1
|
Alqahtani MA, El-Ghiaty MA, El-Mahrouk SR, El-Kadi AOS. Differential Modulatory Effects of Methylmercury (MeHg) on Ahr-regulated Genes in Extrahepatic Tissues of C57BL/6 Mice. Biol Trace Elem Res 2024; 202:5071-5080. [PMID: 38197905 DOI: 10.1007/s12011-023-04050-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024]
Abstract
Methylmercury (MeHg) and 2,3,7,8-tetrachlorodibenzodioxin (TCDD) are potent environmental pollutants implicated in the modulation of xenobiotic-metabolizing enzymes, particularly the cytochrome P450 1 family (CYP1) which is regulated by the aryl hydrocarbon receptor (AHR). However, the co-exposure to MeHg and TCDD raises concerns about their potential combined effects, necessitating thorough investigation. The primary objective of this study was to investigate the individual and combined effects of MeHg and TCDD on AHR-regulated CYP1 enzymes in mouse extrahepatic tissues. Therefore, C57BL/6 mice were administrated with MeHg (2.5 mg/kg) in the absence and presence of TCDD (15 μg/kg) for 6 and 24 h. The AHR-regulated CYP1 mRNA and protein expression levels were measured in the heart, lung, and kidney, using RT real-time PCR and western blot, respectively. Interestingly, treatment with MeHg exhibited mainly inhibitory effect, particularly, it decreased the basal level of Cyp1a1 and Cyp1a2 mRNA and protein, and that was more evident at the 24 h time point in kidney followed by heart. Similarly, when mice were co-exposed, MeHg was able to reduce the TCDD-induced Cyp1a1 and Cyp1a2 expression, however, MeHg potentiated kidney Cyp1b1 mRNA expression, opposing the observed change on its protein level. Also, MeHg induced antioxidant NAD(P)H:quinone oxidoreductase (NQO1) mRNA and protein in kidney, while heme-oxygenase (HO-1) mRNA was up-regulated in heart and kidney. In conclusion, this study reveals intricate interplay between MeHg and TCDD on AHR-regulated CYP1 enzymes, with interesting inhibitory effects observed that might be significant for procarcinogen metabolism. Varied responses across tissues highlight the potential implications for environmental health.
Collapse
Affiliation(s)
- Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, Edmonton, Alberta, T6G 2E1, Canada
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, Edmonton, Alberta, T6G 2E1, Canada
| | - Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, Edmonton, Alberta, T6G 2E1, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, Edmonton, Alberta, T6G 2E1, Canada.
| |
Collapse
|
2
|
Manuel RSJ, Rundquist A, Ambrogi M, Scharpf BR, Peterson NT, Sandhu JK, Chandrashekar S, Ridlon M, Crawford LK, Keil-Stietz KP, Peterson RE, Vezina CM. The aryl hydrocarbon receptor agonist ITE reduces inflammation and urinary dysfunction in a mouse model of autoimmune prostatitis. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:149-161. [PMID: 39308590 PMCID: PMC11411176 DOI: 10.62347/pegk4888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/26/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVES Prostate inflammation is linked to lower urinary tract dysfunction and is a key factor in chronic prostatitis/chronic pelvic pain syndrome. Autoimmunity was recently identified as a driver of prostate inflammation. Agonists of the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor, have been used to suppress autoimmunity in mouse models of colitis, rhinitis, and dermatitis, but whether AHR agonists suppress prostate autoimmunity has not been examined. Here, we test whether ITE (2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester), an AHR agonist, suppresses inflammation, allodynia, and urinary dysfunction in a mouse model of experimental autoimmune prostatitis (EAP). METHODS C57BL/6J adult male mice were immunized with rat prostate antigen to induce EAP or TiterMax Gold® adjuvant (uninflamed control). Mice were also treated with ITE (10 mg/kg/day IP) or DMSO (vehicle, 5 mg/kg/day IP) for 6 days. Using the Nanostring nCounter Inflammation Panel, we evaluated the impact of EAP and ITE on prostatic RNA abundance. We validated EAP and ITE-mediated changes in a subset of RNAs by RT-PCR and RNAScope in situ RNA detection. RESULTS EAP appeared to heighten histological inflammation in the dorsal prostate, induced tactile allodynia, and appeared to increase the frequency of non-voiding bladder contractions. ITE mitigated some actions of EAP. EAP changed abundance of 40 inflammation-related RNAs, while ITE changed abundance of 28 inflammation-related RNAs. We identified a cluster of RNAs for which ITE protected against EAP-induced changes in the abundance of H2-Ab1, S100a8, and S100a9. ITE also increased the abundance of the AHR-responsive Cyp1a1 RNA. CONCLUSIONS These findings support the hypothesis that ITE activates the AHR in the prostate and reduces autoimmune-mediated prostatitis in mice.
Collapse
Affiliation(s)
- Robbie SJ Manuel
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Allison Rundquist
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Marcela Ambrogi
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Brandon R Scharpf
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Nelson T Peterson
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Jaskiran K Sandhu
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
| | - Sneha Chandrashekar
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
| | - Monica Ridlon
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Latasha K Crawford
- Department of Pathological Sciences, University of Wisconsin Madison School of Veterinary MedicineMadison, WI, USA
| | - Kimberly P Keil-Stietz
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Richard E Peterson
- Division of Pharmaceutical Sciences, University of Wisconsin School of PharmacyMadison, WI, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| |
Collapse
|
3
|
Graelmann FJ, Gondorf F, Majlesain Y, Niemann B, Klepac K, Gosejacob D, Gottschalk M, Mayer M, Iriady I, Hatzfeld P, Lindenberg SK, Wunderling K, Thiele C, Abdullah Z, He W, Hiller K, Händler K, Beyer MD, Ulas T, Pfeifer A, Esser C, Weighardt H, Förster I, Reverte-Salisa L. Differential cell type-specific function of the aryl hydrocarbon receptor and its repressor in diet-induced obesity and fibrosis. Mol Metab 2024; 85:101963. [PMID: 38821174 PMCID: PMC11214421 DOI: 10.1016/j.molmet.2024.101963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/02/2024] Open
Abstract
OBJECTIVE The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor regulating xenobiotic responses as well as physiological metabolism. Dietary AhR ligands activate the AhR signaling axis, whereas AhR activation is negatively regulated by the AhR repressor (AhRR). While AhR-deficient mice are known to be resistant to diet-induced obesity (DIO), the influence of the AhRR on DIO has not been assessed so far. METHODS In this study, we analyzed AhRR-/- mice and mice with a conditional deletion of either AhRR or AhR in myeloid cells under conditions of DIO and after supplementation of dietary AhR ligands. Moreover, macrophage metabolism was assessed using Seahorse Mito Stress Test and ROS assays as well as transcriptomic analysis. RESULTS We demonstrate that global AhRR deficiency leads to a robust, but not as profound protection from DIO and hepatosteatosis as AhR deficiency. Under conditions of DIO, AhRR-/- mice did not accumulate TCA cycle intermediates in the circulation in contrast to wild-type (WT) mice, indicating protection from metabolic dysfunction. This effect could be mimicked by dietary supplementation of AhR ligands in WT mice. Because of the predominant expression of the AhRR in myeloid cells, AhRR-deficient macrophages were analyzed for changes in metabolism and showed major metabolic alterations regarding oxidative phosphorylation and mitochondrial activity. Unbiased transcriptomic analysis revealed increased expression of genes involved in de novo lipogenesis and mitochondrial biogenesis. Mice with a genetic deficiency of the AhRR in myeloid cells did not show alterations in weight gain after high fat diet (HFD) but demonstrated ameliorated liver damage compared to control mice. Further, deficiency of the AhR in myeloid cells also did not affect weight gain but led to enhanced liver damage and adipose tissue fibrosis compared to controls. CONCLUSIONS AhRR-deficient mice are resistant to diet-induced metabolic syndrome. Although conditional ablation of either the AhR or AhRR in myeloid cells did not recapitulate the phenotype of the global knockout, our findings suggest that enhanced AhR signaling in myeloid cells deficient for AhRR protects from diet-induced liver damage and fibrosis, whereas myeloid cell-specific AhR deficiency is detrimental.
Collapse
Affiliation(s)
- Frederike J Graelmann
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Fabian Gondorf
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Yasmin Majlesain
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Birte Niemann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Germany
| | - Katarina Klepac
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Germany
| | - Dominic Gosejacob
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Germany
| | - Marlene Gottschalk
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Michelle Mayer
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Irina Iriady
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Philip Hatzfeld
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Sophie K Lindenberg
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Klaus Wunderling
- Biochemistry & Cell Biology of Lipids, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Christoph Thiele
- Biochemistry & Cell Biology of Lipids, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Zeinab Abdullah
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, University of Bonn, Germany
| | - Wei He
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Karsten Hiller
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Kristian Händler
- PRECISE Platform for Single cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn and West German Genome Center, Bonn, Germany; Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany; Institute of Human Genetics, Universitätsklinikum Schleswig-Holstein, University of Lübeck and University of Kiel, 23562 Lübeck, Germany
| | - Marc D Beyer
- PRECISE Platform for Single cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn and West German Genome Center, Bonn, Germany; Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Thomas Ulas
- PRECISE Platform for Single cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn and West German Genome Center, Bonn, Germany; Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Germany
| | - Charlotte Esser
- IUF-Leibniz Research Institute for Environmental Medicine gGmbH, Düsseldorf, Germany
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany; IUF-Leibniz Research Institute for Environmental Medicine gGmbH, Düsseldorf, Germany
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany.
| | - Laia Reverte-Salisa
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany.
| |
Collapse
|
4
|
Uner B, Celik A, Ergin AD, Altay Benetti A, Benetti C. Enhancement of in-vivo cellular uptake of Coenzyme Q10 using saponin derivatives in rTALAP transgenic mice model. J Drug Deliv Sci Technol 2024; 96:105636. [DOI: 10.1016/j.jddst.2024.105636] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
|
5
|
Agarwal M, Roth K, Yang Z, Sharma R, Maddipati K, Westrick J, Petriello MC. Loss of flavin-containing monooxygenase 3 modulates dioxin-like polychlorinated biphenyl 126-induced oxidative stress and hepatotoxicity. ENVIRONMENTAL RESEARCH 2024; 250:118492. [PMID: 38373550 PMCID: PMC11102846 DOI: 10.1016/j.envres.2024.118492] [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: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/21/2024]
Abstract
Dioxin-like pollutants (DLPs), such as polychlorinated biphenyl 126 (PCB 126), are synthetic chemicals classified as persistent organic pollutants. They accumulate in adipose tissue and have been linked to cardiometabolic disorders, including fatty liver disease. The toxicity of these compounds is associated with activation of the aryl hydrocarbon receptor (Ahr), leading to the induction of phase I metabolizing enzyme cytochrome P4501a1 (Cyp1a1) and the subsequent production of reactive oxygen species (ROS). Recent research has shown that DLPs can also induce the xenobiotic detoxification enzyme flavin-containing monooxygenase 3 (FMO3), which plays a role in metabolic homeostasis. We hypothesized whether genetic deletion of Fmo3 could protect mice, particularly in the liver, where Fmo3 is most inducible, against PCB 126 toxicity. To test this hypothesis, male C57BL/6 wild-type (WT) mice and Fmo3 knockout (Fmo3 KO) mice were exposed to PCB 126 or vehicle (safflower oil) during a 12-week study, at weeks 2 and 4. Various analyses were performed, including hepatic histology, RNA-sequencing, and quantitation of PCB 126 and F2-isoprostane concentrations. The results showed that PCB 126 exposure caused macro and microvesicular fat deposition in WT mice, but this macrovesicular fatty change was absent in Fmo3 KO mice. Moreover, at the pathway level, the hepatic oxidative stress response was significantly different between the two genotypes, with the induction of specific genes observed only in WT mice. Notably, the most abundant F2-isoprostane, 8-iso-15-keto PGE2, increased in WT mice in response to PCB 126 exposure. The study's findings also demonstrated that hepatic tissue concentrations of PCB 126 were higher in WT mice compared to Fmo3 KO mice. In summary, the absence of FMO3 in mice led to a distinctive response to dioxin-like pollutant exposure in the liver, likely due to alterations in lipid metabolism and storage, underscoring the complex interplay of genetic factors in the response to environmental toxins.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Zhao Yang
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Rahul Sharma
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Krishnarao Maddipati
- Department of Pathology, Lipidomic Core Facility, Wayne State University, Detroit, MI, 48202, USA
| | - Judy Westrick
- Department of Chemistry, Lumigen Instrumentation Center, Wayne State University, Detroit, MI, 48202, USA
| | - Michael C Petriello
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
6
|
Uner B, Celik A, Ergin AD, Altay Benetti A, Benetti C. Enhancement of in-vivo cellular uptake of Coenzyme Q10 using saponin derivatives in rTALAP transgenic mice model. J Drug Deliv Sci Technol 2024; 96:105636. [DOI: 7.https:/doi.org/10.1016/j.jddst.2024.105636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
|
7
|
Mosca A, Manco M, Braghini MR, Cianfarani S, Maggiore G, Alisi A, Vania A. Environment, Endocrine Disruptors, and Fatty Liver Disease Associated with Metabolic Dysfunction (MASLD). Metabolites 2024; 14:71. [PMID: 38276306 PMCID: PMC10819942 DOI: 10.3390/metabo14010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Ecological theories suggest that environmental factors significantly influence obesity risk and related syndemic morbidities, including metabolically abnormal obesity associated with nonalcoholic fatty liver disease (MASLD). These factors encompass anthropogenic influences and endocrine-disrupting chemicals (EDCs), synergistically interacting to induce metabolic discrepancies, notably in early life, and disrupt metabolic processes in adulthood. This review focuses on endocrine disruptors affecting a child's MASLD risk, independent of their role as obesogens and thus regardless of their impact on adipogenesis. The liver plays a pivotal role in metabolic and detoxification processes, where various lipophilic endocrine-disrupting molecules accumulate in fatty liver parenchyma, exacerbating inflammation and functioning as new anthropogenics that perpetuate chronic low-grade inflammation, especially insulin resistance, crucial in the pathogenesis of MASLD.
Collapse
Affiliation(s)
- Antonella Mosca
- Hepatology and Liver Transplant Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Melania Manco
- Preventive and Predictive Medicine Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Maria Rita Braghini
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.R.B.); (A.A.)
| | - Stefano Cianfarani
- Endocrinology and Diabetes Unit, Bambino Gesù Pediatric Hospital, 00165 Rome, Italy;
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Women’s and Children’s Health, Karolinska Institutet, University Hospital, Solnavägen 1, Solna, 171 77 Stockholm, Sweden
| | - Giuseppe Maggiore
- Hepatology and Liver Transplant Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.R.B.); (A.A.)
| | | |
Collapse
|
8
|
Rance N. How single-cell transcriptomics provides insight on hepatic responses to TCDD. CURRENT OPINION IN TOXICOLOGY 2023; 36:100441. [PMID: 37981901 PMCID: PMC10653208 DOI: 10.1016/j.cotox.2023.100441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
The prototypical aryl hydrocarbon receptor (AHR) ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), has been a valuable model for investigating toxicant-associated fatty liver disease (TAFLD). TCDD induces dose-dependent hepatic lipid accumulation, followed by the development of inflammatory foci and eventual progression to fibrosis in mice. Previously, bulk approaches and in vitro examination of different cell types were relied upon to study the mechanisms underlying TCDD-induced liver pathologies. However, the advent of single-cell transcriptomic technologies, such as single-nuclei RNA sequencing (snRNAseq) and spatial transcriptomics (STx), has provided new insights into the responses of hepatic cell types to TCDD exposure. This review explores the application of these single-cell transcriptomic technologies and highlights their contributions towards unraveling the cell-specific mechanisms mediating the hepatic responses to TCDD.
Collapse
Affiliation(s)
- Nault Rance
- Institute for Integrative Toxicology, Michigan State University, Michigan, USA
- Department of Biochemistry & Molecular Biology, Michigan State University, Michigan, USA
| |
Collapse
|
9
|
Piwarski SA, Salisbury TB. The effects of environmental aryl hydrocarbon receptor ligands on signaling and cell metabolism in cancer. Biochem Pharmacol 2023; 216:115771. [PMID: 37652105 DOI: 10.1016/j.bcp.2023.115771] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Dioxin and dioxin-like compounds are chlorinated organic pollutants formed during the manufacturing of other chemicals. Dioxins are ligands of the aryl hydrocarbon receptor (AHR), that induce AHR-mediated biochemical and toxic responses and are persistent in the environment. 2,3,7,8- tetrachlorodibenzo para dioxin (TCDD) is the prototypical AHR ligand and its effects represent dioxins. TCDD induces toxicity, immunosuppression and is a suspected tumor promoter. The role of TCDD in cancer however is debated and context-dependent. Environmental particulate matter, polycyclic aromatic hydrocarbons, perfluorooctane sulfonamide, endogenous AHR ligands, and cAMP signaling activate AHR through TCDD-independent pathways. The effect of activated AHR in cancer is context-dependent. The ability of FDA-approved drugs to modulate AHR activity has sparked interest in their repurposing for cancer therapy. TCDD by interfering with endogenous pathways, and overstimulating other endogenous pathways influences all stages of cancer. Herein we review signaling mechanisms that activate AHR and mechanisms by which activated AHR modulates signaling in cancer including affected metabolic pathways.
Collapse
Affiliation(s)
- Sean A Piwarski
- Duke Cancer Institute, Department of GU Oncology, Duke University Medical Center, 905 South Lasalle Street, Durham, NC 27710, USA.
| | - Travis B Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| |
Collapse
|
10
|
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
|
11
|
Zhang Y, Feng X, Chen J, Liu J, Wu J, Tan H, Mi Z, Rong P. Controversial role of ILC3s in intestinal diseases: A novelty perspective on immunotherapy. Front Immunol 2023; 14:1134636. [PMID: 37063879 PMCID: PMC10090672 DOI: 10.3389/fimmu.2023.1134636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
ILC3s have been identified as crucial immune regulators that play a role in maintaining host homeostasis and modulating the antitumor response. Emerging evidence supports the idea that LTi cells play an important role in initiating lymphoid tissue development, while other ILC3s can promote host defense and orchestrate adaptive immunity, mainly through the secretion of specific cytokines and crosstalk with other immune cells or tissues. Additionally, dysregulation of ILC3-mediated overexpression of cytokines, changes in subset abundance, and conversion toward other ILC subsets are closely linked with the occurrence of tumors and inflammatory diseases. Regulation of ILC3 cytokines, ILC conversion and LTi-induced TLSs may be a novel strategy for treating tumors and intestinal or extraintestinal inflammatory diseases. Herein, we discuss the development of ILCs, the biology of ILC3s, ILC plasticity, the correlation of ILC3s and adaptive immunity, crosstalk with the intestinal microenvironment, controversial roles of ILC3s in intestinal diseases and potential applications for treatment.
Collapse
Affiliation(s)
- Yunshu Zhang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuefei Feng
- Department of Government & Public Administration, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Juan Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahao Liu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianmin Wu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongpei Tan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| |
Collapse
|
12
|
da Cruz LL, Vesentini G, Sinzato YK, Villaverde AISB, Volpato GT, Damasceno DC. Effects of high-fat diet-induced diabetes on autophagy in the murine liver: A systematic review and meta-analysis. Life Sci 2022; 309:121012. [PMID: 36179817 DOI: 10.1016/j.lfs.2022.121012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/19/2022] [Accepted: 09/25/2022] [Indexed: 12/09/2022]
Abstract
AIMS We conducted a meta-analysis to investigate whether diabetes induced by a high-fat diet (HFD) has the potential to alter the process of autophagy in the murine liver. METHODS A systematic literature search was performed with electronic databases (PubMed, EMBASE, Web of Science). Study design, population, intervention, outcome, and risk of bias were analyzed. Given the availability of studies, a quantitative meta-analysis including 23 studies was performed. KEY FINDINGS The search found 5754 articles, with 48 matching the eligibility criteria, comprising of 1033 animals. The meta-analysis showed that diabetic murines fed with HFD presented an absence of p62 degradation (SMD 4.63, 95 % CI 2.02 to 7.24, p = 0.0005; I2 = 77 %), higher expression of p-mTOR/mTOR (SMD 5.20, 95 % CI 1.00 to 9.39, p = 0.01; I2 = 78 %), and a decreased p-AMPK/AMPK ratio (SMD -2.02, 95 % CI -3.96 to -0.09, p = 0.04; I2 = 85 %) when compared to nondiabetic murines. When associated with streptozotocin, the animals presented decreased ATG-7 and LC3-II. The meta-regression results showed a decrease in autophagy responses due to increased glycemic levels, fat content, and long-term exposure to HFD, and advanced animal age. The common and species-specific protein responses were also consistent with the inhibition of autophagy. SIGNIFICANCE The normal process of autophagy mechanisms in the liver is less competent after HFD consumption. The destabilization of (auto)phagolysosomes contributes to the perpetuation of diabetes, metabolic dysfunction-associated fatty liver disease, and cell death.
Collapse
Affiliation(s)
- Larissa Lopes da Cruz
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil; Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Giovana Vesentini
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Ana Izabel Silva Balbin Villaverde
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| |
Collapse
|
13
|
Yoda T, Tochitani T, Usui T, Kouchi M, Inada H, Hosaka T, Kanno Y, Miyawaki I, Yoshinari K. Involvement of the CYP1A1 inhibition-mediated activation of aryl hydrocarbon receptor in drug-induced hepatotoxicity. J Toxicol Sci 2022; 47:359-373. [PMID: 36047110 DOI: 10.2131/jts.47.359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Hepatotoxicity is one of the most common toxicities observed in non-clinical safety studies of drug candidates, and it is important to understand the hepatotoxicity mechanism to assess the risk of drug-induced liver injury in humans. In this study, we investigated the mechanism of hepatotoxicity caused by 2-[2-Methyl-1-(oxan-4-yl)-1H-benzimidazol-5-yl]-1,3-benzoxazole (DSP-0640), a drug candidate that showed hepatotoxicity characterized by centrilobular hypertrophy and vacuolation of hepatocytes in a 4-week oral repeated-dose toxicity study in male rats. In the liver of rats treated with DSP-0640, the expression of aryl hydrocarbon receptor (AHR) target genes, including Cyp1a1, was upregulated. In in vitro reporter assays, however, DSP-0640 showed only minimal AHR-activating potency. Therefore, we investigated the possibility that DSP-0640 indirectly activated AHR by inhibiting the CYP1 enzyme-dependent clearance of endogenous AHR agonists. In in vitro assays, DSP-0640 showed inhibitory effects on both rat and human CYP1A1 and enhanced rat and human AHR-mediated reporter gene expression induced by 6-formylindolo[3,2-b]carbazole, a well-known endogenous AHR agonist. The possible involvement of CYP1A1 inhibition in AHR activation was also demonstrated with other hepatotoxic compounds tacrine and albendazole. These results suggest that CYP1A1 inhibition-mediated AHR activation is involved in the hepatotoxicity caused by DSP-0640 and that DSP-0640 might induce hepatotoxicity in humans as well. We propose that CYP1A1 inhibition-mediated AHR activation is a novel mechanism for drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Tomomi Yoda
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Toru Usui
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd
| | - Mami Kouchi
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd
| | | | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yuichiro Kanno
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
14
|
Haque N, Tischkau SA. Sexual Dimorphism in Adipose-Hypothalamic Crosstalk and the Contribution of Aryl Hydrocarbon Receptor to Regulate Energy Homeostasis. Int J Mol Sci 2022; 23:ijms23147679. [PMID: 35887027 PMCID: PMC9322714 DOI: 10.3390/ijms23147679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022] Open
Abstract
There are fundamental sex differences in the regulation of energy homeostasis. Better understanding of the underlying mechanisms of energy balance that account for this asymmetry will assist in developing sex-specific therapies for sexually dimorphic diseases such as obesity. Multiple organs, including the hypothalamus and adipose tissue, play vital roles in the regulation of energy homeostasis, which are regulated differently in males and females. Various neuronal populations, particularly within the hypothalamus, such as arcuate nucleus (ARC), can sense nutrient content of the body by the help of peripheral hormones such leptin, derived from adipocytes, to regulate energy homeostasis. This review summarizes how adipose tissue crosstalk with homeostatic network control systems in the brain, which includes energy regulatory regions and the hypothalamic–pituitary axis, contribute to energy regulation in a sex-specific manner. Moreover, development of obesity is contingent upon diet and environmental factors. Substances from diet and environmental contaminants can exert insidious effects on energy metabolism, acting peripherally through the aryl hydrocarbon receptor (AhR). Developmental AhR activation can impart permanent alterations of neuronal development that can manifest a number of sex-specific physiological changes, which sometimes become evident only in adulthood. AhR is currently being investigated as a potential target for treating obesity. The consensus is that impaired function of the receptor protects from obesity in mice. AhR also modulates sex steroid receptors, and hence, one of the objectives of this review is to explain why investigating sex differences while examining this receptor is crucial. Overall, this review summarizes sex differences in the regulation of energy homeostasis imparted by the adipose–hypothalamic axis and examines how this axis can be affected by xenobiotics that signal through AhR.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Shelley A. Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Correspondence:
| |
Collapse
|
15
|
Xu X, Zhang X, Yuan Y, Zhao Y, Fares HM, Yang M, Wen Q, Taha R, Sun L. Species-Specific Differences in Aryl Hydrocarbon Receptor Responses: How and Why? Int J Mol Sci 2021; 22:ijms222413293. [PMID: 34948089 PMCID: PMC8708342 DOI: 10.3390/ijms222413293] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/19/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates a wide range of biological and toxicological effects by binding to specific ligands. AhR ligands exist in various internal and external ecological systems, such as in a wide variety of hydrophobic environmental contaminants and naturally occurring chemicals. Most of these ligands have shown differential responses among different species. Understanding the differences and their mechanisms helps in designing better experimental animal models, improves our understanding of the environmental toxicants related to AhR, and helps to screen and develop new drugs. This review systematically discusses the species differences in AhR activation effects and their modes of action. We focus on the species differences following AhR activation from two aspects: (1) the molecular configuration and activation of AhR and (2) the contrast of cis-acting elements corresponding to AhR. The variations in the responses seen in humans and other species following the activation of the AhR signaling pathway can be attributed to both factors.
Collapse
Affiliation(s)
- Xiaoting Xu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Xi Zhang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Yuzhu Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Yongrui Zhao
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Hamza M. Fares
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Mengjiao Yang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Qing Wen
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Reham Taha
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
| | - Lixin Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; (X.X.); (X.Z.); (Y.Y.); (Y.Z.); (H.M.F.); (M.Y.); (Q.W.); (R.T.)
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China
- Correspondence: ; Tel.: +86-151-9599-9925
| |
Collapse
|
16
|
Activation of the EGFR-PI3K-CaM pathway by PRL-1-overexpressing placenta-derived mesenchymal stem cells ameliorates liver cirrhosis via ER stress-dependent calcium. Stem Cell Res Ther 2021; 12:551. [PMID: 34689832 PMCID: PMC8543968 DOI: 10.1186/s13287-021-02616-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Background Cholesterol accumulation and calcium depletion induce hepatic injury via the endoplasmic reticulum (ER) stress response. ER stress regulates the calcium imbalance between the ER and mitochondria. We previously reported that phosphatase of regenerating liver-1 (PRL-1)-overexpressing placenta-derived mesenchymal stem cells (PD-MSCsPRL−1) promoted liver regeneration via mitochondrial dynamics in a cirrhotic rat model. However, the role of PRL-1 in ER stress-dependent calcium is not clear. Therefore, we demonstrated that PD-MSCsPRL−1 improved hepatic functions by regulating ER stress and calcium channels in a rat model of bile duct ligation (BDL). Methods Liver cirrhosis was induced in Sprague–Dawley (SD) rats using surgically induced BDL for 10 days. PD-MSCs and PD-MSCsPRL−1 (2 × 106 cells) were intravenously administered to animals, and their therapeutic effects were analyzed. WB-F344 cells exposed to thapsigargin (TG) were cocultured with PD-MSCs or PD-MSCsPRL−1. Results ER stress markers, e.g., eukaryotic translation initiation factor 2α (eIF2α), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP), were increased in the nontransplantation group (NTx) compared to the control group. PD-MSCsPRL−1 significantly decreased ER stress markers compared to NTx and induced dynamic changes in calcium channel markers, e.g., sarco/endoplasmic reticulum Ca2+ -ATPase 2b (SERCA2b), inositol 1,4,5-trisphosphate receptor (IP3R), mitochondrial calcium uniporter (MCU), and voltage-dependent anion channel 1 (VDAC1) (*p < 0.05). Cocultivation of TG-treated WB-F344 cells with PD-MSCsPRL−1 decreased cytosolic calmodulin (CaM) expression and cytosolic and mitochondrial Ca2+ concentrations. However, the ER Ca2+ concentration was increased compared to PD-MSCs (*p < 0.05). PRL-1 activated phosphatidylinositol-3-kinase (PI3K) signaling via epidermal growth factor receptor (EGFR), which resulted in calcium increase via CaM expression. Conclusions These findings suggest that PD-MSCsPRL−1 improved hepatic functions via calcium changes and attenuated ER stress in a BDL-injured rat model. Therefore, these results provide useful data for the development of next-generation MSC-based stem cell therapy for regenerative medicine in chronic liver disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02616-y.
Collapse
|
17
|
Hutin D, Long AS, Sugamori K, Shao P, Singh SK, Rasmussen M, Olafsen NE, Pettersen S, Grimaldi G, Grant DM, Matthews J. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD)-Inducible Poly-ADP-Ribose Polymerase (TIPARP/PARP7) Catalytic Mutant Mice (TiparpH532A) Exhibit Increased Sensitivity to TCDD-Induced Hepatotoxicity and Lethality. Toxicol Sci 2021; 183:154-169. [PMID: 34129049 PMCID: PMC8404992 DOI: 10.1093/toxsci/kfab075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly-adenosine diphosphate (ADP)-ribose polymerase (TIPARP/PARP7), an aryl hydrocarbon receptor (AHR) target gene and mono-ADP-ribosyltransferase, acts as part of a negative feedback loop to repress AHR signaling. This process is prevented by a single H532A mutation in TIPARP that destroys its catalytic activity. We hypothesized that the loss of TIPARP catalytic activity would increase sensitivity to TCDD-induced toxicity in vivo. To test this, we created a catalytically deficient mouse line (TiparpH532A) by introducing a single H532A mutation in TIPARP. Treatment of mouse embryonic fibroblasts or hepatocytes isolated from TiparpH532A mice confirmed the increased TCDD-induced expression of the AHR target genes Cyp1a1, Cyp1b1, and Tiparp. TiparpH532A mice given a single injection of 10 µg/kg TCDD, a nonlethal dose in Tiparp+/+ mice, did not survive beyond day 10. All Tiparp+/+ mice survived the 30-day treatment. TCDD-treated TiparpH532A mice displayed increased expression of AHR target genes, increased steatohepatitis and hepatotoxicity. Hepatic RNA-sequencing revealed 7-fold more differentially expressed genes in TiparpH532A mice than in Tiparp+/+ mice (4542 vs 647 genes) 6 days after TCDD treatment. Differentially expressed genes included genes involved in xenobiotic metabolism, lipid homeostasis and inflammation. Taken together, these data further support TIPARP as a critical negative regulator of AHR activity and show that loss of its catalytic activity is sufficient to increase sensitivity to TCDD-induced steatohepatitis and lethality. Since TIPARP inhibition has recently emerged as a potential anticancer therapy, the impact on AHR signaling, TCDD and polycyclic aromatic hydrocarbon toxicity will need to be carefully considered under conditions of therapeutic TIPARP inhibition.
Collapse
Affiliation(s)
- David Hutin
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8 Ontario, Canada
| | - Alexandra S Long
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8 Ontario, Canada
| | - Kim Sugamori
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8 Ontario, Canada
| | - Peng Shao
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8 Ontario, Canada
| | | | - Marit Rasmussen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Ninni Elise Olafsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Solveig Pettersen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Giulia Grimaldi
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Denis M Grant
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8 Ontario, Canada
| | - Jason Matthews
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8 Ontario, Canada.,Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
18
|
Matteo G, Hoyeck MP, Blair HL, Zebarth J, Rick KRC, Williams A, Gagné R, Buick JK, Yauk CL, Bruin JE. Prolonged Low-Dose Dioxin Exposure Impairs Metabolic Adaptability to High-Fat Diet Feeding in Female but Not Male Mice. Endocrinology 2021; 162:bqab050. [PMID: 33693622 PMCID: PMC8101695 DOI: 10.1210/endocr/bqab050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Human studies consistently show an association between exposure to persistent organic pollutants, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, aka "dioxin"), and increased diabetes risk. We previously showed that a single high-dose TCDD exposure (20 µg/kg) decreased plasma insulin levels in male and female mice in vivo, but effects on glucose homeostasis were sex-dependent. OBJECTIVE The current study assessed whether prolonged exposure to a physiologically relevant low-dose of TCDD impacts glucose homeostasis and/or the islet phenotype in a sex-dependent manner in chow-fed or high-fat diet (HFD)-fed mice. METHODS Male and female mice were exposed to 20 ng/kg/d TCDD 2×/week for 12 weeks and simultaneously fed standard chow or a 45% HFD. Glucose homeostasis was assessed by glucose and insulin tolerance tests, and glucose-induced plasma insulin levels were measured in vivo. Histological analysis was performed on pancreas from male and female mice, and islets were isolated from females for TempO-Seq transcriptomic analysis. RESULTS Low-dose TCDD exposure did not lead to adverse metabolic consequences in chow-fed male or female mice, or in HFD-fed males. However, TCDD accelerated the onset of HFD-induced hyperglycemia and impaired glucose-induced plasma insulin levels in females. TCDD caused a modest increase in islet area in males but reduced the percent beta cell area within islets in females. TempO-Seq analysis suggested abnormal changes to endocrine and metabolic pathways in female TCDDHFD islets. CONCLUSION Our data suggest that prolonged low-dose TCDD exposure has minimal effects on glucose homeostasis and islet morphology in chow-fed male and female mice but promotes maladaptive metabolic responses in HFD-fed females.
Collapse
Affiliation(s)
- Geronimo Matteo
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Myriam P Hoyeck
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Hannah L Blair
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Julia Zebarth
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Kayleigh R C Rick
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Rémi Gagné
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Julie K Buick
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
- Department of Biology, University of Ottawa, Ontario, KIN 6N5, Canada
| | - Jennifer E Bruin
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| |
Collapse
|
19
|
Cano R, Pérez JL, Dávila LA, Ortega Á, Gómez Y, Valero-Cedeño NJ, Parra H, Manzano A, Véliz Castro TI, Albornoz MPD, Cano G, Rojas-Quintero J, Chacín M, Bermúdez V. Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. Int J Mol Sci 2021; 22:4807. [PMID: 34062716 PMCID: PMC8125512 DOI: 10.3390/ijms22094807] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder, affecting around 25% of the population worldwide. It is a complex disease spectrum, closely linked with other conditions such as obesity, insulin resistance, type 2 diabetes mellitus, and metabolic syndrome, which may increase liver-related mortality. In light of this, numerous efforts have been carried out in recent years in order to clarify its pathogenesis and create new prevention strategies. Currently, the essential role of environmental pollutants in NAFLD development is recognized. Particularly, endocrine-disrupting chemicals (EDCs) have a notable influence. EDCs can be classified as natural (phytoestrogens, genistein, and coumestrol) or synthetic, and the latter ones can be further subdivided into industrial (dioxins, polychlorinated biphenyls, and alkylphenols), agricultural (pesticides, insecticides, herbicides, and fungicides), residential (phthalates, polybrominated biphenyls, and bisphenol A), and pharmaceutical (parabens). Several experimental models have proposed a mechanism involving this group of substances with the disruption of hepatic metabolism, which promotes NAFLD. These include an imbalance between lipid influx/efflux in the liver, mitochondrial dysfunction, liver inflammation, and epigenetic reprogramming. It can be concluded that exposure to EDCs might play a crucial role in NAFLD initiation and evolution. However, further investigations supporting these effects in humans are required.
Collapse
Affiliation(s)
- Raquel Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - José L. Pérez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Lissé Angarita Dávila
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Sede Concepción 4260000, Chile;
| | - Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Yosselin Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Nereida Josefina Valero-Cedeño
- Carrera de Laboratorio Clínico, Facultad de Ciencias de la Salud, Universidad Estatal del Sur de Manabí, Jipijapa E482, Ecuador; (N.J.V.-C.); (T.I.V.C.)
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Teresa Isabel Véliz Castro
- Carrera de Laboratorio Clínico, Facultad de Ciencias de la Salud, Universidad Estatal del Sur de Manabí, Jipijapa E482, Ecuador; (N.J.V.-C.); (T.I.V.C.)
| | - María P. Díaz Albornoz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Gabriel Cano
- Insitute für Pharmazie, Freie Universitänt Berlin, Königin-Louise-Strabe 2-4, 14195 Berlin, Germany;
| | - Joselyn Rojas-Quintero
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud. Barranquilla, Universidad Simón Bolívar, Barranquilla 55-132, Colombia;
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud. Barranquilla, Universidad Simón Bolívar, Barranquilla 55-132, Colombia;
| |
Collapse
|
20
|
Esteban J, Sánchez-Pérez I, Hamscher G, Miettinen HM, Korkalainen M, Viluksela M, Pohjanvirta R, Håkansson H. Role of aryl hydrocarbon receptor (AHR) in overall retinoid metabolism: Response comparisons to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure between wild-type and AHR knockout mice. Reprod Toxicol 2021; 101:33-49. [PMID: 33607186 DOI: 10.1016/j.reprotox.2021.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/20/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Young adult wild-type and aryl hydrocarbon receptor knockout (AHRKO) mice of both sexes and the C57BL/6J background were exposed to 10 weekly oral doses of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; total dose of 200 μg/kg bw) to further characterize the observed impacts of AHR as well as TCDD on the retinoid system. Unexposed AHRKO mice harboured heavier kidneys, lighter livers and lower serum all-trans retinoic acid (ATRA) and retinol (REOH) concentrations than wild-type mice. Results from the present study also point to a role for the murine AHR in the control of circulating REOH and ATRA concentrations. In wild-type mice, TCDD elevated liver weight and reduced thymus weight, and drastically reduced the hepatic concentrations of 9-cis-4-oxo-13,14-dihydro-retinoic acid (CORA) and retinyl palmitate (REPA). In female wild-type mice, TCDD increased the hepatic concentration of ATRA as well as the renal and circulating REOH concentrations. Renal CORA concentrations were substantially diminished in wild-type male mice exclusively following TCDD-exposure, with a similar tendency in serum. In contrast, TCDD did not affect any of these toxicity or retinoid system parameters in AHRKO mice. Finally, a distinct sex difference occurred in kidney concentrations of all the analysed retinoid forms. Together, these results strengthen the evidence of a mandatory role of AHR in TCDD-induced retinoid disruption, and suggest that the previously reported accumulation of several retinoid forms in the liver of AHRKO mice is a line-specific phenomenon. Our data further support participation of AHR in the control of liver and kidney development in mice.
Collapse
Affiliation(s)
- Javier Esteban
- Instituto De Bioingeniería, Universidad Miguel Hernández De Elche, Elche, Alicante, Spain.
| | - Ismael Sánchez-Pérez
- Instituto De Bioingeniería, Universidad Miguel Hernández De Elche, Elche, Alicante, Spain.
| | - Gerd Hamscher
- Institute of Food Chemistry and Food Biotechnology, Justus Liebig University Giessen, Giessen, Germany.
| | - Hanna M Miettinen
- School of Pharmacy (Toxicology) and Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Merja Korkalainen
- Environmental Health Unit, Finnish Insitute for Health and Welfare (THL), Kuopio, Finland.
| | - Matti Viluksela
- School of Pharmacy (Toxicology) and Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland; Environmental Health Unit, Finnish Insitute for Health and Welfare (THL), Kuopio, Finland.
| | - Raimo Pohjanvirta
- Department of Food Hygiene & Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Mustialankatu 1, FI-00790 Helsinki, Finland.
| | - Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
21
|
Rawls KD, Dougherty BV, Vinnakota KC, Pannala VR, Wallqvist A, Kolling GL, Papin JA. Predicting changes in renal metabolism after compound exposure with a genome-scale metabolic model. Toxicol Appl Pharmacol 2021; 412:115390. [PMID: 33387578 PMCID: PMC7859602 DOI: 10.1016/j.taap.2020.115390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/02/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
The kidneys are metabolically active organs with importance in several physiological tasks such as the secretion of soluble wastes into the urine and synthesizing glucose and oxidizing fatty acids for energy in fasting (non-fed) conditions. Once damaged, the metabolic capability of the kidneys becomes altered. Here, we define metabolic tasks in a computational modeling framework to capture kidney function in an update to the iRno network reconstruction of rat metabolism using literature-based evidence. To demonstrate the utility of iRno for predicting kidney function, we exposed primary rat renal proximal tubule epithelial cells to four compounds with varying levels of nephrotoxicity (acetaminophen, gentamicin, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six and twenty-four hours, and collected transcriptomics and metabolomics data to measure the metabolic effects of compound exposure. For the transcriptomics data, we observed changes in fatty acid metabolism and amino acid metabolism, as well as changes in existing markers of kidney function such as Clu (clusterin). The iRno metabolic network reconstruction was used to predict alterations in these same pathways after integrating transcriptomics data and was able to distinguish between select compound-specific effects on the proximal tubule epithelial cells. Genome-scale metabolic network reconstructions with coupled omics data can be used to predict changes in metabolism as a step towards identifying novel metabolic biomarkers of kidney function and dysfunction.
Collapse
Affiliation(s)
- Kristopher D Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Bonnie V Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kalyan C Vinnakota
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, MD 20817, USA
| | - Venkat R Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, MD 20817, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA
| | - Glynis L Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA; Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
22
|
Girer NG, Tomlinson CR, Elferink CJ. The Aryl Hydrocarbon Receptor in Energy Balance: The Road from Dioxin-Induced Wasting Syndrome to Combating Obesity with Ahr Ligands. Int J Mol Sci 2020; 22:E49. [PMID: 33374508 PMCID: PMC7793057 DOI: 10.3390/ijms22010049] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) has been studied for over 40 years, yet our understanding of this ligand-activated transcription factor remains incomplete. Each year, novel findings continually force us to rethink the role of the AHR in mammalian biology. The AHR has historically been studied within the context of potent activation via AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), with a focus on how the AHR mediates TCDD toxicity. Research has subsequently revealed that the AHR is actively involved in distinct physiological processes ranging from the development of the liver and reproductive organs, to immune system function and wound healing. More recently, the AHR was implicated in the regulation of energy metabolism and is currently being investigated as a potential therapeutic target for obesity. In this review, we re-trace the steps through which the early toxicological studies of TCDD led to the conceptual framework for the AHR as a potential therapeutic target in metabolic disease. We additionally discuss the key discoveries that have been made concerning the role of the AHR in energy metabolism, as well as the current and future directions of the field.
Collapse
Affiliation(s)
- Nathaniel G. Girer
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA;
| | - Craig R. Tomlinson
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Dartmouth Hitchcock Medical Center, Dartmouth College, Lebanon, NH 03756, USA;
| | - Cornelis J. Elferink
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA;
| |
Collapse
|
23
|
Teino I, Matvere A, Pook M, Varik I, Pajusaar L, Uudeküll K, Vaher H, Trei A, Kristjuhan A, Org T, Maimets T. Impact of AHR Ligand TCDD on Human Embryonic Stem Cells and Early Differentiation. Int J Mol Sci 2020; 21:E9052. [PMID: 33260776 PMCID: PMC7731104 DOI: 10.3390/ijms21239052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 12/17/2022] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which mediates the effects of a variety of environmental stimuli in multiple tissues. Recent advances in AHR biology have underlined its importance in cells with high developmental potency, including pluripotent stem cells. Nonetheless, there is little data on AHR expression and its role during the initial stages of stem cell differentiation. The purpose of this study was to investigate the temporal pattern of AHR expression during directed differentiation of human embryonic stem cells (hESC) into neural progenitor, early mesoderm and definitive endoderm cells. Additionally, we investigated the effect of the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the gene expression profile in hESCs and differentiated cells by RNA-seq, accompanied by identification of AHR binding sites by ChIP-seq and epigenetic landscape analysis by ATAC-seq. We showed that AHR is differentially regulated in distinct lineages. We provided evidence that TCDD alters gene expression patterns in hESCs and during early differentiation. Additionally, we identified novel potential AHR target genes, which expand our understanding on the role of this protein in different cell types.
Collapse
Affiliation(s)
- Indrek Teino
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Antti Matvere
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Martin Pook
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Inge Varik
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Laura Pajusaar
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Keyt Uudeküll
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Helen Vaher
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Annika Trei
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Arnold Kristjuhan
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| | - Tõnis Org
- Chair of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia;
- Institute of Genomics, University of Tartu, Riia 23b, 51010 Tartu, Estonia
| | - Toivo Maimets
- Chair of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia; (A.M.); (M.P.); (I.V.); (L.P.); (K.U.); (H.V.); (A.T.); (A.K.); (T.M.)
| |
Collapse
|
24
|
Maziero JS, Thipe VC, Rogero SO, Cavalcante AK, Damasceno KC, Ormenio MB, Martini GA, Batista JGS, Viveiros W, Katti KK, Raphael Karikachery A, Dhurvas Mohandoss D, Dhurvas RD, Nappinnai M, Rogero JR, Lugão AB, Katti KV. Species-Specific in vitro and in vivo Evaluation of Toxicity of Silver Nanoparticles Stabilized with Gum Arabic Protein. Int J Nanomedicine 2020; 15:7359-7376. [PMID: 33061384 PMCID: PMC7537814 DOI: 10.2147/ijn.s250467] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction We report, herein, in vitro, and in vivo toxicity evaluation of silver nanoparticles stabilized with gum arabic protein (AgNP-GP) in Daphnia similis, Danio rerio embryos and in Sprague Dawley rats. Purpose The objective of this investigation was to evaluate in vitro and in vivo toxicity of silver nanoparticles stabilized with gum arabic protein (AgNP-GP), in multispecies due to the recognition that toxicity evaluations beyond a single species reflect the environmental realism. In the present study, AgNP-GP was synthesized through the reduction of silver salt using the tri-alanine-phosphine peptide (commonly referred to as “Katti Peptide”) and stabilized using gum arabic protein. Methods In vitro cytotoxicity tests were performed according to ISO 10993–5 protocols to assess cytotoxicity index (IC50) values. Acute ecotoxicity (EC50) studies were performed using Daphnia similis, according to the ABNT NBR 15088 protocols. In vivo toxicity also included evaluation of acute embryotoxicity using Danio rerio (zebrafish) embryos following the OECD No. 236 guidelines. We also used Sprague Dawley rats to assess the toxicity of AgNP-GP in doses from 2.5 to 10.0 mg kg−1 body weight. Results AgNP-GP nanoparticles were characterized through UV (405 nm), core size (20±5 nm through TEM), hydrodynamic size (70–80 nm), Zeta (ζ) potential (- 26 mV) using DLS and Powder X ray diffraction (PXRD) and EDS. PXRD showed pattern consistent with the Ag (1 1 1) peak. EC50 in Daphnia similis was 4.40 (3.59–5.40) μg L−1. In the zebrafish species, LC50 was 177 μg L−1. Oral administration of AgNP-GP in Sprague Dawley rats for a period of 28 days revealed no adverse effects in doses of up to 10.0 mg kg−1 b.w. in both male and female animals. Conclusion The non-toxicity of AgNP-GP in rats offers a myriad of applications of AgNP-GP in health and hygiene for use as antibiotics, antimicrobial and antifungal agents.
Collapse
Affiliation(s)
- Joana S Maziero
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Velaphi C Thipe
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil.,Institute of Green Nanotechnology, Department of Radiology, University of Missouri Columbia, Columbia, MO, USA
| | - Sizue O Rogero
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Adriana K Cavalcante
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Kelme C Damasceno
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Matheus B Ormenio
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Gisela A Martini
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Jorge G S Batista
- Laboratório de Biomateriais Poliméricos e Nanotheranóstica, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - William Viveiros
- Laboratório de Ecotoxicologia, Companhia Ambiental do Estado de São Paulo (CETESB), São Paulo, SP, Brazil
| | - Kavita K Katti
- Institute of Green Nanotechnology, Department of Radiology, University of Missouri Columbia, Columbia, MO, USA
| | - Alice Raphael Karikachery
- Institute of Green Nanotechnology, Department of Radiology, University of Missouri Columbia, Columbia, MO, USA
| | | | | | | | - José R Rogero
- Laboratório de Biomateriais Poliméricos e Nanotheranóstica, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Ademar B Lugão
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear- IPEN/CNEN-SP, São Paulo, SP, Brazil
| | - Kattesh V Katti
- Institute of Green Nanotechnology, Department of Radiology, University of Missouri Columbia, Columbia, MO, USA
| |
Collapse
|
25
|
Ye M, Xu M, Fan S, Zhang M, Zhou B, Yang S, Wei W, Ji C, Ji J, Ji F. Protective effects of three propolis-abundant flavonoids against ethanol-induced injuries in HepG2 cells involving the inhibition of ERK1/2-AHR-CYP1A1 signaling pathways. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
26
|
Nault R, Fader KA, Bhattacharya S, Zacharewski TR. Single-Nuclei RNA Sequencing Assessment of the Hepatic Effects of 2,3,7,8-Tetrachlorodibenzo-p-dioxin. Cell Mol Gastroenterol Hepatol 2020; 11:147-159. [PMID: 32791302 PMCID: PMC7674514 DOI: 10.1016/j.jcmgh.2020.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Characterization of cell specific transcriptional responses to hepatotoxicants is lost in the averages of bulk RNA-sequencing (RNA-seq). Single-cell/nuclei RNA-seq technologies enable the transcriptomes of individual cell (sub)types to be assessed within the context of in vivo models. METHODS Single-nuclei RNA-sequencing (snSeq) of frozen liver samples from male C57BL/6 mice gavaged with sesame oil vehicle or 30 μg/kg 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) every 4 days for 28 days was used to demonstrate the application of snSeq for the evaluation of xenobiotics. RESULTS A total of 19,907 genes were detected across 16,015 nuclei from control and TCDD-treated livers. Eleven cell (sub)types reflected the expected cell diversity of the liver including distinct pericentral, midzonal, and periportal hepatocyte subpopulations. TCDD altered relative proportions of cell types and elicited cell-specific gene expression profiles. For example, macrophages increased from 0.5% to 24.7%, while neutrophils were only present in treated samples, consistent with histological evaluation. The number of differentially expressed genes (DEGs) in each cell type ranged from 122 (cholangiocytes) to 7625 (midcentral hepatocytes), and loosely correlated with the basal expression level of Ahr, the canonical mediator of TCDD and related compounds. In addition to the expected functions within each cell (sub)types, RAS signaling and related pathways were specifically enriched in nonparenchymal cells while metabolic process enrichment occurred primarily in hepatocytes. snSeq also identified the expansion of a Kupffer cell subtype highly expressing Gpnmb, as reported in a dietary NASH model. CONCLUSIONS We show that snSeq of frozen liver samples can be used to assess cell-specific transcriptional changes and population shifts in models of hepatotoxicity when examining freshly isolated cells is not feasible.
Collapse
Affiliation(s)
- Rance Nault
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Kelly A Fader
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Sudin Bhattacharya
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Tim R Zacharewski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
27
|
Liu Q, Shao W, Weng Z, Zhang X, Ding G, Xu C, Xu J, Jiang Z, Gu A. In vitro evaluation of the hepatic lipid accumulation of bisphenol analogs: A high-content screening assay. Toxicol In Vitro 2020; 68:104959. [PMID: 32763284 DOI: 10.1016/j.tiv.2020.104959] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 08/01/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023]
Abstract
Bisphenol A (BPA) has a variety of adverse effects on human health; therefore, BPA analogs are increasingly used as replacements. Notably, recent studies have revealed that BPA exposure induced hepatic lipid accumulation, but few studies are available regarding the similar effects of other bisphenol analogues (BPs). Thus, in the present study, a high-content screening (HCS) assay was performed to simultaneously evaluate the hepatic lipid accumulation of 13 BPs in vitro. The BPs induced lipid deposition in HepG2 cells ranking as below: 4,4'-thiodiphenol (TDP) < bisphenol S (BPS) < 4,4'-dihydroxybenzophenone (DHBP) < tetrabromobisphenol A (TBBPA) < tetrachlorobisphenol A (TCBPA) < bisphenol E (BPE) < bisphenol F (BPF) < bisphenol B (BPB) < bisphenol AF (BPAF) < bisphenol A (BPA) < bisphenol C (BPC) < tetramethylbisphenol A (TMBPA) < bisphenol AP (BPAP). Meanwhile, Oil Red O staining and triacylglycerol detection further validated the lipid accumulation elicited by the latter 8 BPs, which exhibited the more significant effects on lipid deposition. Mechanistically, significantly increased expressions of genes involved in fatty acid synthesis and nuclear receptors and decreased levels of genes associated with fatty acid β-oxidation were observed under BPs treatment. Therefore, the present work is the first to systematically provide direct evidence for BPs-induced hepatic lipid accumulation in vitro via HCS, which can be helpful for safety assessments of BPs.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Wentao Shao
- Shanghai East Hospital, Institute of Gallstone Disease, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhenkun Weng
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Guipeng Ding
- Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Cheng Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Jin Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhaoyan Jiang
- Shanghai East Hospital, Institute of Gallstone Disease, Tongji University School of Medicine, Shanghai 200120, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
28
|
Doskey CM, Fader KA, Nault R, Lydic T, Matthews J, Potter D, Sharratt B, Williams K, Zacharewski T. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alters hepatic polyunsaturated fatty acid metabolism and eicosanoid biosynthesis in female Sprague-Dawley rats. Toxicol Appl Pharmacol 2020; 398:115034. [PMID: 32387183 PMCID: PMC7294678 DOI: 10.1016/j.taap.2020.115034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent aryl hydrocarbon receptor (AhR) agonist that elicits a broad spectrum of dose-dependent hepatic effects including lipid accumulation, inflammation, and fibrosis. To determine the role of inflammatory lipid mediators in TCDD-mediated hepatotoxicity, eicosanoid metabolism was investigated. Female Sprague-Dawley (SD) rats were orally gavaged with sesame oil vehicle or 0.01-10 μg/kg TCDD every 4 days for 28 days. Hepatic RNA-Seq data was integrated with untargeted metabolomics of liver, serum, and urine, revealing dose-dependent changes in linoleic acid (LA) and arachidonic acid (AA) metabolism. TCDD also elicited dose-dependent differential gene expression associated with the cyclooxygenase, lipoxygenase, and cytochrome P450 epoxidation/hydroxylation pathways with corresponding changes in ω-6 (e.g. AA and LA) and ω-3 polyunsaturated fatty acids (PUFAs), as well as associated eicosanoid metabolites. Overall, TCDD increased the ratio of ω-6 to ω-3 PUFAs. Phospholipase A2 (Pla2g12a) was induced consistent with increased AA metabolism, while AA utilization by induced lipoxygenases Alox5 and Alox15 increased leukotrienes (LTs). More specifically, TCDD increased pro-inflammatory eicosanoids including leukotriene LTB4, and LTB3, known to recruit neutrophils to damaged tissue. Dose-response modeling suggests the cytochrome P450 hydroxylase/epoxygenase and lipoxygenase pathways are more sensitive to TCDD than the cyclooxygenase pathway. Hepatic AhR ChIP-Seq analysis found little enrichment within the regulatory regions of differentially expressed genes (DEGs) involved in eicosanoid biosynthesis, suggesting TCDD-elicited dysregulation of eicosanoid metabolism is a downstream effect of AhR activation. Overall, these results suggest alterations in eicosanoid metabolism may play a key role in TCDD-elicited hepatotoxicity associated with the progression of steatosis to steatohepatitis.
Collapse
Affiliation(s)
- Claire M Doskey
- Department of Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Kelly A Fader
- Department of Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Rance Nault
- Department of Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Todd Lydic
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States
| | - Jason Matthews
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo 0316, Norway
| | - Dave Potter
- Wellington Laboratories Inc., Guelph, Ontario NIG 3M5, Canada
| | - Bonnie Sharratt
- Wellington Laboratories Inc., Guelph, Ontario NIG 3M5, Canada
| | - Kurt Williams
- Department of Pathobiology and Diagnostic Investigation, Michigan State, East Lansing, MI 48824, United States
| | - Tim Zacharewski
- Department of Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
29
|
Renal changes and apoptosis caused by subacute exposure to Aroclor 1254 in selenium-deficient and selenium-supplemented rats. Arh Hig Rada Toksikol 2020; 71:110-120. [PMID: 32975097 PMCID: PMC7968486 DOI: 10.2478/aiht-2020-71-3360] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 06/01/2020] [Indexed: 12/21/2022] Open
Abstract
Aroclor 1254 (A1254), a mixture of polychlorinated biphenyls, exerts hepatic, renal, and reproductive toxicity in rodents. This study aimed to determine a protective role of selenium on histopathological changes, oxidative stress, and apoptosis caused by A1254 in rat kidney. It included a control group, which received regular diet containing 0.15 mg/kg Se (C), a Se-supplemented group (SeS) receiving 1 mg/kg Se, a Se-deficient group (SeD) receiving Se-deficient diet of ≤0.05 mg/kg Se, an A1254-treated group (A) receiving 10 mg/kg of Aroclor 1254 and regular diet, an A1254-treated group receiving Se-supplementation (ASeS), and an A1254-treated group receiving Se-deficient diet (ASeD). Treatments lasted 15 days. After 24 h of the last dose of A1254, the animals were decapitated under anaesthesia and their renal antioxidant enzyme activities, lipid peroxidation (LP), glutathione, protein oxidation, and total antioxidant capacity levels measured. Histopathological changes were evaluated by light and electron microscopy. Apoptosis was detected with the TUNEL assay. Kidney weights, CAT activities, and GSH levels decreased significantly in all A1254-treated groups. Renal atrophic changes and higher apoptotic cell counts were observed in the A and ASeD groups. Both groups also showed a significant drop in GPx1 activities (A – 34.92 % and ASeD – 86.46 %) and rise in LP (A – 30.45 % and ASeD – 20.44 %) vs control. In contrast, LP levels and apoptotic cell counts were significantly lower in the ASeS group vs the A group. Histopathological changes and renal apoptosis were particularly visible in the ASeD group. Our findings suggest that selenium supplementation provides partial protection against renal toxicity of Aroclor 1254.
Collapse
|
30
|
Williams AE, Watt J, Robertson LW, Gadupudi G, Osborn ML, Soares MJ, Iqbal K, Pedersen KB, Shankar K, Littleton S, Maimone C, Eti NA, Suva LJ, Ronis MJJ. Skeletal Toxicity of Coplanar Polychlorinated Biphenyl Congener 126 in the Rat Is Aryl Hydrocarbon Receptor Dependent. Toxicol Sci 2020; 175:113-125. [PMID: 32119087 PMCID: PMC7197949 DOI: 10.1093/toxsci/kfaa030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Epidemiological evidence links polychlorinated biphenyls (PCBs) to skeletal toxicity, however mechanisms whereby PCBs affect bone are poorly studied. In this study, coplanar PCB 126 (5 μmol/kg) or corn oil vehicle was administered to N = 5 and 6 male and female, wild type (WT) or AhR -/- rats via intraperitoneal injection. Animals were sacrificed after 4 weeks. Bone length was measured; bone morphology was assessed by microcomputed tomography and dynamic histomorphometry. Reduced bone length was the only genotype-specific effect and only observed in males (p < .05). WT rats exposed to PCB 126 had reduced serum calcium, and smaller bones with reduced tibial length, cortical area, and medullary area relative to vehicle controls (p < .05). Reduced bone formation rate observed in dynamic histomorphometry was consistent with inhibition of endosteal and periosteal bone growth. The effects of PCB 126 were abolished in AhR -/- rats. Gene expression in bone marrow and shaft were assessed by RNA sequencing. Approximately 75% of the PCB-regulated genes appeared AhR dependent with 89 genes significantly (p < .05) regulated by both PCB 126 and knockout of the AhR gene. Novel targets significantly induced by PCB 126 included Indian hedgehog (Ihh) and connective tissue growth factor (Ctgf/Ccn2), which regulate chondrocyte proliferation and differentiation in the bone growth plate and cell-matrix interactions. These data suggest the toxic effects of PCB 126 on bone are mediated by AhR, which has direct effects on the growth plate and indirect actions related to endocrine disruption. These studies clarify important mechanisms underlying skeletal toxicity of dioxin-like PCBs and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Ashlee E Williams
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana 70112
| | - James Watt
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana 70112
| | - Larry W Robertson
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa
| | - Gopi Gadupudi
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa
| | - Michele L Osborn
- Department of Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Michael J Soares
- Department of Pathology, University of Kansas Medical Center, Kansas City, Missouri
| | - Khursheed Iqbal
- Department of Pathology, University of Kansas Medical Center, Kansas City, Missouri
| | - Kim B Pedersen
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana 70112
| | - Kartik Shankar
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Shana Littleton
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana 70112
| | - Cole Maimone
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana 70112
| | - Nazmin A Eti
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa
| | - Larry J Suva
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, Texas
| | - Martin J J Ronis
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana 70112
| |
Collapse
|
31
|
Konstandi M, Andriopoulou CE, Cheng J, Gonzalez FJ. Sex steroid hormones differentially regulate CYP2D in female wild-type and CYP2D6-humanized mice. J Endocrinol 2020; 245:301-314. [PMID: 32171179 PMCID: PMC7202972 DOI: 10.1530/joe-19-0561] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/15/2022]
Abstract
The CYP2D subfamily catalyses the metabolism of about 25% of prescribed drugs, including the majority of antidepressants and antipsychotics. At present, the mechanism of hepatic CYP2D regulation remains largely unknown. This study investigated the role of sex steroid hormones in CYP2D regulation. For this purpose, Cyp2d22 expression was assessed in the distinct phases of the estrous cycle of normocyclic C57BL/6J (WT) female mice. Cyp2d22 was also evaluated in ovariectomised WT and CYP2D6-humanized (hCYP2D6) mice that received hormonal supplementation with either 17β-estradiol (E2) and/or progesterone. Comparisons were also made to male mice. The data revealed that hepatic Cyp2d22 mRNA, protein and activity levels were higher at estrous compared to the other phases of the estrous cycle and that ovariectomy repressed Cyp2d22 expression in WT mice. Tamoxifen, an anti-estrogenic compound, also repressed hepatic Cyp2d22 via activation of GH/STAT5b and PI3k/AKT signaling pathways. Both hormones prevented the ovariectomy-mediated Cyp2d22 repression. In case of progesterone, this may be mediated by inhibition of the PI3k/AKT/FOX01 pathway. Notably, Cyp2d22 mRNA levels in WT males were similar to those in ovariectomised mice and were markedly lower compared to females at estrous, a differentiation potentially regulated by the GH/STAT5b pathway. Sex steroid hormone-related alterations in Cyp2d22 mRNA expression were highly correlated with Hnf1a mRNA. Interestingly, fluctuations in Cyp2d22 in hippocampus and cerebellum followed those in liver. In contrast to WT mice, ovariectomy induced hepatic CYP2D6 expression in hCYP2D6 mice, whereas E2 and/or progesterone prevented this induction. Apparently, sex steroid hormones display a significant gender- and species-specific role in the regulation of CYP2D.
Collapse
Affiliation(s)
- Maria Konstandi
- University of Ioannina, School of Health Sciences, Faculty of Medicine, Department of Pharmacology, Ioannina GR-451 10, Greece
- National Institutes of Health, National Cancer Institute, Laboratory of Metabolism, Bethesda 20892, MD, USA
- Correspondence should be addressed to Dr Maria Konstandi, Phone +30 2651007554,
| | - Christina E. Andriopoulou
- University of Ioannina, School of Health Sciences, Faculty of Medicine, Department of Pharmacology, Ioannina GR-451 10, Greece
| | - Jie Cheng
- National Institutes of Health, National Cancer Institute, Laboratory of Metabolism, Bethesda 20892, MD, USA
| | - Frank J. Gonzalez
- National Institutes of Health, National Cancer Institute, Laboratory of Metabolism, Bethesda 20892, MD, USA
| |
Collapse
|
32
|
Prokopec SD, Viluksela M, Miettinen HM, Boutros PC, Pohjanvirta R. Transgenerational epigenetic and transcriptomic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure in rat. Arch Toxicol 2020; 94:1613-1624. [PMID: 32277265 DOI: 10.1007/s00204-020-02730-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/26/2020] [Indexed: 12/30/2022]
Abstract
In rats, direct exposure to TCDD causes myriad toxicities. Exposed rats experience hepatotoxicity, wasting syndrome and immune suppression, amongst others. "Inherited exposure", as occurs in the F3 generation of directly exposed F0 animals, has also been shown to cause toxicity: both male and female F3 rats demonstrate an increased incidence of adult onset disease, females also display reproductive abnormalities and increased incidence of ovarian diseases while males show increased incidence of kidney disease and an altered sperm epigenome. Here, we explore the hepatic transcriptomic profile of male and female F3 Sprague-Dawley rats bred through the paternal germ line from F0 dams exposed to a single dose of TCDD (0, 30, 100, 300 or 1000 ng/kg body weight) by oral gavage. We hypothesize that RNA transcripts with altered abundance in livers of unexposed F3 progeny of treated F0 Sprague-Dawley rats may result from epigenetic modifications to the genome. We further survey patterns of differential methylation within male F3 rat testis. Female F3 rats demonstrated more TCDD-mediated hepatic transcriptomic changes than males, with differences primarily in the lowest dose group. In testis from male F3 rats, multiple olfactory receptors displayed patterns of differential methylation. Hypermethylation of Egfr and Mc5r among testes from TCDD lineage rats was observed, but without corresponding changes in hepatic mRNA abundance. Further studies examining these differences in other tissue types are warranted.
Collapse
Affiliation(s)
| | - Matti Viluksela
- Department of Environmental and Biological Sciences, School of Pharmacy (Toxicology), University of Eastern Finland, Kuopio, Finland.,Environmental Health Unit, Finnish Institute for Health and Welfare (THL), Kuopio, Finland
| | - Hanna M Miettinen
- Environmental Health Unit, Finnish Institute for Health and Welfare (THL), Kuopio, Finland.,A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, Canada. .,Department of Pharmacology and Toxicology, Universally of Toronto, Toronto, Canada. .,Department of Human Genetics, University of California, Los Angeles, 12-109 CHS, 10833 Le Conte Avenue, Los Angeles, CA, 90095, USA. .,Department of Urology, University of California, Los Angeles, Los Angeles, USA. .,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, USA. .,Institute for Precision Health, University of California, Los Angeles, Los Angeles, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, USA.
| | - Raimo Pohjanvirta
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, 00014, Helsinki, Finland.
| |
Collapse
|
33
|
Shan Q, Li H, Chen N, Qu F, Guo J. Understanding the Multiple Effects of PCBs on Lipid Metabolism. Diabetes Metab Syndr Obes 2020; 13:3691-3702. [PMID: 33116719 PMCID: PMC7568599 DOI: 10.2147/dmso.s264851] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/19/2020] [Indexed: 12/28/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) are a typical class of environmental contaminants recently shown to be metabolism-disrupting chemicals. Lipids are a highly complex group of biomolecules that not only form the structural basis of biofilms but also act as signaling molecules and energy sources. Lipid metabolic disorders contribute to multiple diseases, including obesity, diabetes, fatty liver, and metabolic syndromes. Although previous literature has reported that PCBs can affect lipid metabolism, including lipid synthesis, uptake, and elimination, few systematic summaries of the detailed process of lipid metabolism caused by PCB exposure have been published. Lipid metabolic processes involve many molecules; however, the key factors that are sensitive to PCB exposure have not been fully clarified. Here, we summarize the recent developments in PCB research with a focus on biomarkers of lipid metabolic disorders related to environmental exposures.
Collapse
Affiliation(s)
- Qiuli Shan
- College of Biological Science and Technology, University of Jinan, Jinan250022, People’s Republic of China
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing100085, People’s Republic of China
- Correspondence: Qiuli Shan Email
| | - Hongmei Li
- College of Biological Science and Technology, University of Jinan, Jinan250022, People’s Republic of China
| | - Ningning Chen
- College of Biological Science and Technology, University of Jinan, Jinan250022, People’s Republic of China
| | - Fan Qu
- College of Biological Science and Technology, University of Jinan, Jinan250022, People’s Republic of China
| | - Jing Guo
- College of Biological Science and Technology, University of Jinan, Jinan250022, People’s Republic of China
| |
Collapse
|
34
|
Ronis MJ, Watt J, Pulliam CF, Williams AE, Alund AW, Haque E, Gadupudi GS, Robertson LW. Skeletal toxicity resulting from exposure of growing male rats to coplanar PCB 126 is associated with disruption of calcium homeostasis and the GH-IGF-1 axis and direct effects on bone formation. Arch Toxicol 2019; 94:389-399. [PMID: 31820026 DOI: 10.1007/s00204-019-02645-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Skeletal toxicity has been reported following exposure to polychlorinated biphenyl (PCB) mixtures. However, molecular mechanisms remain poorly understood. We exposed groups of male 4-5-week-old Sprague-Dawley rats to 3,3', 4, 4', 5-pentachlorobiphenyl (PCB 126), a dioxin-like coplanar PCB congener by a single i.p. injection of 5 µmol/kg in soy oil vehicle or vehicle alone. After 4 weeks, rats were euthanized. PCB exposure resulted in hypocalcemia (P < 0.05) and significant increases in serum PTH without changes in serum phosphorous. Hyperparathyroidism was accompanied by increased expression of mRNAs of vitamin D3 metabolizing cytochrome P450 enzymes CYP27B1 and CYP24 in the kidney (P < 0.05). PCB exposure also reduced body weight, serum IGF-1, and hepatic expression of mRNAs encoding the male-specific GH-pattern-regulated CYP2C11 and CYP3A2 relative to controls (P < 0.05). PCB exposure reduced long bone length, diameter, and surface area, but increased trabecular thickness and volume (P < 0.05). Serum osteocalcin (P < 0.05), a marker and a regulator of bone formation, was reduced, but PCB exposure had no effect on the bone resorption marker RatLaps. Exposure of human intestinal Caco-2 cells to 10-100 nM PCB 126 in the presence of vitamin D3 resulted in inhibition of mRNAs for the calcium transporters TRPV6 and PMCA1b (P < 0.05). In addition, PCB 126 suppressed osteoblastogenesis in primary bone marrow mesenchymal stem cell cultures which was blunted by the AhR antagonist CH-223191. These data provide novel evidence that skeletal toxicity after exposure to PCB 126 is a result of disruption of calcium homeostasis and the GH-IGF-1 axis, and involves direct AhR-mediated effects on bone formation.
Collapse
Affiliation(s)
- Martin J Ronis
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center-New Orleans, 1901 Perdido Str., New Orleans, LA, 70112, USA.
| | - James Watt
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center-New Orleans, 1901 Perdido Str., New Orleans, LA, 70112, USA
| | - Casey F Pulliam
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center-New Orleans, 1901 Perdido Str., New Orleans, LA, 70112, USA
| | - Ashlee E Williams
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center-New Orleans, 1901 Perdido Str., New Orleans, LA, 70112, USA
| | - Alexander W Alund
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ezazul Haque
- IDGP in Human Toxicology and Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, USA
| | - Gopi S Gadupudi
- IDGP in Human Toxicology and Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, USA
| | - Larry W Robertson
- IDGP in Human Toxicology and Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
35
|
Rawls KD, Blais EM, Dougherty BV, Vinnakota KC, Pannala VR, Wallqvist A, Kolling GL, Papin JA. Genome-Scale Characterization of Toxicity-Induced Metabolic Alterations in Primary Hepatocytes. Toxicol Sci 2019; 172:279-291. [PMID: 31501904 PMCID: PMC6876259 DOI: 10.1093/toxsci/kfz197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Context-specific GEnome-scale metabolic Network REconstructions (GENREs) provide a means to understand cellular metabolism at a deeper level of physiological detail. Here, we use transcriptomics data from chemically-exposed rat hepatocytes to constrain a GENRE of rat hepatocyte metabolism and predict biomarkers of liver toxicity using the Transcriptionally Inferred Metabolic Biomarker Response algorithm. We profiled alterations in cellular hepatocyte metabolism following in vitro exposure to four toxicants (acetaminophen, carbon tetrachloride, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six hour. TIMBR predictions were compared with paired fresh and spent media metabolomics data from the same exposure conditions. Agreement between computational model predictions and experimental data led to the identification of specific metabolites and thus metabolic pathways associated with toxicant exposure. Here, we identified changes in the TCA metabolites citrate and alpha-ketoglutarate along with changes in carbohydrate metabolism and interruptions in ATP production and the TCA Cycle. Where predictions and experimental data disagreed, we identified testable hypotheses to reconcile differences between the model predictions and experimental data. The presented pipeline for using paired transcriptomics and metabolomics data provides a framework for interrogating multiple omics datasets to generate mechanistic insight of metabolic changes associated with toxicological responses.
Collapse
Affiliation(s)
- Kristopher D Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
| | - Edik M Blais
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
| | - Bonnie V Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
| | - Kalyan C Vinnakota
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland 20817
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland 21702
| | - Venkat R Pannala
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland 20817
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland 21702
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland 21702
| | - Glynis L Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
- Department of Medicine, Division of Infectious Diseases and International Health
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
36
|
Comparative toxicoproteogenomics of mouse and rat liver identifies TCDD-resistance genes. Arch Toxicol 2019; 93:2961-2978. [DOI: 10.1007/s00204-019-02560-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022]
|
37
|
Viluksela M, Pohjanvirta R. Multigenerational and Transgenerational Effects of Dioxins. Int J Mol Sci 2019; 20:E2947. [PMID: 31212893 PMCID: PMC6627869 DOI: 10.3390/ijms20122947] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/12/2022] Open
Abstract
Dioxins are ubiquitous and persistent environmental contaminants whose background levels are still reason for concern. There is mounting evidence from both epidemiological and experimental studies that paternal exposure to the most potent congener of dioxins, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), can lower the male/female ratio of offspring. Moreover, in laboratory rodents and zebrafish, TCDD exposure of parent animals has been reported to result in reduced reproductive performance along with other adverse effects in subsequent generations, foremost through the paternal but also via the maternal germline. These impacts have been accompanied by epigenetic alterations in placenta and/or sperm cells, including changes in methylation patterns of imprinted genes. Here, we review recent key studies in this field with an attempt to provide an up-to-date picture of the present state of knowledge to the reader. These studies provide biological plausibility for the potential of dioxin exposure at a critical time-window to induce epigenetic alterations across multiple generations and the significance of aryl hydrocarbon receptor (AHR) in mediating these effects. Currently available data do not allow to accurately estimate the human health implications of these findings, although epidemiological evidence on lowered male/female ratio suggests that this effect may take place at realistic human exposure levels.
Collapse
Affiliation(s)
- Matti Viluksela
- School of Pharmacy and Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
- Environmental Health Unit, National Institute for Health and Welfare, P.O. Box 95, FI-70701 Kuopio, Finland.
| | - Raimo Pohjanvirta
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| |
Collapse
|
38
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Fürst P, Håkansson H, Halldorsson T, Lundebye AK, Pohjanvirta R, Rylander L, Smith A, van Loveren H, Waalkens-Berendsen I, Zeilmaker M, Binaglia M, Gómez Ruiz JÁ, Horváth Z, Christoph E, Ciccolallo L, Ramos Bordajandi L, Steinkellner H, Hoogenboom LR. Risk for animal and human health related to the presence of dioxins and dioxin-like PCBs in feed and food. EFSA J 2018; 16:e05333. [PMID: 32625737 PMCID: PMC7009407 DOI: 10.2903/j.efsa.2018.5333] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The European Commission asked EFSA for a scientific opinion on the risks for animal and human health related to the presence of dioxins (PCDD/Fs) and DL-PCBs in feed and food. The data from experimental animal and epidemiological studies were reviewed and it was decided to base the human risk assessment on effects observed in humans and to use animal data as supportive evidence. The critical effect was on semen quality, following pre- and postnatal exposure. The critical study showed a NOAEL of 7.0 pg WHO2005-TEQ/g fat in blood sampled at age 9 years based on PCDD/F-TEQs. No association was observed when including DL-PCB-TEQs. Using toxicokinetic modelling and taking into account the exposure from breastfeeding and a twofold higher intake during childhood, it was estimated that daily exposure in adolescents and adults should be below 0.25 pg TEQ/kg bw/day. The CONTAM Panel established a TWI of 2 pg TEQ/kg bw/week. With occurrence and consumption data from European countries, the mean and P95 intake of total TEQ by Adolescents, Adults, Elderly and Very Elderly varied between, respectively, 2.1 to 10.5, and 5.3 to 30.4 pg TEQ/kg bw/week, implying a considerable exceedance of the TWI. Toddlers and Other Children showed a higher exposure than older age groups, but this was accounted for when deriving the TWI. Exposure to PCDD/F-TEQ only was on average 2.4- and 2.7-fold lower for mean and P95 exposure than for total TEQ. PCDD/Fs and DL-PCBs are transferred to milk and eggs, and accumulate in fatty tissues and liver. Transfer rates and bioconcentration factors were identified for various species. The CONTAM Panel was not able to identify reference values in most farm and companion animals with the exception of NOAELs for mink, chicken and some fish species. The estimated exposure from feed for these species does not imply a risk.
Collapse
|
39
|
Hutin D, Tamblyn L, Gomez A, Grimaldi G, Soedling H, Cho T, Ahmed S, Lucas C, Kanduri C, Grant DM, Matthews J. Hepatocyte-Specific Deletion of TIPARP, a Negative Regulator of the Aryl Hydrocarbon Receptor, Is Sufficient to Increase Sensitivity to Dioxin-Induced Wasting Syndrome. Toxicol Sci 2018; 165:347-360. [PMID: 29873790 PMCID: PMC6154274 DOI: 10.1093/toxsci/kfy136] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) mediates the toxic effects of dioxin (2, 3, 7, 8-tetrachlorodibenzo-p-dioxin; TCDD), which includes thymic atrophy, steatohepatitis, and a lethal wasting syndrome in laboratory rodents. Although the mechanisms of dioxin toxicity remain unknown, AHR signaling in hepatocytes is necessary for dioxin-induced liver toxicity. We previously reported that loss of TCDD-inducible poly(adenosine diphosphate [ADP]-ribose) polymerase (TIPARP/PARP7/ARTD14), an AHR target gene and mono-ADP-ribosyltransferase, increases the sensitivity of mice to dioxin-induced toxicities. To test the hypothesis that TIPARP is a negative regulator of AHR signaling in hepatocytes, we generated Tiparpfl/fl mice in which exon 3 of Tiparp is flanked by loxP sites, followed by Cre-lox technology to create hepatocyte-specific (Tiparpfl/flCreAlb) and whole-body (Tiparpfl/flCreCMV; TiparpEx3-/-) Tiparp null mice. Tiparpfl/flCreAlb and TiparpEx3-/- mice given a single injection of 10 μg/kg dioxin did not survive beyond days 7 and 9, respectively, while all Tiparp+/+ mice survived the 30-day treatment. Dioxin-exposed Tiparpfl/flCreAlb and TiparpEx3-/- mice had increased steatohepatitis and hepatotoxicity as indicated by greater staining of neutral lipids and serum alanine aminotransferase activity than similarly treated wild-type mice. Tiparpfl/flCreAlb and TiparpEx3-/- mice exhibited augmented AHR signaling, denoted by increased dioxin-induced gene expression. Metabolomic studies revealed alterations in lipid and amino acid metabolism in liver extracts from Tiparpfl/flCreAlb mice compared with wild-type mice. Taken together, these data illustrate that TIPARP is an important negative regulator of AHR activity, and that its specific loss in hepatocytes is sufficient to increase sensitivity to dioxin-induced steatohepatitis and lethality.
Collapse
Affiliation(s)
- David Hutin
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Laura Tamblyn
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Alvin Gomez
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Giulia Grimaldi
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Helen Soedling
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Tiffany Cho
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Shaimaa Ahmed
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Christin Lucas
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Chakravarthi Kanduri
- Department of Informatics, Jebsen Centre of Excellence for Celiac Disease Research, University of Oslo, Oslo, Norway
| | - Denis M Grant
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Jason Matthews
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Safe S, Han H, Goldsby J, Mohankumar K, Chapkin RS. Aryl Hydrocarbon Receptor (AhR) Ligands as Selective AhR Modulators: Genomic Studies. CURRENT OPINION IN TOXICOLOGY 2018; 11-12:10-20. [PMID: 31453421 PMCID: PMC6709982 DOI: 10.1016/j.cotox.2018.11.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aryl hydrocarbon receptor (AhR) binds structurally diverse ligands that vary from the environmental toxicant 2,3,7,8-tetrachlorodibenzo-B-dioxin (TCDD) to AhR- active pharmaceuticals and health-promoting phytochemicals. There are remarkable differences in the toxicity of TCDD and related halogenated aromatics (HAs) vs. health promoting AhR ligands, and genomic analysis shows that even among the toxic HAs, there are differences in their regulation of genes and pathways. Thus, like ligands for other receptors, AhR ligands are selective AhR modulators (SAhRMs) which exhibit variable tissue-, organ- and species-specific genomic and functional activities.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology
| | - Huajun Han
- Department of Biochemistry & Biophysics
- Department of Nutrition & Food Science
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX, USA
| | - Jennifer Goldsby
- Department of Nutrition & Food Science
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX, USA
| | | | - Robert S. Chapkin
- Department of Biochemistry & Biophysics
- Department of Nutrition & Food Science
- Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX, USA
| |
Collapse
|
41
|
Andersen ME, Pendse SN, Black MB, McMullen PD. Application of transcriptomic data, visualization tools and bioinformatics resources for informing mode of action. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2018.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
42
|
Gadupudi GS, Elser BA, Sandgruber FA, Li X, Gibson-Corley KN, Robertson LW. PCB126 Inhibits the Activation of AMPK-CREB Signal Transduction Required for Energy Sensing in Liver. Toxicol Sci 2018; 163:440-453. [PMID: 29474705 PMCID: PMC5974782 DOI: 10.1093/toxsci/kfy041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
3,3',4,4',5-pentachlorobiphenyl (PCB126), a dioxin-like PCB, elicits toxicity through a wide array of noncarcinogenic effects, including metabolic syndrome, wasting, and nonalcoholic fatty-liver disease. Previously, we reported decreases in the transcription of several enzymes involved in gluconeogenesis, before the early onset of lipid accumulation. Hence, this study was aimed at understanding the impact of resultant decreases gluconeogenic enzymes on growth, weight, and metabolism in the liver, upon extended exposure. Male Sprague Dawley rats (75-100 g), fed a defined AIN-93G diet, were injected (ip) with single dose of soy oil (5 ml/kg body weight; n = 14) or PCB126 (5 µmol/kg; n = 15), 28 days, prior euthanasia. A subset of rats from each group were fasted for 12 h (vehicle [n = 6] and PCB126 [n = 4]). Rats only showed significant weight loss between days 14 and 28 (p < .05) and some mortality (p = .0413). As in our previous studies, the expression levels of enzymes involved in gluconeogenesis (Pepck-c, G6Pase, Sds, Pc, and Ldh-A) and glycogenolysis (Pygl) were strongly downregulated. The decreased expression of these enzymes in PCB126-treated rats after a 12 h fast decreased hepatic glucose production from glycogen and gluconeogenic substrates, exacerbating the hypoglycemia. Additionally, PCB126 caused hepatic steatosis and decreased the expression of the transcription factor Pparα and its targets, necessary for fatty-acid oxidation. The observed metabolic disruption across multiple branches of fasting metabolism resulted from inhibition in the activation of enzyme AMPK and transcription factor CREB signaling, necessary for "sensing" energy-deprivation and the induction of enzymes that respond to the PCB126 triggered fuel crisis in liver.
Collapse
Affiliation(s)
- Gopi S Gadupudi
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, Iowa
- Department of Occupational and Environmental Health, College of Public Health
| | - Benjamin A Elser
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, Iowa
- Department of Occupational and Environmental Health, College of Public Health
| | - Fabian A Sandgruber
- Department of Occupational and Environmental Health, College of Public Health
| | - Xueshu Li
- Department of Occupational and Environmental Health, College of Public Health
| | | | - Larry W Robertson
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, Iowa
- Department of Occupational and Environmental Health, College of Public Health
| |
Collapse
|
43
|
Wang F, Zhang R, Shi S, Hankinson O. The Effect of Aromatic Hydrocarbon Receptor on the Phenotype of the Hepa 1c1c7 Murine Hepatoma Cells in the Absence of Dioxin. GENE REGULATION AND SYSTEMS BIOLOGY 2017. [DOI: 10.1177/117762500700100006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aromatic hydrocarbon receptor (AhR) mediates biological responses to certain exogenous ligands, such as the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and has also been demonstrated to modulate the cell cycle and differentiated state of several cell lines independently of exogenous ligands. In this study, we used DNA micorarray analysis to elucidate the profile of genes responsive to the expression of unliganded AhR by re-introducing AhR into an AhR-deficient mouse derivative (c19) of the mouse hepatoma cell line Hepa1c1c7. 22 gene products were up-regulated and 8 were down-regulated two-fold or more in c19 cells infected with a retroviral vector expressing mouse AhR. Surprisingly, expression of genes involved in cell proliferation or differentiation were not affected by introduction of AhR. AhR also did not restore expression of the albumin gene in c19 cells. Introduction of AhR into c12, a similar AhR-defective mouse hepatoma cell line, also did not restore albumin expression, and furthermore, did not lead to changes in cellular morphology or cell cycle parameters. These observations fail to support the notion that unliganded AhR regulates proliferation and differentiation of liver-derived cells.
Collapse
Affiliation(s)
- Feng Wang
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, U.S.A
| | - Ruixue Zhang
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, U.S.A
| | - Shengli Shi
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, U.S.A
| | - Oliver Hankinson
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, U.S.A
- Molecular Biology Institute, University of California at Los Angeles
| |
Collapse
|
44
|
LaRocca JL, Rasoulpour RJ, Gollapudi BB, Eisenbrandt DL, Murphy LA, LeBaron MJ. Integration of novel approaches demonstrates simultaneous metabolic inactivation and CAR-mediated hepatocarcinogenesis of a nitrification inhibitor. Toxicol Rep 2017; 4:586-597. [PMID: 29159133 PMCID: PMC5684092 DOI: 10.1016/j.toxrep.2017.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 11/29/2022] Open
Abstract
Nitrapyrin, a nitrification inhibitor, produces liver tumors in mice at high doses. Several experiments were performed to investigate molecular, cellular, and apical endpoints to define the key events leading to the tumor formation. These data support a mode-of-action (MoA) characterized by constitutive androstane receptor (CAR) nuclear receptor activation, increased hepatocellular proliferation leading to hepatocellular foci and tumor formation. Specifically, nitrapyrin induced a dose-related increase in the Cyp2b10/CAR-associated transcript and protein. Interestingly, the corresponding enzyme activity (7-pentoxyresorufin-O-dealkylase (PROD) was not enhanced due to nitrapyrin-mediated suicide inhibition of PROD activity. Nitrapyrin exposure elicited a clear dose-responsive increase in hepatocellular proliferation in wild-type mice, but not in CAR knock-out mice, informing that CAR activation is an obligatory key event in this test material-induced hepatocarcinogenesis. Furthermore, nitrapyrin exposure induced a clear, concentration-responsive increase in cell proliferation in mouse, but not human, hepatocytes in vitro. Evaluation of the data from repeat dose and MoA studies by the Bradford Hill criteria and a Human Relevance Framework (HRF) suggested that nitrapyrin-induced mouse liver tumors are not relevant to human health risk assessment because of qualitative differences between these two species.
Collapse
Affiliation(s)
| | | | - B. Bhaskar Gollapudi
- The Dow Chemical Company, Toxicology and Environmental Research & Consulting, Midland, MI, USA
| | | | - Lynea A. Murphy
- The Dow Chemical Company, Toxicology and Environmental Research & Consulting, Midland, MI, USA
| | - Matthew J. LeBaron
- The Dow Chemical Company, Toxicology and Environmental Research & Consulting, Midland, MI, USA
| |
Collapse
|
45
|
Diani-Moore S, Shoots J, Singh R, Zuk JB, Rifkind AB. NAD + loss, a new player in AhR biology: prevention of thymus atrophy and hepatosteatosis by NAD + repletion. Sci Rep 2017; 7:2268. [PMID: 28536482 PMCID: PMC5442136 DOI: 10.1038/s41598-017-02332-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/18/2017] [Indexed: 01/06/2023] Open
Abstract
Dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) is a carcinogenic and highly toxic industrial byproduct that persists in the environment and produces a pleiotropic toxicity syndrome across vertebrate species that includes wasting, hepatosteatosis, and thymus atrophy. Dioxin toxicities require binding and activation of the aryl hydrocarbon receptor (AhR), a ligand activated transcription factor. However, after nearly 50 years of study, it remains unknown how AhR activation by dioxin produces toxic effects. Here, using the chick embryo close to hatching, a well-accepted model for dioxin toxicity, we identify NAD+ loss through PARP activation as a novel unifying mechanism for diverse effects of dioxin in vivo. We show that NAD+ loss is attributable to increased PARP activity in thymus and liver, as cotreatment with dioxin and the PARP inhibitor PJ34 increased NAD+ levels and prevented both thymus atrophy and hepatosteatosis. Our findings additionally support a role for decreased NAD+ dependent Sirt6 activity in mediating dioxin toxicity following PARP activation. Strikingly, treatment in vivo with the NAD+ repleting agent nicotinamide, a form of vitamin B3, prevented thymus atrophy and hepatosteatosis by dioxin and increased sirtuin activity, providing a therapeutic approach for preventing dioxin toxicities in vivo.
Collapse
Affiliation(s)
- Silvia Diani-Moore
- Department of Pharmacology and Pharmacology PhD Program, Weill Cornell Medicine, 1300 York Avenue, NY, NY, 10021, USA
| | - Jenny Shoots
- Department of Pharmacology and Pharmacology PhD Program, Weill Cornell Medicine, 1300 York Avenue, NY, NY, 10021, USA
| | - Rubi Singh
- Department of Pharmacology and Pharmacology PhD Program, Weill Cornell Medicine, 1300 York Avenue, NY, NY, 10021, USA
| | - Joshua B Zuk
- Department of Pharmacology and Pharmacology PhD Program, Weill Cornell Medicine, 1300 York Avenue, NY, NY, 10021, USA
| | - Arleen B Rifkind
- Department of Pharmacology and Pharmacology PhD Program, Weill Cornell Medicine, 1300 York Avenue, NY, NY, 10021, USA.
| |
Collapse
|
46
|
Fader KA, Zacharewski TR. Beyond the Aryl Hydrocarbon Receptor: Pathway Interactions in the Hepatotoxicity of 2,3,7,8-Tetrachlorodibenzo- p-dioxin and Related Compounds. CURRENT OPINION IN TOXICOLOGY 2017; 2:36-41. [PMID: 28948239 PMCID: PMC5609723 DOI: 10.1016/j.cotox.2017.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is the prototypical ligand for a group of environmental halogenated aromatic hydrocarbon contaminants which elicit hepatotoxicity and other toxic responses through activation of the aryl hydrocarbon receptor (AhR). Despite the conservation of the AhR and its signaling pathway, TCDD-elicited differential gene expression networks are species-specific, consistent with differences in sensitivity and toxic responses between species. This review integrates gene expression studies with complementary phenotypic analyses (e.g., metabolomics, clinical biochemistry, and histopathology) to elucidate the pathways through which TCDD and related compounds cause hepatotoxicity beyond AhR activation. We propose that AhR-mediated toxicity is a collective response to the cumulative burden of metabolic reprogramming across multiple pathways. Consequently, nutrition, health status, and genetic background establish the basis for differences in sensitivity and predisposition to adverse outcomes between species, sub-populations, tissues, and cells.
Collapse
Affiliation(s)
- Kelly A. Fader
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824
| | - Timothy R. Zacharewski
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824
| |
Collapse
|
47
|
Nguyen HT, Tsuchiya MCL, Yoo J, Iida M, Agusa T, Hirano M, Kim EY, Miyazaki T, Nose M, Iwata H. Strain differences in the proteome of dioxin-sensitive and dioxin-resistant mice treated with 2,3,7,8-tetrabromodibenzo-p-dioxin. Arch Toxicol 2016; 91:1763-1782. [PMID: 27604104 DOI: 10.1007/s00204-016-1834-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/24/2016] [Indexed: 11/28/2022]
Abstract
Dioxins cause various toxic effects through the aryl hydrocarbon receptor (AHR) in vertebrates, with dramatic species and strain differences in susceptibility. Although inbred mouse strains C3H/HeJ-lpr/lpr (C3H/lpr) and MRL/MpJ-lpr/lpr (MRL/lpr) are known as dioxin-sensitive and dioxin-resistant mice, respectively, the molecular mechanism underlying this difference remains unclear. The difference in the hepatic proteome of the two mouse strains treated with vehicle or 2,3,7,8-tetrabromodibenzo-p-dioxin (TBDD) was investigated by a proteomic approach of two-dimensional electrophoresis (2-DE) coupled with matrix-assisted laser desorption/ionization time-of-flight/time-of-flight tandem mass spectrometry (MALDI-TOF/TOF). To confirm the strain-difference in response to TBDD treatment, cytochrome P450 (CYP) 1A1 and 1A2 protein levels were measured in both strains. A dose of 10 µg/kg body weight of TBDD induced hepatic CYP1A1 and CYP1A2 expression in both strains, but the expression levels of both CYP1A proteins were higher in C3H/lpr mice than in MRL/lpr mice, supporting that C3H/lpr mice are more sensitive to dioxins than MRL/lpr mice. Proteins that were more induced or suppressed by TBDD treatment in C3H/lpr mice were successfully identified by 2-DE and MALDI-TOF/TOF, including proteins responsible for AHR activation through production of endogenous ligands such as aspartate aminotransferase, indolethylamine N-methyltransferase, and aldehyde dehydrogenases, as well as proteins reducing oxidative stress, such as superoxide dismutase and peroxiredoxins. Taken together, our results provide insights into the molecular mechanism underlying the high dioxin susceptibility of the C3H/lpr strain, in which AHR activation by TBDD is more prompted by the production of endogenous ligands, but the adaptation to oxidative stress is also acquired.
Collapse
Affiliation(s)
- Hoa Thanh Nguyen
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan
| | - Maria Claret Lauan Tsuchiya
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan.,Institute of Biological Sciences, University of the Philippines Los Baños, Laguna, Philippines
| | - Jean Yoo
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan
| | - Midori Iida
- Graduate School of Computer Science and System Engineering, Kyushu Institute of Technology, Iizuka, 820-0067, Japan
| | - Tetsuro Agusa
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan.,Graduate School of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, 862-8502, Japan
| | - Masashi Hirano
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Seoul, 130-701, Korea
| | | | - Masato Nose
- Institute for Promotion of Advanced Science and Technology, Ehime University, Matsuyama, 790-8577, Japan
| | - Hisato Iwata
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan.
| |
Collapse
|
48
|
Jaeger C, Tischkau SA. Role of Aryl Hydrocarbon Receptor in Circadian Clock Disruption and Metabolic Dysfunction. ENVIRONMENTAL HEALTH INSIGHTS 2016; 10:133-141. [PMID: 27559298 PMCID: PMC4990151 DOI: 10.4137/ehi.s38343] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 06/01/2023]
Abstract
The prevalence of metabolic syndrome, a clustering of three or more risk factors that include abdominal obesity, increased blood pressure, and high levels of glucose, triglycerides, and high-density lipoproteins, has reached dangerous and costly levels worldwide. Increases in morbidity and mortality result from a combination of factors that promote altered glucose metabolism, insulin resistance, and metabolic dysfunction. Although diet and exercise are commonly touted as important determinants in the development of metabolic dysfunction, other environmental factors, including circadian clock disruption and activation of the aryl hydrocarbon receptor (AhR) by dietary or other environmental sources, must also be considered. AhR binds a range of ligands, which prompts protein-protein interactions with other Per-Arnt-Sim (PAS)-domain-containing proteins and subsequent transcriptional activity. This review focuses on the reciprocal crosstalk between the activated AhR and the molecular circadian clock. AhR exhibits a rhythmic expression and time-dependent sensitivity to activation by AhR agonists. Conversely, AhR activation influences the amplitude and phase of expression of circadian clock genes, hormones, and the behavioral responses of the clock system to changes in environmental illumination. Both the clock and AhR status and activation play significant and underappreciated roles in metabolic homeostasis. This review highlights the state of knowledge regarding how AhR may act together with the circadian clock to influence energy metabolism. Understanding the variety of AhR-dependent mechanisms, including its interactions with the circadian timing system that promote metabolic dysfunction, reveals new targets of interest for maintenance of healthy metabolism.
Collapse
|
49
|
Yin J, Sheng B, Qiu Y, Yang K, Xiao W, Yang H. Role of AhR in positive regulation of cell proliferation and survival. Cell Prolif 2016; 49:554-60. [PMID: 27523394 DOI: 10.1111/cpr.12282] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/29/2016] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is an important nuclear transcription factor that is best known for mediating toxic responses by adjusting numbers of metabolism-related enzymes, including CYP1A1 and CYP1B1. Previous findings have revealed that, in addition to negatively regulating cell proliferation and survival, AhR may also positively regulate these pathways. Here, we review these findings and summarize distinct mechanisms by which AhR promotes cell proliferation and survival, including modulation of receptor expression, growth factor signalling and apoptosis, regulating the cell cycle and promoting cytokine expression. This review will aid better understanding the role of AhR in positive regulation of cell proliferation and survival.
Collapse
Affiliation(s)
- Jiuheng Yin
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Baifa Sheng
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Kunqiu Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
50
|
Watson JD, Prokopec SD, Smith AB, Okey AB, Pohjanvirta R, Boutros PC. 2,3,7,8 Tetrachlorodibenzo-p-dioxin-induced RNA abundance changes identify Ackr3, Col18a1, Cyb5a and Glud1 as candidate mediators of toxicity. Arch Toxicol 2016; 91:325-338. [PMID: 27136898 PMCID: PMC5225275 DOI: 10.1007/s00204-016-1720-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/20/2016] [Indexed: 12/11/2022]
Abstract
2,3,7,8 Tetrachlorodibenzo-p-dioxin (TCDD) is an aromatic, long-lived environmental contaminant. While the pathogenesis of TCDD-induced toxicity is poorly understood, it has been shown that the aryl hydrocarbon receptor (AHR) is required. However, the specific transcriptomic changes that lead to toxic outcomes have not yet been identified. We previously identified a panel of 33 genes that respond to TCDD treatment in two TCDD-sensitive rodent species. To identify genes involved in the onset of hepatic toxicity, we explored 25 of these in-depth using liver from two rat strains: the TCDD-resistant Han/Wistar (H/W) and the TCDD-sensitive Long–Evans (L–E). Time course and dose–response analyses of mRNA abundance following TCDD insult indicate that eight genes are similarly regulated in livers of both strains of rat, suggesting that they are not central to the severe L–E-specific TCDD-induced toxicities. The remaining 17 genes exhibited various divergent mRNA abundances between L–E and H/W strains after TCDD treatment. Several genes displayed a biphasic response where the initial response to TCDD treatment was followed by a secondary response, usually of larger magnitude in L–E liver. This secondary response was most often an exaggeration of the original TCDD-induced response. Only cytochrome b5 type A (microsomal) (Cyb5a) had equivalent TCDD sensitivity to the prototypic AHR-responsive cytochrome P450, family 1, subfamily a, polypeptide 1 (Cyp1a1), while six genes were less sensitive. Four genes showed an early inter-strain difference that was sustained throughout most of the time course (atypical chemokine receptor 3 (Ackr3), collagen, type XVIII, alpha 1 (Col18a1), Cyb5a and glutamate dehydrogenase 1 (Glud1)), and of those genes examined in this study, are most likely to represent genes involved in the pathogenesis of TCDD-induced hepatotoxicity in L–E rats.
Collapse
Affiliation(s)
- John D Watson
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Stephenie D Prokopec
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Ashley B Smith
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Allan B Okey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Raimo Pohjanvirta
- Laboratory of Toxicology, National Institute for Health and Welfare, Kuopio, Finland.,Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Paul C Boutros
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| |
Collapse
|