1
|
Vrzalova A, Vrzal R. Orchestra of ligand-activated transcription factors in the molecular symphony of SERPINE 1 / PAI-1 gene regulation. Biochimie 2025; 228:138-157. [PMID: 39321911 DOI: 10.1016/j.biochi.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Plasminogen activator inhibitor 1 (PAI-1) is a crucial serine protease inhibitor that prevents plasminogen activation by inhibiting tissue- and urokinase-type plasminogen activators (tPA, uPA). PAI-1 is well-known for its role in modulating hemocoagulation or extracellular matrix formation by inhibiting plasmin or matrix metalloproteinases, respectively. PAI-1 is induced by pro-inflammatory cytokines across various tissues, yet its regulation by ligand-activated transcription factors is partly disregarded. Therefore, we have attempted to summarize the current knowledge on the transcriptional regulation of PAI-1 expression by the most relevant xenobiotic and endocrine receptors implicated in modulating PAI-1 levels. This review aims to contribute to the understanding of the specific, often tissue-dependent regulation of PAI-1 and provide insights into the modulation of PAI-1 levels beyond its direct inhibition.
Collapse
Affiliation(s)
- Aneta Vrzalova
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Radim Vrzal
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
2
|
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
|
3
|
Sink WJ, Fling R, Yilmaz A, Nault R, Goniwiecha D, Harkema JR, Graham SF, Zacharewski T. 2,3,7,8-Tetrachlorodibenzo- p-dioxin (TCDD) elicited dose-dependent shifts in the murine urinary metabolome associated with hepatic AHR-mediated differential gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619714. [PMID: 39484576 PMCID: PMC11526911 DOI: 10.1101/2024.10.22.619714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Epidemiological evidence suggests an association between dioxin and dioxin-like compound (DLC) exposure and human liver disease. The prototypical DLC, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), has been shown to induce the progression of reversible hepatic steatosis to steatohepatitis with periportal fibrosis and biliary hyperplasia in mice. Although the effects of TCDD toxicity are mediated by aryl hydrocarbon receptor (AHR) activation, the underlying mechanisms of TCDD-induced hepatotoxicity are unresolved. In the present study, male C57BL/6NCrl mice were gavaged every 4 days for 28 days with 0.03 - 30 μg/kg TCDD and evaluated for liver histopathology and gene expression as well as complementary 1-dimensional proton magnetic resonance (1D- 1H NMR) urinary metabolic profiling. Urinary trimethylamine (TMA), trimethylamine N-oxide (TMAO), and 1-methylnicotinamide (1MN) levels were altered by TCDD at doses ≤ 3 μg/kg; other urinary metabolites, like glycolate, urocanate, and 3-hydroxyisovalerate, were only altered at doses that induced moderate to severe steatohepatitis. Bulk liver RNA-seq data suggested altered urinary metabolites correlated with hepatic differential gene expression corresponding to specific metabolic pathways. In addition to evaluating whether altered urinary metabolites were liver-dependent, published single-nuclear RNA-seq (snRNA-seq), AHR ChIP-seq, and AHR knockout gene expression datasets provide further support for hepatic cell-type and AHR-regulated dependency, respectively. Overall, TCDD-induced liver effects were preceded by and occurred with changes in urinary metabolite levels due to AHR-mediated changes in hepatic gene expression.
Collapse
Affiliation(s)
- Warren J Sink
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| | - Russell Fling
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| | - Ali Yilmaz
- Corewell Health Research Institute, Royal Oak, MI 48073, USA
| | - Rance Nault
- Michigan State University, Department of Pharmacology and Toxicology, East Lansing, MI 48824, USA
| | - Delanie Goniwiecha
- Middlebury College, Neuroscience Faculty, 14 Old Chapel Rd, Middlebury, VT 05753, USA
| | - Jack R Harkema
- Michigan State University, Pathobiology & Diagnostic Investigation, East Lansing, MI, United States of America
| | - Stewart F Graham
- Corewell Health Research Institute, Royal Oak, MI 48073, USA
- Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Timothy Zacharewski
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| |
Collapse
|
4
|
Cholico GN, Nault R, Zacharewski T. Cell-specific AHR-driven differential gene expression in the mouse liver cell following acute TCDD exposure. BMC Genomics 2024; 25:809. [PMID: 39198768 PMCID: PMC11351262 DOI: 10.1186/s12864-024-10730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant that disrupts hepatic function leading to steatotic liver disease (SLD)-like pathologies, such as steatosis, steatohepatitis, and fibrosis. These effects are mediated by the aryl hydrocarbon receptor following changes in gene expression. Although diverse cell types are involved, initial cell-specific changes in gene expression have not been reported. In this study, differential gene expression in hepatic cell types was examined in male C57BL/6 mice gavaged with 30 µg/kg of TCDD using single-nuclei RNA-sequencing. Ten liver cell types were identified with the proportions of most cell types remaining unchanged, except for neutrophils which increased at 72 h. Gene expression suggests TCDD induced genes related to oxidative stress in hepatocytes as early as 2 h. Lipid homeostasis was disrupted in hepatocytes, macrophages, B cells, and T cells, characterized by the induction of genes associated with lipid transport, steroid hormone biosynthesis, and the suppression of β-oxidation, while linoleic acid metabolism was altered in hepatic stellate cells (HSCs), B cells, portal fibroblasts, and plasmacytoid dendritic cells. Pro-fibrogenic processes were also enriched, including the induction retinol metabolism genes in HSCs and the early induction of anti-fibrolysis genes in hepatocytes, endothelial cells, HSCs, and macrophages. Hepatocytes also had gene expression changes consistent with hepatocellular carcinoma. Collectively, these findings underscore the effects of TCDD in initiating SLD-like phenotypes and identified cell-specific gene expression changes related to oxidative stress, steatosis, fibrosis, cell proliferation and the development of HCC.
Collapse
Affiliation(s)
- Giovan N Cholico
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Rance Nault
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
- Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Tim Zacharewski
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
5
|
Hoyeck MP, Angela Ching ME, Basu L, van Allen K, Palaniyandi J, Perera I, Poleo-Giordani E, Hanson AA, Ghorbani P, Fullerton MD, Bruin JE. The aryl hydrocarbon receptor in β-cells mediates the effects of TCDD on glucose homeostasis in mice. Mol Metab 2024; 81:101893. [PMID: 38309623 PMCID: PMC10867573 DOI: 10.1016/j.molmet.2024.101893] [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: 11/13/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
OBJECTIVE Chronic exposure to persistent organic pollutants (POPs) is associated with increased incidence of type 2 diabetes, hyperglycemia, and poor insulin secretion in humans. Dioxins and dioxin-like compounds are a broad class of POPs that exert cellular toxicity through activation of the aryl hydrocarbon receptor (AhR). We previously showed that a single high-dose injection of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, aka dioxin; 20 μg/kg) in vivo reduced fasted and glucose-stimulated plasma insulin levels for up to 6 weeks in male and female mice. TCDD-exposed male mice were also modestly hypoglycemic and had increased insulin sensitivity, whereas TCDD-exposed females were transiently glucose intolerant. Whether these effects are driven by AhR activation in β-cells requires investigation. METHODS We exposed female and male β-cell specific Ahr knockout (βAhrKO) mice and littermate Ins1-Cre genotype controls (βAhrWT) to a single high dose of 20 μg/kg TCDD and tracked the mice for 6 weeks. RESULTS Under baseline conditions, deleting AhR from β-cells caused hypoglycemia in female mice, increased insulin secretion ex vivo in female mouse islets, and promoted modest weight gain in male mice. Importantly, high-dose TCDD exposure impaired glucose homeostasis and β-cell function in βAhrWT mice, but these phenotypes were largely abolished in TCDD-exposed βAhrKO mice. CONCLUSION Our study demonstrates that AhR signaling in β-cells is important for regulating baseline β-cell function in female mice and energy homeostasis in male mice. We also show that β-cell AhR signaling largely mediates the effects of TCDD on glucose homeostasis in both sexes, suggesting that the effects of TCDD on β-cell function and health are driving metabolic phenotypes in peripheral tissues.
Collapse
Affiliation(s)
- Myriam P Hoyeck
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Ma Enrica Angela Ching
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Lahari Basu
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Kyle van Allen
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Jana Palaniyandi
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Ineli Perera
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Emilia Poleo-Giordani
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Antonio A Hanson
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, Ottawa, ON, Canada; Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Jennifer E Bruin
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
6
|
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
|
7
|
Steidemann MM, Liu J, Bayes K, Castro LP, Ferguson-Miller S, LaPres JJ. Evidence for crosstalk between the aryl hydrocarbon receptor and the translocator protein in mouse lung epithelial cells. Exp Cell Res 2023; 429:113617. [PMID: 37172753 PMCID: PMC10330775 DOI: 10.1016/j.yexcr.2023.113617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/07/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
Cellular homeostasis requires the use of multiple environmental sensors that can respond to a variety of endogenous and exogenous compounds. The aryl hydrocarbon receptor (AHR) is classically known as a transcription factor that induces genes that encode drug metabolizing enzymes when bound to toxicants such as 2,3,7,8-tetrachlorodibenzo-ρ-dioxin (TCDD). The receptor has a growing number of putative endogenous ligands, such as tryptophan, cholesterol, and heme metabolites. Many of these compounds are also linked to the translocator protein (TSPO), an outer mitochondrial membrane protein. Given a portion of the cellular pool of the AHR has also been localized to mitochondria and the overlap in putative ligands, we tested the hypothesis that crosstalk exists between the two proteins. CRISPR/Cas9 was used to create knockouts for AHR and TSPO in a mouse lung epithelial cell line (MLE-12). WT, AHR-/-, and TSPO-/- cells were then exposed to AHR ligand (TCDD), TSPO ligand (PK11195), or both and RNA-seq was performed. More mitochondrial-related genes were altered by loss of both AHR and TSPO than would have been expected just by chance. Some of the genes altered included those that encode for components of the electron transport system and the mitochondrial calcium uniporter. Both proteins altered the activity of the other as AHR loss caused the increase of TSPO at both the mRNA and protein level and loss of TSPO significantly increased the expression of classic AHR battery genes after TCDD treatment. This research provides evidence that AHR and TSPO participate in similar pathways that contribute to mitochondrial homeostasis.
Collapse
Affiliation(s)
- Michelle M Steidemann
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, United States
| | - Jian Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, United States
| | - Kalin Bayes
- Department of Integrative Biology, Michigan State University, East Lansing, MI, 48824, United States
| | - Lizbeth P Castro
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, United States; Department of Cell and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States
| | - Shelagh Ferguson-Miller
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, United States
| | - John J LaPres
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, United States; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, United States.
| |
Collapse
|
8
|
Cholico GN, Orlowska K, Fling RR, Sink WJ, Zacharewski NA, Fader KA, Nault R, Zacharewski T. Consequences of reprogramming acetyl-CoA metabolism by 2,3,7,8-tetrachlorodibenzo-p-dioxin in the mouse liver. Sci Rep 2023; 13:4138. [PMID: 36914879 PMCID: PMC10011583 DOI: 10.1038/s41598-023-31087-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant that induces the progression of steatosis to steatohepatitis with fibrosis in mice. Furthermore, TCDD reprograms hepatic metabolism by redirecting glycolytic intermediates while inhibiting lipid metabolism. Here, we examined the effect of TCDD on hepatic acetyl-coenzyme A (acetyl-CoA) and β-hydroxybutyrate levels as well as protein acetylation and β-hydroxybutyrylation. Acetyl-CoA is not only a central metabolite in multiple anabolic and catabolic pathways, but also a substrate used for posttranslational modification of proteins and a surrogate indicator of cellular energy status. Targeted metabolomic analysis revealed a dose-dependent decrease in hepatic acetyl-CoA levels coincident with the phosphorylation of pyruvate dehydrogenase (E1), and the induction of pyruvate dehydrogenase kinase 4 and pyruvate dehydrogenase phosphatase, while repressing ATP citrate lyase and short-chain acyl-CoA synthetase gene expression. In addition, TCDD dose-dependently reduced the levels of hepatic β-hydroxybutyrate and repressed ketone body biosynthesis gene expression. Moreover, levels of total hepatic protein acetylation and β-hydroxybutyrylation were reduced. AMPK phosphorylation was induced consistent with acetyl-CoA serving as a cellular energy status surrogate, yet subsequent targets associated with re-establishing energy homeostasis were not activated. Collectively, TCDD reduced hepatic acetyl-CoA and β-hydroxybutyrate levels eliciting starvation-like conditions despite normal levels of food intake.
Collapse
Affiliation(s)
- Giovan N Cholico
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Karina Orlowska
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Russell R Fling
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
- Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Warren J Sink
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Nicholas A Zacharewski
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
| | - Kelly A Fader
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Rance Nault
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Tim Zacharewski
- Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
9
|
Nault R, Saha S, Bhattacharya S, Sinha S, Maiti T, Zacharewski T. Single-cell transcriptomics shows dose-dependent disruption of hepatic zonation by TCDD in mice. Toxicol Sci 2023; 191:135-148. [PMID: 36222588 PMCID: PMC9887712 DOI: 10.1093/toxsci/kfac109] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) dose-dependently induces the development of hepatic fat accumulation and inflammation with fibrosis in mice initially in the portal region. Conversely, differential gene and protein expression is first detected in the central region. To further investigate cell-specific and spatially resolved dose-dependent changes in gene expression elicited by TCDD, single-nuclei RNA sequencing and spatial transcriptomics were used for livers of male mice gavaged with TCDD every 4 days for 28 days. The proportion of 11 cell (sub)types across 131 613 nuclei dose-dependently changed with 68% of all portal and central hepatocyte nuclei in control mice being overtaken by macrophages following TCDD treatment. We identified 368 (portal fibroblasts) to 1339 (macrophages) differentially expressed genes. Spatial analyses revealed initial loss of portal identity that eventually spanned the entire liver lobule with increasing dose. Induction of R-spondin 3 (Rspo3) and pericentral Apc, suggested dysregulation of the Wnt/β-catenin signaling cascade in zonally resolved steatosis. Collectively, the integrated results suggest disruption of zonation contributes to the pattern of TCDD-elicited NAFLD pathologies.
Collapse
Affiliation(s)
- Rance Nault
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Satabdi Saha
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan 48824, USA
| | - Sudin Bhattacharya
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
- Biomedical Engineering Department, Pharmacology & Toxicology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - Samiran Sinha
- Department of Statistics, Texas A&M University, College Station, Texas 77840, USA
| | - Tapabrata Maiti
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan 48824, USA
| | - Tim Zacharewski
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
10
|
Orlowska K, Fling RR, Nault R, Sink WJ, Schilmiller AL, Zacharewski T. Dioxin-elicited decrease in cobalamin redirects propionyl-CoA metabolism to the β-oxidation-like pathway resulting in acrylyl-CoA conjugate buildup. J Biol Chem 2022; 298:102301. [PMID: 35931118 PMCID: PMC9418907 DOI: 10.1016/j.jbc.2022.102301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant that induces diverse biological and toxic effects, including reprogramming intermediate metabolism, mediated by the aryl hydrocarbon receptor. However, the specific reprogramming effects of TCDD are unclear. Here, we performed targeted LC-MS analysis of hepatic extracts from mice gavaged with TCDD. We detected an increase in S-(2-carboxyethyl)-L-cysteine, a conjugate from the spontaneous reaction between the cysteine sulfhydryl group and highly reactive acrylyl-CoA, an intermediate in the cobalamin (Cbl)-independent β-oxidation-like metabolism of propionyl-CoA. TCDD repressed genes in both the canonical Cbl-dependent carboxylase and the alternate Cbl-independent β-oxidation-like pathways as well as inhibited methylmalonyl-CoA mutase (MUT) at lower doses. Moreover, TCDD decreased serum Cbl levels and hepatic cobalt levels while eliciting negligible effects on gene expression associated with Cbl absorption, transport, trafficking, or derivatization to 5'-deoxy-adenosylcobalamin (AdoCbl), the required MUT cofactor. Additionally, TCDD induced the gene encoding aconitate decarboxylase 1 (Acod1), the enzyme responsible for decarboxylation of cis-aconitate to itaconate, and dose-dependently increased itaconate levels in hepatic extracts. Our results indicate MUT inhibition is consistent with itaconate activation to itaconyl-CoA, a MUT suicide inactivator that forms an adduct with adenosylcobalamin. This adduct in turn inhibits MUT activity and reduces Cbl levels. Collectively, these results suggest the decrease in MUT activity is due to Cbl depletion following TCDD treatment, which redirects propionyl-CoA metabolism to the alternate Cbl-independent β-oxidation-like pathway. The resulting hepatic accumulation of acrylyl-CoA likely contributes to TCDD-elicited hepatotoxicity and the multihit progression of steatosis to steatohepatitis with fibrosis.
Collapse
Affiliation(s)
- Karina Orlowska
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Russ R. Fling
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA,Microbiology & Molecular Genetics, Michigan Sptate University, East Lansing, Michigan, USA
| | - Rance Nault
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Warren J. Sink
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Anthony L. Schilmiller
- Mass Spectrometry and Metabolomics Core, Michigan State University, East Lansing, Michigan, USA
| | - Tim Zacharewski
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
11
|
Cholico GN, Nault R, Zacharewski TR. Genome-Wide ChIPseq Analysis of AhR, COUP-TF, and HNF4 Enrichment in TCDD-Treated Mouse Liver. Int J Mol Sci 2022; 23:1558. [PMID: 35163483 PMCID: PMC8836158 DOI: 10.3390/ijms23031558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor known for mediating the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds. Although the canonical mechanism of AhR activation involves heterodimerization with the aryl hydrocarbon receptor nuclear translocator, other transcriptional regulators that interact with AhR have been identified. Enrichment analysis of motifs in AhR-bound genomic regions implicated co-operation with COUP transcription factor (COUP-TF) and hepatocyte nuclear factor 4 (HNF4). The present study investigated AhR, HNF4α and COUP-TFII genomic binding and effects on gene expression associated with liver-specific function and cell differentiation in response to TCDD. Hepatic ChIPseq data from male C57BL/6 mice at 2 h after oral gavage with 30 µg/kg TCDD were integrated with bulk RNA-sequencing (RNAseq) time-course (2-72 h) and dose-response (0.01-30 µg/kg) datasets to assess putative AhR, HNF4α and COUP-TFII interactions associated with differential gene expression. Functional enrichment analysis of differentially expressed genes (DEGs) identified differential binding enrichment for AhR, COUP-TFII, and HNF4α to regions within liver-specific genes, suggesting intersections associated with the loss of liver-specific functions and hepatocyte differentiation. Analysis found that the repression of liver-specific, HNF4α target and hepatocyte differentiation genes, involved increased AhR and HNF4α binding with decreased COUP-TFII binding. Collectively, these results suggested TCDD-elicited loss of liver-specific functions and markers of hepatocyte differentiation involved interactions between AhR, COUP-TFII and HNF4α.
Collapse
Affiliation(s)
| | | | - Tim R. Zacharewski
- Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; (G.N.C.); (R.N.)
| |
Collapse
|
12
|
Yang Y, Filipovic D, Bhattacharya S. A Negative Feedback Loop and Transcription Factor Cooperation Regulate Zonal Gene Induction by 2, 3, 7, 8-Tetrachlorodibenzo-p-Dioxin in the Mouse Liver. Hepatol Commun 2021; 6:750-764. [PMID: 34726355 PMCID: PMC8948569 DOI: 10.1002/hep4.1848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 10/02/2021] [Accepted: 10/10/2021] [Indexed: 01/04/2023] Open
Abstract
The cytochrome P450 (Cyp) proteins Cyp1A1 and Cyp1A2 are strongly induced in the mouse liver by the potent environmental toxicant 2, 3, 7, 8‐tetrachlorodibenzo‐p‐dioxin (TCDD), acting through the aryl hydrocarbon receptor (AHR). The induction of Cyp1A1 is localized within the centrilobular regions of the mouse liver at low doses of TCDD, progressing to pan‐lobular induction at higher doses. Even without chemical perturbation, metabolic functions and associated genes are basally zonated in the liver lobule along the central‐to‐portal axis. To investigate the mechanistic basis of spatially restricted gene induction by TCDD, we have developed a multiscale computational model of the mouse liver lobule with single‐cell resolution. The spatial location of individual hepatocytes in the model was calibrated from previously published high‐resolution images. A systems biology model of the network of biochemical signaling pathways underlying Cyp1A1 and Cyp1A2 induction was then incorporated into each hepatocyte in the model. Model simulations showed that a negative feedback loop formed by binding of the induced Cyp1A2 protein to TCDD, together with cooperative gene induction by the β‐catenin/AHR/TCDD transcription factor complex and β‐catenin, help produce the spatially localized induction pattern of Cyp1A1. Although endogenous WNT regulates the metabolic zonation of many genes, it was not a driver of zonal Cyp1A1 induction in our model. Conclusion: In this work, we used data‐driven computational modeling to identify the mechanistic basis of zonally restricted gene expression induced by the potent and persistent environmental pollutant TCDD. The multiscale model and derived results clarify the mechanisms of dose‐dependent hepatic gene induction responses to TCDD. Additionally, this work contributes to our broader understanding of spatial gene regulation along the liver lobule.
Collapse
Affiliation(s)
- Yongliang Yang
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.,Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, USA
| | - David Filipovic
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.,Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, USA.,Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.,Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, USA.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
13
|
Rodríguez-Fdez S, Lorenzo-Martín LF, Fabbiano S, Menacho-Márquez M, Sauzeau V, Dosil M, Bustelo XR. New Functions of Vav Family Proteins in Cardiovascular Biology, Skeletal Muscle, and the Nervous System. BIOLOGY 2021; 10:biology10090857. [PMID: 34571735 PMCID: PMC8472352 DOI: 10.3390/biology10090857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary In this review, we provide information on the role of Vav proteins, a group of signaling molecules that act as both Rho GTPase activators and adaptor molecules, in the cardiovascular system, skeletal muscle, and the nervous system. We also describe how these functions impact in other physiological and pathological processes such as sympathoregulation, blood pressure regulation, systemic metabolism, and metabolic syndrome. Abstract Vav proteins act as tyrosine phosphorylation-regulated guanosine nucleotide exchange factors for Rho GTPases and as molecular scaffolds. In mammals, this family of signaling proteins is composed of three members (Vav1, Vav2, Vav3) that work downstream of protein tyrosine kinases in a wide variety of cellular processes. Recent work with genetically modified mouse models has revealed that these proteins play key signaling roles in vascular smooth and skeletal muscle cells, specific neuronal subtypes, and glia cells. These functions, in turn, ensure the proper regulation of blood pressure levels, skeletal muscle mass, axonal wiring, and fiber myelination events as well as systemic metabolic balance. The study of these mice has also led to the discovery of new physiological interconnection among tissues that contribute to the ontogeny and progression of different pathologies such as, for example, hypertension, cardiovascular disease, and metabolic syndrome. Here, we provide an integrated view of all these new Vav family-dependent signaling and physiological functions.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - L. Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Salvatore Fabbiano
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Mauricio Menacho-Márquez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
| | - Vincent Sauzeau
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Institut du Thorax, UMR1087 CNRS 6291, INSERM, Université de Nantes, 44096 Nantes, France
| | - Mercedes Dosil
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-663-194-634
| |
Collapse
|
14
|
Liu Q, Zhang L, Allman EL, Hubbard TD, Murray IA, Hao F, Tian Y, Gui W, Nichols RG, Smith PB, Anitha M, Perdew GH, Patterson AD. The aryl hydrocarbon receptor activates ceramide biosynthesis in mice contributing to hepatic lipogenesis. Toxicology 2021; 458:152831. [PMID: 34097992 DOI: 10.1016/j.tox.2021.152831] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 12/18/2022]
Abstract
Aryl hydrocarbon receptor (AHR) activation via 2,3,7,8-tetrachlorodibenzofuran (TCDF) induces the accumulation of hepatic lipids. Here we report that AHR activation by TCDF (24 μg/kg body weight given orally for five days) induced significant elevation of hepatic lipids including ceramides in mice, was associated with increased expression of key ceramide biosynthetic genes, and increased activity of their respective enzymes. Results from chromatin immunoprecipitation (ChIP), electrophoretic mobility shift assay (EMSA) and cell-based reporter luciferase assays indicated that AHR directly activated the serine palmitoyltransferase long chain base subunit 2 (Sptlc2, encodes serine palmitoyltransferase 2 (SPT2)) gene whose product catalyzes the initial rate-limiting step in de novo sphingolipid biosynthesis. Hepatic ceramide accumulation was further confirmed by mass spectrometry-based lipidomics. Taken together, our results revealed that AHR activation results in the up-regulation of Sptlc2, leading to ceramide accumulation, thus promoting lipogenesis, which can induce hepatic lipid accumulation.
Collapse
Affiliation(s)
- Qing Liu
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Limin Zhang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan, 430071, China
| | - Erik L Allman
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Troy D Hubbard
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Iain A Murray
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Fuhua Hao
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yuan Tian
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Wei Gui
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Robert G Nichols
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Philip B Smith
- Huck Institutes of the Life Sciences, University Park, PA, 16802, USA
| | - Mallappa Anitha
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Gary H Perdew
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Andrew D Patterson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
15
|
Olivero-Verbel J, Harkema JR, Roth RA, Ganey PE. Fenofibrate, a peroxisome proliferator-activated receptor-alpha agonist, blocks steatosis and alters the inflammatory response in a mouse model of inflammation-dioxin interaction. Chem Biol Interact 2021; 345:109521. [PMID: 34052195 DOI: 10.1016/j.cbi.2021.109521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/07/2021] [Accepted: 05/14/2021] [Indexed: 12/01/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (dioxin; TCDD) is an environmental contaminant that elicits a variety of toxic effects, many of which are mediated through activation of the aryl hydrocarbon receptor (AhR). Interaction between AhR and the peroxisome proliferator-activated receptor-alpha (PPAR-α), which regulates fatty acid metabolism, has been suggested. Furthermore, with recognition of the prevalence of inflammatory conditions, there is current interest in the potential for inflammatory stress to modulate the response to environmental agents. The aim of this work was to assess the interaction of TCDD with hepatic inflammation modulated by fenofibrate, a PPAR-α agonist. Female, C57BL/6 mice were treated orally with vehicle or fenofibrate (250 mg/kg) for 13 days, and then were given vehicle or 30 μg/kg TCDD. Four days later, the animals received an i.p. injection of lipopolysaccharide-galactosamine (LPS-GalN) (0.05x107 EU/kg and 500 mg/kg, respectively) to incite inflammation, or saline as vehicle control. After 4 h, the mice were euthanized, and blood and liver samples were collected for analysis. Livers of animals treated with TCDD with or without LPS-GalN had increased lipid deposition, and this effect was blocked by fenofibrate. In TCDD/LPS-GalN-treated mice, fenofibrate caused an increase in plasma activity of alanine aminotransferase, a marker of hepatocellular injury. TCDD reduced LPS-GalN-induced apoptosis, an effect that was prevented by fenofibrate pretreatment. LPS-GalN induced an increase in the concentration of interleukin-6 in plasma and accumulation of neutrophils in liver. TCDD exposure enhanced the former response and inhibited the latter one. These results suggest that fenofibrate counteracts the changes in lipid metabolism induced by TCDD but increases inflammation and liver injury in this model of inflammation-TCDD interaction.
Collapse
Affiliation(s)
- Jesus Olivero-Verbel
- Department of Pharmacology and Toxicology. Michigan State University, East Lansing, MI, USA; Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, University of Cartagena, Cartagena, 130014, Colombia
| | - Jack R Harkema
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, USA
| | - Robert A Roth
- Department of Pharmacology and Toxicology. Michigan State University, East Lansing, MI, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology. Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
16
|
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: 17] [Impact Index Per Article: 4.3] [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
|
17
|
Ye G, Gao H, Zhang X, Liu X, Chen J, Liao X, Zhang H, Huang Q. Aryl hydrocarbon receptor mediates benzo[a]pyrene-induced metabolic reprogramming in human lung epithelial BEAS-2B cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 756:144130. [PMID: 33288249 DOI: 10.1016/j.scitotenv.2020.144130] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/09/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
Polycyclic aromatic hydrocarbon exposure accelerates the initiation and progression of lung cancer through aryl hydrocarbon receptor (AHR) signaling. Metabolic reprogramming is a hallmark of cancer. However, how AHR reprograms metabolism related to the malignant transformation in of benzo[a]pyrene (BaP)-exposed lung cells remains unclear. After confirming that BaP exposure activated AHR signaling and relevant downstream factors and then promoted epithelial-mesenchymal transition, an untargeted metabolomics approach was employed to discover AHR-mediated metabolic reprogramming and potential therapeutic targets in BaP-exposed BEAS-2B cells. We found that 52 metabolites were significantly altered in BaP-exposed BEAS-2B cells and responsive to resveratrol (RSV) intervention. Pathway analysis revealed that 28 and 30 metabolic pathways were significantly altered in response to BaP exposure and RSV intervention, respectively. Notably, levels of most amino acids were significantly decreased, while those of most fatty acids were significantly increased in BaP-exposed BEAS-2B cells, and above changes were abolished by RSV intervention. Besides, levels of amino acids and fatty acids were highly correlated with those of many metabolites and AHR signaling upon BaP exposure and RSV intervention (the absolute values of Pearson correlation coefficients above 0.8). We further discovered a decrease in peroxisome proliferator-activated receptor (PPAR) A/G signaling and an increase in fatty acid import by the transporter FATP1 in BaP-exposed BEAS-2B cells. Furthermore, inhibition of AHR signaling by CH-223191 abolished BaP-induced repression of PPARA/G signaling and activation of FATP1 in BEAS-2B cells, demonstrating the regulatory role of AHR signaling in fatty acid accumulation via mediating PPARA/G-FATP1 signaling. These data suggested amino acid and fatty acid metabolism, AHR and PPAR-FATP1 signaling as potential therapeutic targets for intervening BaP-induced toxicity and related diseases. As far as we known, fatty acid accumulation and high correlations of AHR signaling with amino acid and fatty acid metabolism are novel phenomena discovered in BaP-exposed lung epithelial cells.
Collapse
Affiliation(s)
- Guozhu Ye
- Center for Excellence in Regional Atmospheric Environment, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China; Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China.
| | - Han Gao
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Xu Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Jinsheng Chen
- Center for Excellence in Regional Atmospheric Environment, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China; Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Xu Liao
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Han Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Qiansheng Huang
- Center for Excellence in Regional Atmospheric Environment, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China; Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China.
| |
Collapse
|
18
|
Coady KK, Burgoon L, Doskey C, Davis JW. Assessment of Transcriptomic and Apical Responses of Daphnia magna Exposed to a Polyethylene Microplastic in a 21-d Chronic Study. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2020; 39:1578-1589. [PMID: 32388890 DOI: 10.1002/etc.4745] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/10/2020] [Accepted: 05/04/2020] [Indexed: 06/11/2023]
Abstract
There is global concern regarding the fate and effects of microplastics in the environment, particularly in aquatic systems. In the present study, ethylene acrylic acid copolymer particles were evaluated in a chronic toxicity study with the aquatic invertebrate Daphnia magna. The study design included a natural particle control treatment (silica) to differentiate any potential physical effects of a particle from the intrinsic toxicity of the test material. In addition to the standard endpoints of survival, growth, and reproduction, the transcriptomic profiles of control and ethylene acrylic acid copolymer-exposed D. magna were evaluated at the termination of the 21-d toxicity study. No significant effects on D. magna growth, survival, or reproduction were observed in comparison with both particle and untreated control groups. Significant transcriptomic alterations were induced at the highest treatment level of 2.3 × 1012 particles of the ethylene acrylic acid copolymer/L in key pathways linked to central metabolism and energy reserves, oxidative stress, and ovulation and molting, indicating a global transcriptomic response pattern. To put the results in perspective is challenging at this time, because, to date, microplastic environmental monitoring approaches have not been equipped to detect particles in the nanosize range. However, our results indicate that ethylene acrylic acid copolymer microplastics in the upper nanosize range are not expected to adversely affect D. magna growth, survival, or reproductive outcomes at concentrations of up to 1012 particles/L. Environ Toxicol Chem 2020;39:1578-1589. © 2020 SETAC.
Collapse
Affiliation(s)
- Katherine K Coady
- Toxicology and Environmental Research and Consulting, Dow, Midland, Michigan, USA
| | - Lyle Burgoon
- US Army Engineer Research and Development Center, Raleigh-Durham, North Carolina, USA
| | - Claire Doskey
- Toxicology and Environmental Research and Consulting, Dow, Midland, Michigan, USA
| | - John W Davis
- Toxicology and Environmental Research and Consulting, Dow, Midland, Michigan, USA
| |
Collapse
|
19
|
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
|
20
|
Huerta-Yepez S, Tirado-Rodriguez A, Montecillo-Aguado MR, Yang J, Hammock BD, Hankinson O. Aryl Hydrocarbon Receptor-Dependent inductions of omega-3 and omega-6 polyunsaturated fatty acid metabolism act inversely on tumor progression. Sci Rep 2020; 10:7843. [PMID: 32398692 PMCID: PMC7217871 DOI: 10.1038/s41598-020-64146-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/13/2020] [Indexed: 12/21/2022] Open
Abstract
The Western diet contains a high ratio of omega-6 (ω6) to omega-3 (ω3) polyunsaturated fatty acids (PUFA). The prototypical aryl hydrocarbon receptor (AHR) ligand, 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), induces CYP1 family enzymes, which can metabolize PUFA to epoxides. Mice fed ω3-rich or ω6-rich diets were treated with TCDD and injected subcutaneously with AHR-competent Hepa1-GFP hepatoma cells or AHR-deficient LLC lung cancer cells. TCDD reduced the growth rates of the resulting tumors in ω3-fed mice and inhibited their metastasis to the liver and/or lung, but had the opposite effects in mice fed ω6 PUFA. These responses were likely attributable to the corresponding PUFA epoxides generated in tumor cells and/or host, since many depended upon co-administration of a soluble epoxide hydrolase (EPHX2) inhibitor in males, and/or were associated with increases in epoxide levels in tumors and sites of metastasis. Equivalent effects occurred in females in the absence of EPHX2 inhibition, probably because this sex expressed reduced levels of EPHX2. The responses elicited by TCDD were associated with effects on tumor vascularity, tumor cell proliferation and/or apoptosis. Thus environmental AHR agonists, and potentially also endogenous, nutritional, and microbiome-derived agonists, may reduce or enhance cancer progression depending on the composition of dietary PUFA, particularly in females.
Collapse
Affiliation(s)
- Sara Huerta-Yepez
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Ana Tirado-Rodriguez
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Mayra R Montecillo-Aguado
- Research Unit of Oncology Diseases. Hospital Infantil de Mexico, Federico Gomez, Mexico City, Mexico
| | - Jun Yang
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| | - Oliver Hankinson
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA.
- Molecular Toxicology Interdepartmental Program and Department of Environmental Health Sciences, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
21
|
Josyula N, Andersen ME, Kaminski NE, Dere E, Zacharewski TR, Bhattacharya S. Gene co-regulation and co-expression in the aryl hydrocarbon receptor-mediated transcriptional regulatory network in the mouse liver. Arch Toxicol 2019; 94:113-126. [PMID: 31728591 DOI: 10.1007/s00204-019-02620-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/06/2019] [Indexed: 01/16/2023]
Abstract
Four decades after its discovery, the aryl hydrocarbon receptor (AHR), a ligand-inducible transcription factor (TF) activated by the persistent environmental contaminant 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), remains an enigmatic molecule with a controversial endogenous role. Here, we have assembled a global map of the AHR gene regulatory network in female C57BL/6 mice orally gavaged with 30 µg/kg of TCDD from a combination of previously published gene expression and genome-wide TF-binding data sets. Using Kohonen self-organizing maps and subspace clustering, we show that genes co-regulated by common upstream TFs in the AHR network exhibit a pattern of co-expression. Directly bound, indirectly bound, and non-genomic AHR target genes exhibit distinct expression patterns, with the directly bound targets associated with highest median expression. Interestingly, among the directly bound AHR target genes, the expression level increases with the number of AHR-binding sites in the proximal promoter regions. Finally, we show that co-regulated genes in the AHR network activate distinct groups of downstream biological processes. Although the specific findings described here are restricted to hepatic effects under short-term TCDD exposure, this work describes a generalizable approach to the reconstruction and analysis of transcriptional regulatory cascades underlying cellular stress response, revealing network hierarchy and the nature of information flow from the initial signaling events to phenotypic outcomes. Such reconstructed networks can form the basis of a new generation of quantitative adverse outcome pathways.
Collapse
Affiliation(s)
- Navya Josyula
- Biomedical and Translational Informatics Program, Geisinger Health System, Rockville, MD, 20850, USA
| | | | - Norbert E Kaminski
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.,Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, 48824, USA
| | - Edward Dere
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.,Genentech, South San Francisco, CA, 94080, USA
| | - Timothy R Zacharewski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.,Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Sudin Bhattacharya
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA. .,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA. .,Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824-1226, USA. .,Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, 48824, USA. .,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
22
|
Nynca A, Sadowska A, Paukszto L, Molcan T, Ruszkowska M, Swigonska S, Orlowska K, Myszczynski K, Jastrzebski JP, Ciereszko RE. Temporal changes in the transcriptomic profile of granulosa cells of pigs treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Anim Reprod Sci 2019; 207:83-94. [PMID: 31213330 DOI: 10.1016/j.anireprosci.2019.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/15/2019] [Accepted: 06/04/2019] [Indexed: 01/17/2023]
Abstract
The 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) compound is an environmental chemical adversely affecting reproductive processes. Intracellular TCDD effects are mediated via aryl hydrocarbon receptor (AhR). The aim of the current study was to identify genes linking the AhR pathway with phenotypic consequences of TCDD action in granulosa cells of pigs. By applying multifactorial analysis, with TCDD and incubation time as factors, it was possible to determine temporal changes induced by TCDD in the cell transcriptome. Among the identified 144 differentially expressed genes (DEGs; Padjusted<0.05, log2 fold change (FC)≥1), 111 DEGs were classified as sustained genes (FC values changing between 3 and 24 h). Eighty six DEGs were classified as early genes and only nine as late genes (FC changes observed between 3 and 12 h or 12 and 24 h, respectively). The sustained gene category included genes related to TCDD mechanism of action (AHR, ARNTL, CYP1A1), cell proliferation (TGFβ3), follicular development and ovulation (PTGS2) as well as stress response (NR3C1). The early gene category contained DEGs associated with cell proliferation (DUSP4, TAB1) and cellular response to stress (DHX34). The CYP1A1 gene was the only DEG classified as an early, late and sustained gene. The multifactorial approach allowed for statistically analyzing TCDD-induced changes over time in the gene expression in granulosa cells of pigs. Changes over time in the granulosal transcriptome profile indicated the involvement of stress related molecules in the cellular response to TCDD and TCDD effects on ovulation. The TCDD effects were particularly evident during the early stage of action by this compound.
Collapse
Affiliation(s)
- Anna Nynca
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Prawochenskiego 5, 10-720, Olsztyn, Poland.
| | - Agnieszka Sadowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Lukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Tomasz Molcan
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Monika Ruszkowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Sylwia Swigonska
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Prawochenskiego 5, 10-720, Olsztyn, Poland.
| | - Karina Orlowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Kamil Myszczynski
- Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Plac Lodzki 1, 10-727, Olsztyn, Poland.
| | - Jan P Jastrzebski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Renata E Ciereszko
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Prawochenskiego 5, 10-720, Olsztyn, Poland; Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| |
Collapse
|
23
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin abolishes circadian regulation of hepatic metabolic activity in mice. Sci Rep 2019; 9:6514. [PMID: 31015483 PMCID: PMC6478849 DOI: 10.1038/s41598-019-42760-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 04/05/2019] [Indexed: 12/19/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) activation is reported to alter the hepatic expression of circadian clock regulators, however the impact on clock-controlled metabolism has not been thoroughly investigated. This study examines the effects of AhR activation on hepatic transcriptome and metabolome rhythmicity in male C57BL/6 mice orally gavaged with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) every 4 days for 28 days. TCDD diminished the rhythmicity of several core clock regulators (e.g. Arntl, Clock, Nr1d1, Per1, Cry1, Nfil3) in a dose-dependent manner, involving either a ≥ 3.3-fold suppression in amplitude or complete loss of oscillation. Accordingly, protein levels (ARNTL, REV-ERBα, NFIL3) and genomic binding (ARNTL) of select regulators were reduced and arrhythmic following treatment. As a result, the oscillating expression of 99.6% of 5,636 clock-controlled hepatic genes was abolished including genes associated with the metabolism of lipids, glucose/glycogen, and heme. For example, TCDD flattened expression of the rate-limiting enzymes in both gluconeogenesis (Pck1) and glycogenesis (Gys2), consistent with the depletion and loss of rhythmicity in hepatic glycogen levels. Examination of polar hepatic extracts by untargeted mass spectrometry revealed that virtually all oscillating metabolites lost rhythmicity following treatment. Collectively, these results suggest TCDD disrupted circadian regulation of hepatic metabolism, altering metabolic efficiency and energy storage.
Collapse
|
24
|
Formononetin and biochanin A protects against ritonavir induced hepatotoxicity via modulation of NfκB/pAkt signaling molecules. Life Sci 2018; 213:174-182. [DOI: 10.1016/j.lfs.2018.10.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/07/2018] [Accepted: 10/12/2018] [Indexed: 01/13/2023]
|
25
|
Nault R, Doskey CM, Fader KA, Rockwell CE, Zacharewski T. Comparison of Hepatic NRF2 and Aryl Hydrocarbon Receptor Binding in 2,3,7,8-Tetrachlorodibenzo- p-dioxin-Treated Mice Demonstrates NRF2-Independent PKM2 Induction. Mol Pharmacol 2018; 94:876-884. [PMID: 29752288 PMCID: PMC6022803 DOI: 10.1124/mol.118.112144] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/04/2018] [Indexed: 12/23/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces hepatic oxidative stress following activation of the aryl hydrocarbon receptor (AhR). Our recent studies showed TCDD induced pyruvate kinase muscle isoform 2 (Pkm2) as a novel antioxidant response in normal differentiated hepatocytes. To investigate cooperative regulation between nuclear factor, erythroid derived 2, like 2 (Nrf2) and the AhR in the induction of Pkm2, hepatic chromatin immunoprecipitation sequencing (ChIP-seq) analyses were integrated with RNA sequencing (RNA-seq) time-course data from mice treated with TCDD for 2-168 hours. ChIP-seq analysis 2 hours after TCDD treatment identified genome-wide NRF2 enrichment. Approximately 842 NRF2-enriched regions were located in the regulatory region of differentially expressed genes (DEGs), whereas 579 DEGs showed both NRF2 and AhR enrichment. Sequence analysis of regions with overlapping NRF2 and AhR enrichment showed over-representation of either antioxidant or dioxin response elements, although 18 possessed both motifs. NRF2 exhibited negligible enrichment within a closed Pkm chromatin region, whereas the AhR was enriched 29-fold. Furthermore, TCDD induced Pkm2 in primary hepatocytes from wild-type and Nrf2-null mice, indicating NRF2 is not required. Although NRF2 and AhR cooperate to regulate numerous antioxidant gene expression responses, the induction of Pkm2 by TCDD is independent of reactive oxygen species-mediated NRF2 activation.
Collapse
Affiliation(s)
- Rance Nault
- Departments of Biochemistry and Molecular Biology (R.N., C.M.D., K.A.F., T.Z.) and Pharmacology and Toxicology (C.E.R.) and Institute for Integrative Toxicology (R.N., C.M.D., K.A.F., C.E.R., T.Z.), Michigan State University, East Lansing, Michigan
| | - Claire M Doskey
- Departments of Biochemistry and Molecular Biology (R.N., C.M.D., K.A.F., T.Z.) and Pharmacology and Toxicology (C.E.R.) and Institute for Integrative Toxicology (R.N., C.M.D., K.A.F., C.E.R., T.Z.), Michigan State University, East Lansing, Michigan
| | - Kelly A Fader
- Departments of Biochemistry and Molecular Biology (R.N., C.M.D., K.A.F., T.Z.) and Pharmacology and Toxicology (C.E.R.) and Institute for Integrative Toxicology (R.N., C.M.D., K.A.F., C.E.R., T.Z.), Michigan State University, East Lansing, Michigan
| | - Cheryl E Rockwell
- Departments of Biochemistry and Molecular Biology (R.N., C.M.D., K.A.F., T.Z.) and Pharmacology and Toxicology (C.E.R.) and Institute for Integrative Toxicology (R.N., C.M.D., K.A.F., C.E.R., T.Z.), Michigan State University, East Lansing, Michigan
| | - Tim Zacharewski
- Departments of Biochemistry and Molecular Biology (R.N., C.M.D., K.A.F., T.Z.) and Pharmacology and Toxicology (C.E.R.) and Institute for Integrative Toxicology (R.N., C.M.D., K.A.F., C.E.R., T.Z.), Michigan State University, East Lansing, Michigan
| |
Collapse
|
26
|
Lamas B, Natividad JM, Sokol H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol 2018; 11:1024-1038. [PMID: 29626198 DOI: 10.1038/s41385-018-0019-2] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 02/04/2023]
Abstract
Aryl hydrocarbon receptor (AhR) is a member of the basic helix-loop-helix-(bHLH) superfamily of transcription factors, which are associated with cellular responses to environmental stimuli, such as xenobiotics and oxygen levels. Unlike other members of bHLH, AhR is the only bHLH transcription factor that is known to be ligand activated. Early AhR studies focused on understanding the role of AhR in mediating the toxicity and carcinogenesis properties of the prototypic ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In recent years, however, it has become apparent that, in addition to its toxicological involvement, AhR is highly receptive to a wide array of endogenous and exogenous ligands, and that its activation leads to a myriad of key host physiological functions. In this study, we review the current understanding of the functions of AhR in the mucosal immune system with a focus on its role in intestinal barrier function and intestinal immune cells, as well as in intestinal homeostasis.
Collapse
Affiliation(s)
- Bruno Lamas
- Laboratoire de biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Hôpital Saint-Antoine, Paris, F-75005, France.,Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy en Josas, 78350, France
| | - Jane M Natividad
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy en Josas, 78350, France
| | - Harry Sokol
- Laboratoire de biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Hôpital Saint-Antoine, Paris, F-75005, France. .,Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy en Josas, 78350, France.
| |
Collapse
|
27
|
Kim KH, Choi JM, Li F, Arizpe A, Wooton-Kee CR, Anakk S, Jung SY, Finegold MJ, Moore DD. Xenobiotic Nuclear Receptor Signaling Determines Molecular Pathogenesis of Progressive Familial Intrahepatic Cholestasis. Endocrinology 2018; 159:2435-2446. [PMID: 29718219 PMCID: PMC7263843 DOI: 10.1210/en.2018-00110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/20/2018] [Indexed: 01/14/2023]
Abstract
Progressive familial intrahepatic cholestasis (PFIC) is a genetically heterogeneous disorder of bile flow disruption due to abnormal canalicular transport or impaired bile acid (BA) metabolism, causing excess BA accumulation and liver failure. We previously reported an intrahepatic cholestasis mouse model based on loss of function of both farnesoid X receptor (FXR; NR1H4) and a small heterodimer partner (SHP; NR0B2) [double knockout (DKO)], which has strong similarities to human PFIC5. We compared the pathogenesis of DKO livers with that of another intrahepatic cholestasis model, Bsep-/-, which represents human PFIC2. Both models exhibit severe hepatomegaly and hepatic BA accumulation, but DKO showed greater circulating BA and liver injury, and Bsep-/- had milder phenotypes. Molecular profiling of BAs uncovered specific enrichment of cholic acid (CA)-derived BAs in DKO livers but chenodeoxycholate-derived BAs in Bsep-/- livers. Transcriptomic and proteomic analysis revealed specific activation of CA synthesis and alternative basolateral BA transport in DKO but increased chenodeoxycholic acid synthesis and canalicular transport in Bsep-/-. The constitutive androstane receptor (CAR)/pregnane X receptor (PXR)-CYP2B/CYP2C axis is activated in DKO livers but not in other cholestasis models. Loss of this axis in Fxr:Shp:Car:Pxr quadruple knockouts blocked Cyp2b/Cyp2c gene induction, impaired bilirubin conjugation/elimination, and increased liver injury. Differential CYP2B expression in DKO and Bsep-/- was recapitulated in human PFIC5 and PFIC2 livers. In conclusion, loss of FXR/SHP results in distinct molecular pathogenesis and CAR/PXR activation, which promotes Cyp2b/Cyp2c gene transcription and bilirubin clearance. CAR/PXR activation was not observed in Bsep-/- mice or PFIC2 patients. These findings provide a deeper understanding of the heterogeneity of intrahepatic cholestasis.
Collapse
Affiliation(s)
- Kang Ho Kim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jong Min Choi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Feng Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas
| | - Armando Arizpe
- School of Natural Science, University of Texas at Austin, Austin, Texas
| | - Clavia Ruth Wooton-Kee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Milton J Finegold
- Department of Pathology and Immunology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Correspondence: David D. Moore, PhD, Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030. E-mail:
| |
Collapse
|
28
|
Fader KA, Nault R, Raehtz S, McCabe LR, Zacharewski TR. 2,3,7,8-Tetrachlorodibenzo-p-dioxin dose-dependently increases bone mass and decreases marrow adiposity in juvenile mice. Toxicol Appl Pharmacol 2018; 348:85-98. [PMID: 29673856 PMCID: PMC5984050 DOI: 10.1016/j.taap.2018.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/03/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and other aryl hydrocarbon receptor (AhR) agonists have been shown to regulate bone development and remodeling in a species-, ligand-, and age-specific manner, however the underlying mechanisms remain poorly understood. In this study, we characterized the effect of 0.01-30 μg/kg TCDD on the femoral morphology of male and female juvenile mice orally gavaged every 4 days for 28 days and used RNA-Seq to investigate gene expression changes associated with the resultant phenotype. Micro-computed tomography revealed that TCDD dose-dependently increased trabecular bone volume fraction (BVF) 2.9- and 3.3-fold in male and female femurs, respectively. Decreased serum tartrate-resistant acid phosphatase (TRAP) levels, combined with a reduced osteoclast surface to bone surface ratio and repression of femoral proteases (cathepsin K, matrix metallopeptidase 13), suggests that TCDD impaired bone resorption. Increased osteoblast counts at the trabecular bone surface were consistent with a reciprocal reduction in the number of bone marrow adipocytes, suggesting AhR activation may direct mesenchymal stem cell differentiation towards osteoblasts rather than adipocytes. Notably, femoral expression of transmembrane glycoprotein NMB (Gpnmb; osteoactivin), a positive regulator of osteoblast differentiation and mineralization, was dose-dependently induced up to 18.8-fold by TCDD. Moreover, increased serum levels of 1,25-dihydroxyvitamin D3 were in accordance with the renal induction of 1α-hydroxylase Cyp27b1 and may contribute to impaired bone resorption. Collectively, the data suggest AhR activation tipped the bone remodeling balance towards bone formation, resulting in increased bone mass with reduced marrow adiposity.
Collapse
Affiliation(s)
- Kelly A Fader
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Rance Nault
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States
| | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States; Department of Radiology, Michigan State University, East Lansing, MI 48824, United States
| | - Timothy R Zacharewski
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
29
|
Erdemli ME, Yigitcan B, Gul M, Bag HG, Gul S, Aksungur Z. Thymoquinone is protective against 2,3,7,8-tetrachlorodibenzo-p-dioxin induced hepatotoxicity. Biotech Histochem 2018; 93:453-462. [PMID: 29701106 DOI: 10.1080/10520295.2018.1453549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
We investigated changes in rat liver tissues following administration of thymoquinone (TQ) against 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced hepatotoxicity. Fifty rats were assigned randomly to five groups of 10 as follows: control, corn oil, TCDD, TQ and TCDD + TQ. Biochemical and histopathological analyses were conducted on liver tissue. We found that 30 day TCDD administration caused histopathological changes in liver including thickening of Glisson's capsule, intracytoplasmic vacuolization in hepatocytes, sinusoidal dilation, vascular and sinusoidal congestion and inflammatory cell infiltration. TCDD administration increased malondialdehyde (MDA), total oxidant status (TOS), alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) levels in rat liver tissue and reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT) and total antioxidant status (TAS) levels compared to all other groups. In the TQ treated group, GSH, SOD, CAT and TAS levels increased compared to all other groups. MDA, TOS, ALT, AST, ALP levels decreased compared to all other groups. Our histological findings were consistent with the biochemical findings. The oxidative and histologic effects of TCDD were eliminated by TQ treatment. TCDD administration caused oxidative stress in rat liver and TQ administered with TCDD prevented TCDD induced hepatotoxicity. TQ could be considered an alternative anti-TCDD toxicity agent.
Collapse
Affiliation(s)
- M E Erdemli
- a Department of Medical Biochemistry, Medical Faculty , Nigde Omer Halisdemir University , Nigde
| | - B Yigitcan
- b Departments of Histology and Embryology, Medical Faculty , Inonu University , Malatya
| | - M Gul
- b Departments of Histology and Embryology, Medical Faculty , Inonu University , Malatya
| | - H G Bag
- c Department of Biostatistics, Medical Faculty , Inonu University , Malatya
| | - S Gul
- b Departments of Histology and Embryology, Medical Faculty , Inonu University , Malatya
| | - Z Aksungur
- d Department of Medical Biochemistry, Medical Faculty , Inonu University , Malatya , Turkey
| |
Collapse
|
30
|
Csanaky IL, Lickteig AJ, Klaassen CD. Aryl hydrocarbon receptor (AhR) mediated short-term effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on bile acid homeostasis in mice. Toxicol Appl Pharmacol 2018; 343:48-61. [PMID: 29452137 DOI: 10.1016/j.taap.2018.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/25/2018] [Accepted: 02/12/2018] [Indexed: 01/05/2023]
Abstract
The effects of the most potent aryl hydrocarbon receptor (AhR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on bile acid (BA) homeostasis was examined in male and female wild-type and AhR-null mice shortly after 4-day exposure, rather than at a later time when secondary non-AhR dependent effects are more likely to occur. TCDD had similar effects on BA homeostasis in male and female mice. TCDD decreased the concentration of total-(Σ) BAs in liver by approximately 50% (all major BA categories except for the non-6,12-OH BAs), without decreasing the expression of the rate limiting BA synthetic enzyme (Cyp7a1) or altering the major BA regulatory pathways (FXR) in liver and intestine. Even though the Σ-BAs in liver were markedly decreased, the Σ-BAs excreted into bile were not altered. TCDD decreased the relative amount of 12-OH BAs (TCA, TDCA, CA, DCA) in bile and increased the biliary excretion of TCDCA and its metabolites (TαMCA, TUDCA); this was likely due to the decreased Cyp8b1 (12α-hydroxylase) in liver. The concentration of Σ-BAs in serum was not altered by TCDD, indicating that serum BAs do not reflect BA status in liver. However, proportions of individual BAs in serum reflected the decreased expression of Cyp8b1. All these TCDD-induced changes in BA homeostasis were absent in AhR-null mice. In summary, through the AhR, TCDD markedly decreases BA concentrations in liver and reduces the 12α-hydroxylation of BAs without altering Cyp7a1 and FXR signaling. The TCDD-induced decrease in Σ-BAs in liver did not result in a decrease in biliary excretion or serum concentrations of Σ-BAs.
Collapse
Affiliation(s)
- Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital, Kansas City, MO 64108; USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
31
|
The aryl hydrocarbon receptor regulates the expression of TIPARP and its cis long non-coding RNA, TIPARP-AS1. Biochem Biophys Res Commun 2018; 495:2356-2362. [DOI: 10.1016/j.bbrc.2017.12.113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
|
32
|
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
|
33
|
Loss of liver-specific and sexually dimorphic gene expression by aryl hydrocarbon receptor activation in C57BL/6 mice. PLoS One 2017; 12:e0184842. [PMID: 28922406 PMCID: PMC5602546 DOI: 10.1371/journal.pone.0184842] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a highly conserved transcription factor that mediates a broad spectrum of species-, strain-, sex-, age-, tissue-, and cell-specific responses elicited by structurally diverse ligands including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Dose-dependent effects on liver-specific and sexually dimorphic gene expression were examined in male and female mice gavaged with TCDD every 4 days for 28 or 92 days. RNA-seq data revealed the coordinated repression of 181 genes predominately expressed in the liver including albumin (3.7-fold), α-fibrinogen (14.5-fold), and β-fibrinogen (17.4-fold) in males with corresponding AhR enrichment at 2 hr. Liver-specific genes exhibiting sexually dimorphic expression also demonstrated diminished divergence between sexes. For example, male-biased Gstp1 was repressed 3.0-fold in males and induced 4.5-fold in females, which were confirmed at the protein level. Disrupted regulation is consistent with impaired GHR-JAK2-STAT5 signaling and inhibition of female specific CUX2-mediated transcription as well as the repression of other key transcriptional regulators including Ghr, Stat5b, Bcl6, Hnf4a, Hnf6, Foxa1/2/3, and Zhx2. Attenuated liver-specific and sexually dimorphic gene expression was concurrent with the induction of fetal genes such as alpha-fetoprotein. The results suggest AhR activation causes the loss of liver-specific and sexually dimorphic gene expression producing a functionally "de-differentiated" hepatic phenotype.
Collapse
|
34
|
Alassane-Kpembi I, Gerez JR, Cossalter AM, Neves M, Laffitte J, Naylies C, Lippi Y, Kolf-Clauw M, Bracarense APL, Pinton P, Oswald IP. Intestinal toxicity of the type B trichothecene mycotoxin fusarenon-X: whole transcriptome profiling reveals new signaling pathways. Sci Rep 2017; 7:7530. [PMID: 28790326 PMCID: PMC5548841 DOI: 10.1038/s41598-017-07155-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/23/2017] [Indexed: 12/02/2022] Open
Abstract
The few data available on fusarenon-X (FX) do not support the derivation of health-based guidance values, although preliminary results suggest higher toxicity than other regulated trichothecenes. Using histo-morphological analysis and whole transcriptome profiling, this study was designed to obtain a global view of the intestinal alterations induced by FX. Deoxynivalenol (DON) served as a benchmark. FX induced more severe histological alterations than DON. Inflammation was the hallmark of the molecular toxicity of both mycotoxins. The benchmark doses for the up-regulation of key inflammatory genes by FX were 4- to 45-fold higher than the previously reported values for DON. The transcriptome analysis revealed that both mycotoxins down-regulated the peroxisome proliferator-activated receptor (PPAR) and liver X receptor - retinoid X receptor (LXR-RXR) signaling pathways that control lipid metabolism. Interestingly, several pathways, including VDR/RXR activation, ephrin receptor signaling, and GNRH signaling, were specific to FX and thus discriminated the transcriptomic fingerprints of the two mycotoxins. These results demonstrate that FX induces more potent intestinal inflammation than DON. Moreover, although the mechanisms of toxicity of both mycotoxins are similar in many ways, this study emphasize specific pathways targeted by each mycotoxin, highlighting the need for specific mechanism-based risk assessments of Fusarium mycotoxins.
Collapse
Affiliation(s)
- Imourana Alassane-Kpembi
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
- Hôpital d'Instruction des Armées, Camp Guézo, 01BP517, Cotonou, Benin
| | - Juliana Rubira Gerez
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Anne-Marie Cossalter
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
| | - Manon Neves
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
| | - Joëlle Laffitte
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
| | - Claire Naylies
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
| | - Yannick Lippi
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
| | - Martine Kolf-Clauw
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
- Université de Toulouse, Ecole Nationale Vétérinaire (ENVT), Toulouse, France
| | - Ana Paula L Bracarense
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Philippe Pinton
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France
| | - Isabelle P Oswald
- Toxalim, Research Center in Food Toxicology, Université de Toulouse, INRA, ENVT, INP- PURPAN, UPS, F-31027, Toulouse, France.
| |
Collapse
|
35
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)-elicited effects on bile acid homeostasis: Alterations in biosynthesis, enterohepatic circulation, and microbial metabolism. Sci Rep 2017; 7:5921. [PMID: 28725001 PMCID: PMC5517430 DOI: 10.1038/s41598-017-05656-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/01/2017] [Indexed: 01/14/2023] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant which elicits hepatotoxicity through activation of the aryl hydrocarbon receptor (AhR). Male C57BL/6 mice orally gavaged with TCDD (0.01–30 µg/kg) every 4 days for 28 days exhibited bile duct proliferation and pericholangitis. Mass spectrometry analysis detected a 4.6-fold increase in total hepatic bile acid levels, despite the coordinated repression of genes involved in cholesterol and primary bile acid biosynthesis including Cyp7a1. Specifically, TCDD elicited a >200-fold increase in taurolithocholic acid (TLCA), a potent G protein-coupled bile acid receptor 1 (GPBAR1) agonist associated with bile duct proliferation. Increased levels of microbial bile acid metabolism loci (bsh, baiCD) are consistent with accumulation of TLCA and other secondary bile acids. Fecal bile acids decreased 2.8-fold, suggesting enhanced intestinal reabsorption due to induction of ileal transporters (Slc10a2, Slc51a) and increases in whole gut transit time and intestinal permeability. Moreover, serum bile acids were increased 45.4-fold, consistent with blood-to-hepatocyte transporter repression (Slco1a1, Slc10a1, Slco2b1, Slco1b2, Slco1a4) and hepatocyte-to-blood transporter induction (Abcc4, Abcc3). These results suggest that systemic alterations in enterohepatic circulation, as well as host and microbiota bile acid metabolism, favor bile acid accumulation that contributes to AhR-mediated hepatotoxicity.
Collapse
|
36
|
Yu Y, Liu Q, Guo S, Zhang Q, Tang J, Liu G, Kong D, Li J, Yan S, Wang R, Wang P, Su X, Yu Y. 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin promotes endothelial cell apoptosis through activation of EP3/p38MAPK/Bcl-2 pathway. J Cell Mol Med 2017; 21:3540-3551. [PMID: 28699682 PMCID: PMC5706494 DOI: 10.1111/jcmm.13265] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 05/02/2017] [Indexed: 12/18/2022] Open
Abstract
Endothelial injury or dysfunction is an early event in the pathogenesis of atherosclerosis. Epidemiological and animal studies have shown that 2, 3, 7, 8‐tetrachlorodibenzo‐p‐dioxin (TCDD) exposure increases morbidity and mortality from chronic cardiovascular diseases, including atherosclerosis. However, whether or how TCDD exposure causes endothelial injury or dysfunction remains largely unknown. Cultured human umbilical vein endothelial cells (HUVECs) were exposed to different doses of TCDD, and cell apoptosis was examined. We found that TCDD treatment increased caspase 3 activity and apoptosis in HUVECs in a dose‐dependent manner,at doses from 10 to 40 nM. TCDD increased cyclooxygenase enzymes (COX)‐2 expression and its downstream prostaglandin (PG) production (mainly PGE2 and 6‐keto‐PGF1α) in HUVECs. Interestingly, inhibition of COX‐2, but not COX‐1, markedly attenuated TCDD‐triggered apoptosis in HUVECs. Pharmacological inhibition or gene silencing of the PGE2 receptor subtype 3 (EP3) suppressed the augmented apoptosis in TCDD‐treated HUVECs. Activation of the EP3 receptor enhanced p38 MAPK phosphorylation and decreased Bcl‐2 expression following TCDD treatment. Both p38 MAPK suppression and Bcl‐2 overexpression attenuated the apoptosis in TCDD‐treated HUVECs. TCDD increased EP3‐dependent Rho activity and subsequently promoted p38MAPK/Bcl‐2 pathway‐mediated apoptosis in HUVECs. In addition, TCDD promoted apoptosis in vascular endothelium and delayed re‐endothelialization after femoral artery injury in wild‐type (WT) mice, but not in EP3−/− mice. In summary, TCDD promotes endothelial apoptosis through the COX‐2/PGE2/EP3/p38MAPK/Bcl‐2 pathway. Given the cardiovascular hazard of a COX‐2 inhibitor, our findings indicate that the EP3 receptor and its downstream pathways may be potential targets for prevention of TCDD‐associated cardiovascular diseases.
Collapse
Affiliation(s)
- Yu Yu
- Department of Pharmacology, Tianjin Medical University, Tianjin, China.,Department of Pediatric Cardiology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Liu
- Department of Pharmacology, Tianjin Medical University, Tianjin, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shumin Guo
- Department of Pharmacology, Tianjin Medical University, Tianjin, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qianqian Zhang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juan Tang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guizhu Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Deping Kong
- Department of Pharmacology, Tianjin Medical University, Tianjin, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juanjuan Li
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuai Yan
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruiguo Wang
- Institute of Quality Standards and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Peilong Wang
- Institute of Quality Standards and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoou Su
- Institute of Quality Standards and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Medical University, Tianjin, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
37
|
Ostrowska A, Gostomska-Pampuch K, Lesków A, Kuropka P, Gamian E, Ziólkowski P, Kowalczyk A, Lukaszewicz E, Gamian A, Calkosinski I. Expression of advanced glycation end-products and NFκB in chick embryos exposed to dioxins and treated with acetylsalicylic acid and α-tocopherol. Poult Sci 2017; 96:1874-1883. [PMID: 28158694 DOI: 10.3382/ps/pew450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/16/2016] [Indexed: 01/04/2023] Open
Abstract
Dioxins have adverse and multifaceted effect on body functions. They are known to be carcinogens, immunotoxins, and teratogenic agents. In vivo, transformation of dioxins occurs after their interaction with the aryl hydrocarbon receptor (AhR) and leads to formation of proinflammatory and toxic metabolites. The aim of this study was to verify whether α-tocopherol (vitamin E) and acetylsalicylic acid (ASA), could reduce the damage caused by the action of dioxins. Fertile chicken eggs were injected with a solution of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), followed by the injection of α-tocopherol or acetylsalicylic acid. Organs such as heart and liver were dissected from the chick embryos at d 13 and 19 of development and subjected to immunohistochemical analysis of presence of advanced glycation end products (AGEs) and nuclear factor kappa B (NFκB) in tissues. The AGEs were used as the marker for exposure to dioxins, since it is well established that their level increases in dioxin-damaged tissues. Formation of AGEs was evaluated in embryos exposed to dioxin and treated with vitamin E and/or ASA (against dioxin-exposed, untreated controls). We have found that TCDD causes developmental disorders and increases the level of AGEs in chick embryo tissues. The use of such pharmacological agents as vitamin E, ASA, and combination of ASA and vitamin E, inhibited formation of the AGEs in 13-day-old embryos and reduced the AGEs level in embryos after 19 d of the development.
Collapse
Affiliation(s)
- A Ostrowska
- Independent Laboratory of Neurotoxicology and Environmental Diagnostics, Wroclaw Medical University, Wroclaw, Poland
| | - K Gostomska-Pampuch
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - A Lesków
- Independent Laboratory of Neurotoxicology and Environmental Diagnostics, Wroclaw Medical University, Wroclaw, Poland
| | - P Kuropka
- Department of Animal Physiology and Biostructure, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - E Gamian
- Department of Pathomorphology, Wroclaw Medical University, Wroclaw, Poland
| | - P Ziólkowski
- Department of Pathomorphology, Wroclaw Medical University, Wroclaw, Poland
| | - A Kowalczyk
- Institute of Animal Breeding, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - E Lukaszewicz
- Institute of Animal Breeding, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - A Gamian
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland.,Wroclaw Research Centre EIT+, Wroclaw, Poland
| | - I Calkosinski
- Independent Laboratory of Neurotoxicology and Environmental Diagnostics, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
38
|
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
|
39
|
Nault R, Fader KA, Lydic TA, Zacharewski TR. Lipidomic Evaluation of Aryl Hydrocarbon Receptor-Mediated Hepatic Steatosis in Male and Female Mice Elicited by 2,3,7,8-Tetrachlorodibenzo-p-dioxin. Chem Res Toxicol 2017; 30:1060-1075. [PMID: 28238261 PMCID: PMC5896278 DOI: 10.1021/acs.chemrestox.6b00430] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces hepatic steatosis mediated by the aryl hydrocarbon receptor. To further characterize TCDD-elicited hepatic lipid accumulation, mice were gavaged with TCDD every 4 days for 28 days. Liver samples were examined using untargeted lipidomics with structural confirmation of lipid species by targeted high-resolution MS/MS, and data were integrated with complementary RNA-Seq analyses. Approximately 936 unique spectral features were detected, of which 379 were confirmed as unique lipid species. Both male and female samples exhibited similar qualitative changes (lipid species) but differed in quantitative changes. A shift to higher mass lipid species was observed, indicative of increased free fatty acid (FFA) packaging. For example, of the 13 lipid classes examined, triglycerides increased from 46 to 48% of total lipids to 68-83% in TCDD treated animals. Hepatic cholesterol esters increased 11.3-fold in male mice with moieties consisting largely of dietary fatty acids (FAs) (i.e., linolenate, palmitate, and oleate). Phosphatidylserines, phosphatidylethanolamines, phosphatidic acids, and cardiolipins decreased 4.1-, 5.0-, 5.4- and 7.4-fold, respectively, while ceramides increased 6.6-fold. Accordingly, the integration of lipidomic data with differential gene expression associated with lipid metabolism suggests that in addition to the repression of de novo fatty acid synthesis and β-oxidation, TCDD also increased hepatic uptake and packaging of lipids, while inhibiting VLDL secretion, consistent with hepatic fat accumulation and the progression to steatohepatitis with fibrosis.
Collapse
Affiliation(s)
- Rance Nault
- Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kelly A. Fader
- Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Todd A. Lydic
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Timothy R. Zacharewski
- Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
40
|
Duval C, Teixeira-Clerc F, Leblanc AF, Touch S, Emond C, Guerre-Millo M, Lotersztajn S, Barouki R, Aggerbeck M, Coumoul X. Chronic Exposure to Low Doses of Dioxin Promotes Liver Fibrosis Development in the C57BL/6J Diet-Induced Obesity Mouse Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:428-436. [PMID: 27713108 PMCID: PMC5332187 DOI: 10.1289/ehp316] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/11/2016] [Accepted: 08/19/2016] [Indexed: 05/21/2023]
Abstract
BACKGROUND Exposure to persistent organic pollutants (POPs) has been associated with the progression of chronic liver diseases, yet the contribution of POPs to the development of fibrosis in non-alcoholic fatty liver disease (NAFLD), a condition closely linked to obesity, remains poorly documented. OBJECTIVES We investigated the effects of subchronic exposure to low doses of the POP 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an aryl hydrocarbon receptor ligand, on NAFLD progression in diet-induced obese C57BL/6J mice. METHODS Male C57BL/6J mice were fed either a 10% low-fat (LFD) or a 45% high-fat (HFD) purified diet for 14 weeks and TCDD-exposed groups were injected once a week with 5 μg/kg TCDD or the vehicle for the last 6 weeks of the diet. RESULTS Liver histology and triglyceride levels showed that exposure of HFD fed mice to TCDD worsened hepatic steatosis, as compared to either HFD alone or LFD plus TCDD and the mRNA levels of key genes of hepatic lipid metabolism were strongly altered in co-treated mice. Further, increased liver collagen staining and serum transaminase levels showed that TCDD induced liver fibrosis in the HFD fed mice. TCDD in LFD fed mice increased the expression of several inflammation and fibrosis marker genes with no additional effect from a HFD. CONCLUSIONS Exposure to TCDD amplifies the impairment of liver functions observed in mice fed an enriched fat diet as compared to a low fat diet. The results provide new evidence that environmental pollutants promote the development of liver fibrosis in obesity-related NAFLD in C57BL/6J mice. Citation: Duval C, Teixeira-Clerc F, Leblanc AF, Touch S, Emond C, Guerre-Millo M, Lotersztajn S, Barouki R, Aggerbeck M, Coumoul X. 2017. Chronic exposure to low doses of dioxin promotes liver fibrosis development in the C57BL/6J diet-induced obesity mouse model. Environ Health Perspect 125:428-436; http://dx.doi.org/10.1289/EHP316.
Collapse
Affiliation(s)
- Caroline Duval
- INSERM UMR (Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche)-S1124, Paris, France
- Université Paris Descartes, ComUE (Communauté d’universités et d’établissements), Sorbonne Paris Cité, Paris, France
| | - Fatima Teixeira-Clerc
- INSERM UMR-S955, Hôpital Henri Mondor, Créteil, France
- Université Paris-Est, Créteil, France
| | - Alix F. Leblanc
- INSERM UMR (Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche)-S1124, Paris, France
- Université Paris Descartes, ComUE (Communauté d’universités et d’établissements), Sorbonne Paris Cité, Paris, France
| | - Sothea Touch
- INSERM UMR-S1166, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | - Claude Emond
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Quebec, Canada
| | | | - Sophie Lotersztajn
- INSERM UMR-S955, Hôpital Henri Mondor, Créteil, France
- Université Paris-Est, Créteil, France
| | - Robert Barouki
- INSERM UMR (Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche)-S1124, Paris, France
- Université Paris Descartes, ComUE (Communauté d’universités et d’établissements), Sorbonne Paris Cité, Paris, France
- AP-HP (Assistance Publique - Hôpitaux de Paris), Hôpital Necker-Enfants Malades, Paris, France
| | - Martine Aggerbeck
- INSERM UMR (Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche)-S1124, Paris, France
- Université Paris Descartes, ComUE (Communauté d’universités et d’établissements), Sorbonne Paris Cité, Paris, France
| | - Xavier Coumoul
- INSERM UMR (Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche)-S1124, Paris, France
- Université Paris Descartes, ComUE (Communauté d’universités et d’établissements), Sorbonne Paris Cité, Paris, France
- Address correspondence to X. Coumoul, INSERM UMR-S1124, Université Paris Descartes, 45 rue des Saints-Pères, 75006 Paris, France. Telephone: 33142863359. E-mail:
| |
Collapse
|
41
|
Convergence of hepcidin deficiency, systemic iron overloading, heme accumulation, and REV-ERBα/β activation in aryl hydrocarbon receptor-elicited hepatotoxicity. Toxicol Appl Pharmacol 2017; 321:1-17. [PMID: 28213091 DOI: 10.1016/j.taap.2017.02.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/10/2017] [Accepted: 02/07/2017] [Indexed: 12/13/2022]
Abstract
Persistent aryl hydrocarbon receptor (AhR) agonists elicit dose-dependent hepatic lipid accumulation, oxidative stress, inflammation, and fibrosis in mice. Iron (Fe) promotes AhR-mediated oxidative stress by catalyzing reactive oxygen species (ROS) production. To further characterize the role of Fe in AhR-mediated hepatotoxicity, male C57BL/6 mice were orally gavaged with sesame oil vehicle or 0.01-30μg/kg 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) every 4days for 28days. Duodenal epithelial and hepatic RNA-Seq data were integrated with hepatic AhR ChIP-Seq, capillary electrophoresis protein measurements, and clinical chemistry analyses. TCDD dose-dependently repressed hepatic expression of hepcidin (Hamp and Hamp2), the master regulator of systemic Fe homeostasis, resulting in a 2.6-fold increase in serum Fe with accumulating Fe spilling into urine. Total hepatic Fe levels were negligibly increased while transferrin saturation remained unchanged. Furthermore, TCDD elicited dose-dependent gene expression changes in heme biosynthesis including the induction of aminolevulinic acid synthase 1 (Alas1) and repression of uroporphyrinogen decarboxylase (Urod), leading to a 50% increase in hepatic hemin and a 13.2-fold increase in total urinary porphyrins. Consistent with this heme accumulation, differential gene expression suggests that heme activated BACH1 and REV-ERBα/β, causing induction of heme oxygenase 1 (Hmox1) and repression of fatty acid biosynthesis, respectively. Collectively, these results suggest that Hamp repression, Fe accumulation, and increased heme levels converge to promote oxidative stress and the progression of TCDD-elicited hepatotoxicity.
Collapse
|
42
|
Wright EJ, De Castro KP, Joshi AD, Elferink CJ. Canonical and non-canonical aryl hydrocarbon receptor signaling pathways. CURRENT OPINION IN TOXICOLOGY 2017; 2:87-92. [PMID: 32296737 DOI: 10.1016/j.cotox.2017.01.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Decades of research on the Aryl hydrocarbon Receptor (AhR) has unveiled its involvement in the toxicity of halogenated and polycyclic aromatic hydrocarbons, and a myriad of normal physiological processes. The molecular dissection of AhR biology has centered on a canonical signaling pathway in an effort to mechanistically reconcile the diverse pathophysiological effects of exposure to environmental pollutants. As a consequence, we now know that canonical signaling can explain many but not all of the AhR-mediated effects. Here we describe recent findings that point to non-canonical signaling pathways, and focus on a novel AhR interaction with the Krüppel-like Factor 6 protein responsible for previously un-recognized epigenetic changes in the chromatin affecting gene expression.
Collapse
Affiliation(s)
- Eric J Wright
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0654, USA
| | - Karen Pereira De Castro
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0654, USA
| | - Aditya D Joshi
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0654, USA
| | - Cornelis J Elferink
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0654, USA
| |
Collapse
|
43
|
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
|
44
|
Hwang HJ, Dornbos P, Steidemann M, Dunivin TK, Rizzo M, LaPres JJ. Mitochondrial-targeted aryl hydrocarbon receptor and the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin on cellular respiration and the mitochondrial proteome. Toxicol Appl Pharmacol 2016; 304:121-32. [PMID: 27105554 DOI: 10.1016/j.taap.2016.04.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/02/2016] [Accepted: 04/08/2016] [Indexed: 11/18/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor within the Per-Arnt-Sim (PAS) domain superfamily. Exposure to the most potent AHR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is associated with various pathological effects including metabolic syndrome. While research over the last several years has demonstrated a role for oxidative stress and metabolic dysfunction in AHR-dependent TCDD-induced toxicity, the role of the mitochondria in this process has not been fully explored. Our previous research suggested that a portion of the cellular pool of AHR could be found in the mitochondria (mitoAHR). Using a protease protection assay with digitonin extraction, we have now shown that this mitoAHR is localized to the inter-membrane space (IMS) of the organelle. TCDD exposure induced a degradation of mitoAHR similar to that of cytosolic AHR. Furthermore, siRNA-mediated knockdown revealed that translocase of outer-mitochondrial membrane 20 (TOMM20) was involved in the import of AHR into the mitochondria. In addition, TCDD altered cellular respiration in an AHR-dependent manner to maintain respiratory efficiency as measured by oxygen consumption rate (OCR). Stable isotope labeling by amino acids in cell culture (SILAC) identified a battery of proteins within the mitochondrial proteome influenced by TCDD in an AHR-dependent manner. Among these, 17 proteins with fold changes≥2 are associated with various metabolic pathways, suggesting a role of mitochondrial retrograde signaling in TCDD-mediated pathologies. Collectively, these studies suggest that mitoAHR is localized to the IMS and AHR-dependent TCDD-induced toxicity, including metabolic dysfunction, wasting syndrome, and hepatic steatosis, involves mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hye Jin Hwang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Center for Mitochondrial Science and Medicine, Michigan State University, East Lansing, MI 48824, United States
| | - Peter Dornbos
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824-1319, United States
| | - Michelle Steidemann
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824-1319, United States; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Taylor K Dunivin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, United States
| | - Mike Rizzo
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824-1319, United States; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI 48824, United States
| | - John J LaPres
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Center for Mitochondrial Science and Medicine, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
45
|
Fader KA, Nault R, Ammendolia DA, Harkema JR, Williams KJ, Crawford RB, Kaminski NE, Potter D, Sharratt B, Zacharewski TR. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin Alters Lipid Metabolism and Depletes Immune Cell Populations in the Jejunum of C57BL/6 Mice. Toxicol Sci 2015; 148:567-80. [PMID: 26377647 PMCID: PMC5009443 DOI: 10.1093/toxsci/kfv206] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent aryl hydrocarbon receptor agonist that elicits dose-dependent hepatic fat accumulation and inflammation that can progress to steatohepatitis. To investigate intestine-liver interactions that contribute to TCDD-elicited steatohepatitis, we examined the dose-dependent effects of TCDD (0.01, 0.03, 0.1, 0.3, 1, 3, 10, or 30 µg/kg) on jejunal epithelial gene expression in C57BL/6 mice orally gavaged every 4 days for 28 days. Agilent 4x44K whole-genome microarray analysis of the jejunal epithelium identified 439 differentially expressed genes (|fold change| ≥ 1.5, P1(t) ≥ 0.999) across 1 or more doses, many related to lipid metabolism and immune system processes. TCDD-elicited differentially expressed genes were associated with lipolysis, fatty acid/cholesterol absorption and transport, the Kennedy pathway, and retinol metabolism, consistent with increased hepatic fat accumulation. Moreover, several major histocompatibility complex (MHC) class II genes (H2-Aa, H2-Ab1, H2-DMb1, Cd74) were repressed, coincident with decreased macrophage and dendritic cell levels in the lamina propria, suggesting migration of antigen-presenting cells out of the intestine. In contrast, hepatic RNA-Seq analysis identified increased expression of MHC class II genes, as well as chemokines and chemokine receptors involved in macrophage recruitment (Ccr1, Ccr5, Ccl5, Cx3cr1), consistent with hepatic F4/80 labeling and macrophage infiltration into the liver. Collectively, these results suggest TCDD elicits changes that support hepatic lipid accumulation, macrophage migration, and the progression of hepatic steatosis to steatohepatitis.
Collapse
Affiliation(s)
- Kelly A Fader
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Rance Nault
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Dustin A Ammendolia
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Jack R Harkema
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824; Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824
| | - Kurt J Williams
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824
| | - Robert B Crawford
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824; and
| | - Norbert E Kaminski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824; and
| | - Dave Potter
- Wellington Laboratories Inc., Guelph, Ontario N1G 3M5, Canada
| | - Bonnie Sharratt
- Wellington Laboratories Inc., Guelph, Ontario N1G 3M5, Canada
| | - Timothy R Zacharewski
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824; *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824;
| |
Collapse
|
46
|
Nault R, Fader KA, Kirby MP, Ahmed S, Matthews J, Jones AD, Lunt SY, Zacharewski TR. Pyruvate Kinase Isoform Switching and Hepatic Metabolic Reprogramming by the Environmental Contaminant 2,3,7,8-Tetrachlorodibenzo-p-Dioxin. Toxicol Sci 2015; 149:358-71. [PMID: 26582802 DOI: 10.1093/toxsci/kfv245] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) elicits dose-dependent hepatotoxicity that includes fat accumulation, inflammation, and fibrosis that may progress to hepatocellular carcinoma. To further investigate these effects, RNA-Seq data were integrated with computationally identified putative dioxin response elements, and complementary targeted metabolomic and aryl hydrocarbon receptor (AhR) ChIP-Seq data from female C57BL/6 mice gavaged with TCDD every 4 days for 28 days. Data integration using CytoKEGG with manual curation identified dose-dependent alterations in central carbon and amino acid metabolism. More specifically, TCDD increased pyruvate kinase isoform M2 (PKM2) gene and protein expression. PKM2 has lower catalytic activity resulting in decreased glycolytic flux and the accumulation of upstream intermediates that were redirected to the pentose phosphate pathway and serine/folate biosynthesis, 2 important NADPH producing pathways stemming from glycolysis. In addition, the GAC:KGA glutaminase (GLS1) protein isoform ratio was increased, consistent with increases in glutaminolysis which serves an anaplerotic role for the TCA cycle and compensates for the reduced glycolytic flux. Collectively, gene expression, protein, and metabolite changes were indicative of increases in NADPH production in support of cytochrome P450 activity and ROS defenses. This AhR-mediated metabolic reprogramming is similar to the Warburg effect and represents a novel advantageous defense mechanism to increase anti-oxidant capacity in normal differentiated hepatocytes.
Collapse
Affiliation(s)
- Rance Nault
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, Institute for Integrative Toxicology
| | - Kelly A Fader
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, Institute for Integrative Toxicology
| | - Mathew P Kirby
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Shaimaa Ahmed
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jason Matthews
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada, Department of Nutrition, University of Oslo, Oslo, 0316, Norway, and
| | - A Daniel Jones
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, Department of Chemistry, Michigan State University, East Lansing, Michigan 48824
| | - Sophia Y Lunt
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Timothy R Zacharewski
- *Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, Institute for Integrative Toxicology,
| |
Collapse
|
47
|
Jackson DP, Joshi AD, Elferink CJ. Ah Receptor Pathway Intricacies; Signaling Through Diverse Protein Partners and DNA-Motifs. Toxicol Res (Camb) 2015; 4:1143-1158. [PMID: 26783425 PMCID: PMC4714567 DOI: 10.1039/c4tx00236a] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The Ah receptor is a transcription factor that modulates gene expression via interactions with multiple protein partners; these are reviewed, including the novel NC-XRE pathway involving KLF6.
Collapse
|
48
|
Shmarakov IO. Retinoid-xenobiotic interactions: the Ying and the Yang. Hepatobiliary Surg Nutr 2015; 4:243-67. [PMID: 26311625 DOI: 10.3978/j.issn.2304-3881.2015.05.05] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/13/2015] [Indexed: 12/20/2022]
Abstract
The literature provides compelling evidence pointing to tight metabolic interactions between retinoids and xenobiotics. These are extensive and important for understanding xenobiotic actions in the body. Within the body, retinoids affect xenobiotic metabolism and actions and conversely, xenobiotics affect retinoid metabolism and actions. This article summarizes data that establish the importance of retinoid-dependent metabolic pathways for sustaining the body's responses to xenobiotic exposure, including the roles of all-trans- and 9-cis-retinoic acid for protecting mammals from harmful xenobiotic effects and for ensuring xenobiotic elimination from the body. This review will also consider molecular mechanisms underlying xenobiotic toxicity focusing on how this may contribute to retinoid deficiency and disruption of normal retinoid homeostasis. Special attention is paid to xenobiotic molecular targets (nuclear receptors, regulatory proteins, enzymes, and transporters) which affect retinoid metabolism and signaling.
Collapse
Affiliation(s)
- Igor O Shmarakov
- Department of Biochemistry and Biotechnology, Chernivtsi National University, Chernivtsi, Ukraine
| |
Collapse
|
49
|
Tang Z, Li S, Guan X, Schmitt-Kopplin P, Lin S, Cai Z. Rapid assessment of the coenzyme Q10 redox state using ultrahigh performance liquid chromatography tandem mass spectrometry. Analyst 2015; 139:5600-4. [PMID: 25140668 DOI: 10.1039/c4an00760c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An improved method for accurate and rapid assessment of the coenzyme Q10 (CoQ10) redox state using ultrahigh performance liquid chromatography tandem mass spectrometry was described, with particular attention given to the instability of the reduced form of CoQ10 during sample preparation, chromatographic separation and mass spectrometric detection. As highly lipophilic compounds in complex biological matrices, both reduced and oxidized forms of CoQ10 were extracted simultaneously from the tissue samples by methanol which is superior to ethanol and isopropanol. After centrifugation, the supernatants were immediately separated on a C18 column with isocratic elution using methanol containing 2 mM ammonium acetate as a non-aqueous mobile phase, and detected by positive electrospray ionization tandem mass spectrometry in multiple reaction monitoring (MRM) mode. Ammonium acetate as an additive in methanol provided enhanced mass spectrometric responses for both forms of CoQ10, primarily due to stable formation of adduct ions [M + NH4](+), which served as precursor ions in positive ionization MRM transitions. The assay showed a linear range of 8.6-8585 ng mL(-1) for CoQ10H2 and 8.6-4292 ng mL(-1) for CoQ10. The limits of detection (LODs) were 7.0 and 1.0 ng mL(-1) and limits of quantification (LOQs) were 15.0 and 5.0 ng mL(-1) for CoQ10H2 and CoQ10, respectively. This rapid extractive and analytical method could avoid artificial auto-oxidation of the reduced form of CoQ10, enabling the native redox state assessment. This reliable method was also successfully applied for the measurement of the CoQ10 redox state in liver tissues of mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin, revealing the down-regulated mitochondrial electron transport chain.
Collapse
Affiliation(s)
- Zhi Tang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China.
| | | | | | | | | | | |
Collapse
|
50
|
Lee J, Prokopec SD, Watson JD, Sun RX, Pohjanvirta R, Boutros PC. Male and female mice show significant differences in hepatic transcriptomic response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. BMC Genomics 2015; 16:625. [PMID: 26290441 PMCID: PMC4546048 DOI: 10.1186/s12864-015-1840-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/13/2015] [Indexed: 12/21/2022] Open
Abstract
Background 2,3,7,8–tetrachlorodibenzo-p-dixion (TCDD) is the most potent of the dioxin congeners, capable of causing a wide range of toxic effects across numerous animal models. Previous studies have demonstrated that males and females of the same species can display divergent sensitivity phenotypes to TCDD toxicities. Although it is now clear that most TCDD-induced toxic outcomes are mediated by the aryl hydrocarbon receptor (AHR), the mechanism of differential responses to TCDD exposure between sexes remains largely unknown. To investigate the differential sensitivities in male and female mice, we profiled the hepatic transcriptomic responses 4 days following exposure to various amounts of TCDD (125, 250, 500 or 1000 μg/kg) in adult male and female C57BL/6Kuo mice. Results Several key findings were revealed by our study. 1) Hepatic transcriptomes varied significantly between the sexes at all doses examined. 2) The liver transcriptome of males was more dysregulated by TCDD than that of females. 3) The alteration of “AHR-core” genes was consistent in magnitude, regardless of sex. 4) A subset of genes demonstrated sex-dependent TCDD-induced transcriptional changes, including Fmo3 and Nr1i3, which were significantly induced in livers of male mice only. In addition, a meta-analysis was performed to contrast transcriptomic profiles of various organisms and tissues following exposure to equitoxic doses of TCDD. Minimal overlap was observed in the differences between TCDD-sensitive or TCDD-resistant models. Conclusions Sex-dependent sensitivities to TCDD exposure are associated with a set of sex-specific TCDD-responsive genes. In addition, complex interactions between the aryl hydrocarbon and sex hormone receptors may affect the observable differences in sensitivity phenotypes between the sexes. Further work is necessary to better understand the roles of those genes altered by TCDD in a sex-dependent manner, and their association with changes to sex hormones and receptors. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1840-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jamie Lee
- Informatics and Bio-computing Program, Ontario Institute for Cancer Research, Toronto, Canada.
| | - Stephenie D Prokopec
- Informatics and Bio-computing Program, Ontario Institute for Cancer Research, Toronto, Canada.
| | - John D Watson
- Informatics and Bio-computing Program, Ontario Institute for Cancer Research, Toronto, Canada.
| | - Ren X Sun
- Informatics and Bio-computing Program, Ontario Institute for Cancer Research, Toronto, Canada. .,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada.
| | - Raimo Pohjanvirta
- Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland. .,Laboratory of Toxicology, National Institute for Health and Welfare, Kuopio, Finland.
| | - Paul C Boutros
- Informatics and Bio-computing Program, Ontario Institute for Cancer Research, Toronto, Canada. .,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| |
Collapse
|