orally-exposed chemicals. The constitutive androstane receptor (CAR/Nr1i3) is known to up-regulate many genes encoding drug-metabolizing enzymes and transporters (drug-processing genes/DPGs) in liver, but less is known regarding its effect in intestine. Sixty-day-old wild-type and Car−/− mice were administered the CAR-ligand TCPOBOP or vehicle once daily for 4 days. In wild-type mice, Car mRNA was down-regulated by TCPOBOP in liver and duodenum. Car−/− mice had altered basal intestinal expression of many DPGs in a section-specific manner. Consistent with the liver data (Aleksunes and Klaassen, 2012), TCPOBOP up-regulated many DPGs (Cyp2b10, Cyp3a11, Aldh1a1, Aldh1a7, Gsta1, Gsta4, Gstm1-m4, Gstt1, Ugt1a1, Ugt2b34, Ugt2b36, and Mrp2–4) in specific sections of small intestine in a CAR-dependent manner. However, the mRNAs of Nqo1 and Papss2 were previously known to be up-regulated by TCPOBOP in liver but were not altered in intestine. Interestingly, many known CAR-target genes were highest expressed in colon where CAR is minimally expressed, suggesting that additional regulators are involved in regulating their expression. In conclusion, CAR regulates the basal expression of many DPGs in intestine, and although many hepatic CAR-targeted DPGs were bona fide CAR-targets in intestine, pharmacological activation of CAR in liver and intestine are not identical.
Collapse
Key Words
- Aldh, aldehyde dehydrogenase
- Asbt, solute carrier family 10, member 2 (apical sodium/bile acid cotransporter)
- CAR
- CAR, constitutive androstane receptor
- CITCO, 6-(4-chlorophenyl)imidazo [2,1-b](1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
- Cq, quantification cycle
- Cyp, cytochrome P450
- DPGs, drug-processing genes (genes that encodes drug metabolizing enzymes or transporters)
- Drug-metabolizing enzymes
- Drug-processing genes
- Gst, glutathione S-trasnferase
- H3, Histone 3
- HRP, horseradish peroxidase
- Intestine
- Mice
- Mrp, multi-drug resistance-associated protein (ABC transporter family C member)
- Nqo1, NAD(P)H dehydrogenase quinone 1
- Nrf2, nuclear factor erythroid 2-related factor 2
- Oatp, organic anion transporting polypeptide (solute carrier organic anion transporter family member)
- PBS, phosphate-buffered saline
- PBST, phosphate-buffered saline with 0.05% tween 20
- PPARα, peroxisome proliferator activated receptor alpha
- PVDF, polyvinylidene difluoride
- Papss2, 3ʹ-phosphoadenosine 5ʹ-phosphosulfate synthase 2
- ST buffer, sucrose Tris buffer
- Sult, sulfotransferase
- TCPOBOP, 3,3ʹ,5,5ʹ-tetrachloro-1,4-bis(pyridyloxy)benzene
- Transporters
- Ugt, UDP glucuronosyltransferase
- WT, wild-type
- cDNA, complementary DNA
- ddCq, delta delta Cq
- hCAR, human constitutive androstane receptor
- qPCR, quantitative polymerase chain reaction
Collapse
|
48
|
Hirashima R, Michimae H, Takemoto H, Sasaki A, Kobayashi Y, Itoh T, Tukey RH, Fujiwara R. Induction of the UDP-Glucuronosyltransferase 1A1 during the Perinatal Period Can Cause Neurodevelopmental Toxicity. Mol Pharmacol 2016; 90:265-74. [PMID: 27413119 PMCID: PMC4998668 DOI: 10.1124/mol.116.104174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/11/2016] [Indexed: 12/17/2022] Open
Abstract
Anticonvulsants can increase the risk of developing neurotoxicity in infants; however, the underlying mechanism has not been elucidated to date. Thyroxine [3,5,3',5'-l-tetraiodothyronine (T4)] plays crucial roles in the development of the central nervous system. In this study, we hypothesized that induction of UDP-glucuronosyltransferase 1A1 (UGT1A1)-an enzyme involved in the metabolism of T4-by anticonvulsants would reduce serum T4 levels and cause neurodevelopmental toxicity. Exposure of mice to phenytoin during both the prenatal and postnatal periods significantly induced UGT1A1 and decreased serum T4 levels on postnatal day 14. In the phenytoin-treated mice, the mRNA levels of synaptophysin and synapsin I in the hippocampus were lower than those in the control mice. The thickness of the external granule cell layer was greater in phenytoin-treated mice, indicating that induction of UGT1A1 during the perinatal period caused neurodevelopmental disorders. Exposure to phenytoin during only the postnatal period also caused these neurodevelopmental disorders. A T4 replacement attenuated the increase in thickness of the external granule cell layer, indicating that the reduced T4 was specifically associated with the phenytoin-induced neurodevelopmental disorder. In addition, these neurodevelopmental disorders were also found in the carbamazepine- and pregnenolone-16-α-carbonitrile-treated mice. Our study is the first to indicate that UGT1A1 can control neurodevelopment by regulating serum T4 levels.
Collapse
Affiliation(s)
- Rika Hirashima
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Hirofumi Michimae
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Hiroaki Takemoto
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Aya Sasaki
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Yoshinori Kobayashi
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Tomoo Itoh
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Robert H Tukey
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Ryoichi Fujiwara
- Department of Pharmaceutics (R.H., A.S., T.I., R.F.), Division of Biostatistics (H.M.), and Department of Pharmacognosy (H.T., Y.K.), School of Pharmacy, Kitasato University, Tokyo, Japan; and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| |
Collapse
|
49
|
Bright AS, Herrera-Garcia G, Moscovitz JE, You D, Guo GL, Aleksunes LM. Regulation of Drug Disposition Gene Expression in Pregnant Mice with Car Receptor Activation. NUCLEAR RECEPTOR RESEARCH 2016; 3. [PMID: 27818994 DOI: 10.11131/2016/101193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
More than half of pregnant women use prescription medications in order to maintain both maternal and fetal health. The constitutive androstane receptor (Car) critically affects the disposition of chemicals by regulating the transcription of genes encoding metabolic enzymes and transporters. However, the effects of Car activation on chemical disposition during pregnancy are unclear. This study aims to determine the degree to which pregnancy alters the expression of drug metabolizing enzymes and transporters in response to the pharmacological activation of Car. To test this, pregnant C57BL/6 mice were administered IP doses of vehicle, or a potent Car agonist, TCPOBOP, on gestation days 14, 15 and 16. Hepatic mRNA and protein expression of Car target genes (phase I, II and transporters) were quantified on gestation day 17. Pregnancy-related changes, such as induction of Cyp2b10, Ugt1a1 and Sult1a1 and repression of Ugt1a6, Gsta1, Gsta2 and Mrp6, were observed. Interestingly, the induction of Cyp2b10, Gsta1, Gsta2 and Mrp2-4 mRNAs by TCPOBOP was attenuated in maternal livers suggesting that Car activation is impeded by the biochemical and/or physiological changes that occur during gestation. Taken together, these findings suggest that pregnancy and pharmacological activation of Car can differentially regulate the expression of drug metabolism and transport genes.
Collapse
Affiliation(s)
- Amanda S Bright
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Guadalupe Herrera-Garcia
- Department of Obstetrics and Gynecology, Rutgers-Robert Wood Johnson Medical School, 1 Robert Wood Johnson Place, New Brunswick, NJ 08901, USA
| | - Jamie E Moscovitz
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Dahea You
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| |
Collapse
|
50
|
Zhu L, Qu K, Xia B, Sun X, Chen B. Transcriptomic response to water accommodated fraction of crude oil exposure in the gill of Japanese flounder, Paralichthys olivaceus. MARINE POLLUTION BULLETIN 2016; 106:283-291. [PMID: 27001715 DOI: 10.1016/j.marpolbul.2015.12.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 06/05/2023]
Abstract
Illumina-based RNA-seq was used to determine the short-term transcriptomic responses of Paralichthys olivaceus gill to an environmentally relevant level of water accommodated fraction (WAF) of crude oil. 213,979 transcripts and 128,482 unigenes were obtained. Differential expression analysis revealed that 1641 and 2142 genes were significantly up- and down-regulated. Enrichment analysis identified a set of GO terms and putative pathways involved in the response of P. olivaceus to WAF exposure. Analysis of the transcripts revealed the effective protective mechanisms of P. olivaceus to reduce the toxic effects of WAF. Moreover, WAF exposure induced the metabolism of energy substrates, and downstream pathway genes were modified to provide protection against toxic damage. Transcripts analysis demonstrated that the genes involved in circadian rhythm signaling were regulated in gills of P. olivaceus exposed to WAF. These results provide insights into the mechanisms of WAF-induced toxicity in fishes and into the WAF-sensitive biomarkers in P. olivaceus.
Collapse
Affiliation(s)
- Lin Zhu
- Marine Fishery Environment and Bioremediation Laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Keming Qu
- Marine Fishery Environment and Bioremediation Laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Bin Xia
- Marine Fishery Environment and Bioremediation Laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266200, China
| | - Xuemei Sun
- Marine Fishery Environment and Bioremediation Laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Bijuan Chen
- Marine Fishery Environment and Bioremediation Laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China.
| |
Collapse
|