51
|
Fougerat A, Montagner A, Loiseau N, Guillou H, Wahli W. Peroxisome Proliferator-Activated Receptors and Their Novel Ligands as Candidates for the Treatment of Non-Alcoholic Fatty Liver Disease. Cells 2020; 9:E1638. [PMID: 32650421 PMCID: PMC7408116 DOI: 10.3390/cells9071638] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, frequently associated with obesity and type 2 diabetes. Steatosis is the initial stage of the disease, which is characterized by lipid accumulation in hepatocytes, which can progress to non-alcoholic steatohepatitis (NASH) with inflammation and various levels of fibrosis that further increase the risk of developing cirrhosis and hepatocellular carcinoma. The pathogenesis of NAFLD is influenced by interactions between genetic and environmental factors and involves several biological processes in multiple organs. No effective therapy is currently available for the treatment of NAFLD. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that regulate many functions that are disturbed in NAFLD, including glucose and lipid metabolism, as well as inflammation. Thus, they represent relevant clinical targets for NAFLD. In this review, we describe the determinants and mechanisms underlying the pathogenesis of NAFLD, its progression and complications, as well as the current therapeutic strategies that are employed. We also focus on the complementary and distinct roles of PPAR isotypes in many biological processes and on the effects of first-generation PPAR agonists. Finally, we review novel and safe PPAR agonists with improved efficacy and their potential use in the treatment of NAFLD.
Collapse
Affiliation(s)
- Anne Fougerat
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Alexandra Montagner
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Institut National de la Santé et de la Recherche Médicale (Inserm), Institute of Metabolic and Cardiovascular Diseases, UMR1048 Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases, University of Toulouse, UMR1048 Toulouse, France
| | - Nicolas Loiseau
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Hervé Guillou
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Walter Wahli
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
52
|
Brauner C, Joveleviths D, Álvares-da-Silva MR, Marroni N, Bona S, Schemitt E, Nardi R. Exposure to organic solvents and hepatotoxicity. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2020; 55:1173-1178. [PMID: 32602765 DOI: 10.1080/10934529.2020.1779532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 06/11/2023]
Abstract
The purpose of this study was to identify the long-term effect of chemical exposure on the liver. Laboratory tests included alanine aminotransferase (ALT) dosage and oxidative stress tests, such as thiobarbituric acid reactive substances in plasma and superoxide dismutase (SOD) and glutathione S-transferase analysis in erythrocytes. The cross-sectional study comprised 70 workers, 30 of them exposed to organic solvents and 40 not exposed. All those exposed presented at least 5 years of exposure to solvents. Hepatitis B and C, known hepatic disease, comorbidities, use of alcohol, illicit drugs or hepatotoxic medications, smoking, body mass index >30, female sex and age (<18 or >65) were excluded from the sample. Results indicated that elevated ALT was more frequent in the exposed group compared to controls: 33% vs. 10.5%, with a statistical significance (p < 0.05). Thiobarbituric acid reactive substances were significantly elevated (p < 0.01) in the exposed group in comparison to controls. Antioxidant enzymes were more elevated in the exposed group compared to controls: SOD 7.29 (4.30-8.91) USOD/mg of protein vs. 3.48 (2.98-5.28) USOD/mg of protein and GST 2.57 µmol/min/mg of protein (1.80-4.78) vs. 1.81 µmol/min/mg of protein (1.45- 2.30) µM/min/mg of protein. The results suggest an association between exposure to organic solvents and hepatotoxicity.
Collapse
Affiliation(s)
- Cristiano Brauner
- Postgraduate Program - Gastroenterology and Hepatology Sciences, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Dvora Joveleviths
- Gastroenterology and Hepatology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- FAMED, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Mário R Álvares-da-Silva
- Gastroenterology and Hepatology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- FAMED, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Norma Marroni
- Gastroenterology and Hepatology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- FAMED, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Silvia Bona
- Postgraduate Program - Gastroenterology and Hepatology Sciences, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Elizângela Schemitt
- Postgraduate Program - Gastroenterology and Hepatology Sciences, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Raissa Nardi
- Postgraduate Program - Gastroenterology and Hepatology Sciences, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
53
|
Ramaiahgari SC, Auerbach SS, Saddler TO, Rice JR, Dunlap PE, Sipes NS, DeVito MJ, Shah RR, Bushel PR, Merrick BA, Paules RS, Ferguson SS. The Power of Resolution: Contextualized Understanding of Biological Responses to Liver Injury Chemicals Using High-throughput Transcriptomics and Benchmark Concentration Modeling. Toxicol Sci 2020; 169:553-566. [PMID: 30850835 DOI: 10.1093/toxsci/kfz065] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prediction of human response to chemical exposures is a major challenge in both pharmaceutical and toxicological research. Transcriptomics has been a powerful tool to explore chemical-biological interactions, however, limited throughput, high-costs, and complexity of transcriptomic interpretations have yielded numerous studies lacking sufficient experimental context for predictive application. To address these challenges, we have utilized a novel high-throughput transcriptomics (HTT) platform, TempO-Seq, to apply the interpretive power of concentration-response modeling with exposures to 24 reference compounds in both differentiated and non-differentiated human HepaRG cell cultures. Our goals were to (1) explore transcriptomic characteristics distinguishing liver injury compounds, (2) assess impacts of differentiation state of HepaRG cells on baseline and compound-induced responses (eg, metabolically-activated), and (3) identify and resolve reference biological-response pathways through benchmark concentration (BMC) modeling. Study data revealed the predictive utility of this approach to identify human liver injury compounds by their respective BMCs in relation to human internal exposure plasma concentrations, and effectively distinguished drug analogs with varied associations of human liver injury (eg, withdrawn therapeutics trovafloxacin and troglitazone). Impacts of cellular differentiation state (proliferated vs differentiated) were revealed on baseline drug metabolizing enzyme expression, hepatic receptor signaling, and responsiveness to metabolically-activated toxicants (eg, cyclophosphamide, benzo(a)pyrene, and aflatoxin B1). Finally, concentration-response modeling enabled efficient identification and resolution of plausibly-relevant biological-response pathways through their respective pathway-level BMCs. Taken together, these findings revealed HTT paired with differentiated in vitro liver models as an effective tool to model, explore, and interpret toxicological and pharmacological interactions.
Collapse
Affiliation(s)
- Sreenivasa C Ramaiahgari
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Scott S Auerbach
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Trey O Saddler
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Julie R Rice
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Paul E Dunlap
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Nisha S Sipes
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Michael J DeVito
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Ruchir R Shah
- Sciome, LLC, Research Triangle Park, Durham, North Carolina 27709
| | - Pierre R Bushel
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709.,Division of Intramural Research, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Bruce A Merrick
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Richard S Paules
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Stephen S Ferguson
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| |
Collapse
|
54
|
Adomshick V, Pu Y, Veiga-Lopez A. Automated lipid droplet quantification system for phenotypic analysis of adipocytes using CellProfiler. Toxicol Mech Methods 2020; 30:378-387. [PMID: 32208812 DOI: 10.1080/15376516.2020.1747124] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipogenic differentiation is the process by which preadipocytes become mature adipocytes, cells that store energy and regulate metabolic homeostasis. During differentiation, neutral lipids that accumulate in adipocytes can be detected using stains and used as an index of cell differentiation. However, imaging tools for evaluating intracellular lipid droplets remain at their infancy. Nutrition, stress, or chemical exposure can dysregulate adipogenic differentiation and lipid metabolism. Therefore, the aims of this study were to develop an accurate, standardized approach to quantify lipid droplet size of mature adipocytes and a clustering approach to analyze the total lipid content per adipocyte. For the lipid droplet analysis, we used two approaches, the free online computer software of reference, ImageJ, and another free online computer software, CellProfiler. For ImageJ, we used an already developed macro designed to identify particles and quantify their area, and for CellProfiler, we developed a new analysis pipeline. Our results show that CellProfiler is able to accurately identify a greater number of lipid droplets compared to ImageJ. A clustering analysis is also possible using CellProfiler which allows for the quantification of total lipid content per individual adipocyte to provide insight into single-cell responsiveness to adipogenic stimuli. CellProfiler streamlines the lipid droplet phenotypic analysis of adipocytes compared to more traditional analysis methods. In conclusion, this novel image analysis tool can provide a more precise evaluation of lipid droplet and adipogenesis dysregulation, a critical need in the understanding of metabolic disorders.
Collapse
Affiliation(s)
- Victoria Adomshick
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Yong Pu
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | | |
Collapse
|
55
|
Matsuzaka Y, Hosaka T, Ogaito A, Yoshinari K, Uesawa Y. Prediction Model of Aryl Hydrocarbon Receptor Activation by a Novel QSAR Approach, DeepSnap-Deep Learning. Molecules 2020; 25:molecules25061317. [PMID: 32183141 PMCID: PMC7144728 DOI: 10.3390/molecules25061317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that senses environmental exogenous and endogenous ligands or xenobiotic chemicals. In particular, exposure of the liver to environmental metabolism-disrupting chemicals contributes to the development and propagation of steatosis and hepatotoxicity. However, the mechanisms for AhR-induced hepatotoxicity and tumor propagation in the liver remain to be revealed, due to the wide variety of AhR ligands. Recently, quantitative structure–activity relationship (QSAR) analysis using deep neural network (DNN) has shown superior performance for the prediction of chemical compounds. Therefore, this study proposes a novel QSAR analysis using deep learning (DL), called the DeepSnap–DL method, to construct prediction models of chemical activation of AhR. Compared with conventional machine learning (ML) techniques, such as the random forest, XGBoost, LightGBM, and CatBoost, the proposed method achieves high-performance prediction of AhR activation. Thus, the DeepSnap–DL method may be considered a useful tool for achieving high-throughput in silico evaluation of AhR-induced hepatotoxicity.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, 204-8588 Tokyo, Japan;
| | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8529, Japan; (T.H.); (A.O.); (K.Y.)
| | - Anna Ogaito
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8529, Japan; (T.H.); (A.O.); (K.Y.)
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8529, Japan; (T.H.); (A.O.); (K.Y.)
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, 204-8588 Tokyo, Japan;
- Correspondence:
| |
Collapse
|
56
|
Lee JW, Choi K, Park K, Seong C, Yu SD, Kim P. Adverse effects of perfluoroalkyl acids on fish and other aquatic organisms: A review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 707:135334. [PMID: 31874399 DOI: 10.1016/j.scitotenv.2019.135334] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 06/10/2023]
Abstract
Perfluoroalkyl acids (PFAAs) have been widely used in many industrial and consumer products. They have been detected ubiquitously in ambient water along with other environmental matrices, and their adverse effects on aquatic organisms have been a subject of active investigation. Here, we intended to summarize and synthesize the existing body of knowledge on PFAA toxicity through an extensive literature review, and shed light on areas where further research is warranted. PFAA toxicity appears to be influenced by the sex and developmental stages of aquatic organisms, but not significantly by exposure route. PFAA-induced aquatic toxicity could be classified as metabolism disturbance, reproduction disruption, oxidative stress, developmental toxicity, thyroid disruption, etc. At the molecular level, these responses can be initiated by key events, such as nuclear receptor activation, reactive oxygen species induction, or interaction with a membrane, followed by a cascade of downstream responses. PFAA-induced toxicity involves diverse metabolic processes, and therefore elucidating crosstalk or interactions among diverse metabolic pathways is a challenging task. In the presence of other chemicals, PFAAs can function as agonists or antagonists, resulting in different directions of combined toxicity. Therefore, mixture toxicity with other groups of chemicals is another research opportunity. Experimental evidence supports the trans-generational toxicity of PFAAs, suggesting that their long-term consequences for aquatic ecosystems should become of concern. A recent global ban of several PFAAs resulted in an increasing dependence on PFAA alternatives. The lack of sufficient toxicological information on this emerging group of chemicals warrant caution and rigorous toxicological assessments.
Collapse
Affiliation(s)
- Jin Wuk Lee
- Research Department of Environmental Health, National Institute of Environmental Research, Incheon 404-708, Republic of Korea
| | - Kyungho Choi
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Republic of Korea
| | - Kyunghwa Park
- Research Department of Environmental Health, National Institute of Environmental Research, Incheon 404-708, Republic of Korea
| | - Changho Seong
- Research Department of Environmental Health, National Institute of Environmental Research, Incheon 404-708, Republic of Korea
| | - Seung Do Yu
- Research Department of Environmental Health, National Institute of Environmental Research, Incheon 404-708, Republic of Korea
| | - Pilje Kim
- Research Department of Environmental Health, National Institute of Environmental Research, Incheon 404-708, Republic of Korea.
| |
Collapse
|
57
|
Lasch A, Alarcan J, Lampen A, Braeuning A, Lichtenstein D. Combinations of LXR and RXR agonists induce triglyceride accumulation in human HepaRG cells in a synergistic manner. Arch Toxicol 2020; 94:1303-1320. [DOI: 10.1007/s00204-020-02685-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/21/2020] [Indexed: 12/15/2022]
|
58
|
Understanding Environmental Contaminants' Direct Effects on Non-alcoholic Fatty Liver Disease Progression. Curr Environ Health Rep 2020; 6:95-104. [PMID: 31090041 DOI: 10.1007/s40572-019-00231-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Environmental contaminants are considered one of the major factors in the development and progression of NAFLD, the most common liver disease in the USA. RECENT FINDINGS The evolving knowledge of mechanisms of hepatic steatosis and steatohepatitis has recently been reviewed and characterized as ALD, NAFLD, and TAFLD. The most recent mechanistic studies on PFAS and PCBs have revealed a greater role for toxicants in the initiation of not only TAFLD but also NAFLD and the more progressive inflammatory stage of NAFLD, non-alcoholic steatohepatitis. In addition to insecticides, recent studies support a significant contribution of fungicides and herbicides to NAFLD. The mechanisms of PFAS, PCBs, and fungicides in contributing to the increased prevalence of NAFLD remain unclear. Addressing whether chronic, low-dose exposures could result in liver pathology and whether real-world exposure to mixtures of environmental contaminants pose a significant risk factor for NAFLD is paramount to understand the impact of NAFLD on populations today.
Collapse
|
59
|
Kennedy MC, Hart ADM, Kruisselbrink JW, van Lenthe M, de Boer WJ, van der Voet H, Rorije E, Sprong C, van Klaveren J. A retain and refine approach to cumulative risk assessment. Food Chem Toxicol 2020; 138:111223. [PMID: 32088251 DOI: 10.1016/j.fct.2020.111223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 12/27/2022]
Abstract
Mixtures of substances to which humans are exposed may lead to cumulative exposure and health effects. To study their effects, it is first necessary to identify a cumulative assessment group (CAG) of substances for risk assessment or hazard testing. Excluding substances from consideration before there is sufficient evidence may underestimate the risk. Conversely, including everything and treating the inevitable uncertainties using conservative assumptions is inefficient and may overestimate the risk, with an unknown level of protection. An efficient, transparent strategy is described to retain a large group, quantifying the uncertainty of group membership and other uncertainties. Iterative refinement of the CAG then focuses on adding information for the substances with high probability of contributing significantly to the risk. Probabilities can be estimated using expert opinion or derived from data on substance properties. An example is presented with 100 pesticides, in which the retain step identified a single substance to target refinement. Using an updated hazard characterisation for this substance reduced the mean exposure estimate from 0.43 to 0.28 μg kg-bw-1 day-1 and reduced the 99.99th percentile exposure from 24.9 to 5.1 μg kg-bw-1 day-1. Other retained substances contributed little to the risk estimates, even after accounting for uncertainty.
Collapse
Affiliation(s)
- Marc C Kennedy
- Fera Science Ltd, Sand Hutton, York, YO41 1LZ, United Kingdom.
| | - Andy D M Hart
- Fera Science Ltd, Sand Hutton, York, YO41 1LZ, United Kingdom
| | - Johannes W Kruisselbrink
- Wageningen University & Research, Biometrics, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Marco van Lenthe
- Wageningen University & Research, Biometrics, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Waldo J de Boer
- Wageningen University & Research, Biometrics, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Hilko van der Voet
- Wageningen University & Research, Biometrics, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Emiel Rorije
- RIVM, National Institute for Public Health and the Environment, The Netherlands, PO Box 1, 3720 BA, Bilthoven, the Netherlands
| | - Corinne Sprong
- RIVM, National Institute for Public Health and the Environment, The Netherlands, PO Box 1, 3720 BA, Bilthoven, the Netherlands
| | - Jacob van Klaveren
- RIVM, National Institute for Public Health and the Environment, The Netherlands, PO Box 1, 3720 BA, Bilthoven, the Netherlands
| |
Collapse
|
60
|
Wahlang B, Appana S, Falkner KC, McClain CJ, Brock G, Cave MC. Insecticide and metal exposures are associated with a surrogate biomarker for non-alcoholic fatty liver disease in the National Health and Nutrition Examination Survey 2003-2004. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:6476-6487. [PMID: 31873887 PMCID: PMC7047555 DOI: 10.1007/s11356-019-07066-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/13/2019] [Indexed: 04/16/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), the most common form of liver disease, affects over 30% of the US population. Our group and others have previously demonstrated that low-level environmental pollutant exposures were associated with increased odds ratios for unexplained alanine aminotransferase (ALT) elevation, a surrogate biomarker for NAFLD, in the adult National Health and Nutrition Examination Survey (NHANES). However, recently, more sensitive and lower ALT cutoffs have been proposed. The objective of this observational study is to utilize these ALT cutoffs to determine new associations between environmental chemicals and the surrogate NAFLD biomarker. Adult NHANES 2003-2004 participants without viral hepatitis, hemochromatosis, or alcoholic liver disease were analyzed in this cross-sectional study. ALT elevation was defined as > 30 IU/L in men and > 19 IU/L in women. Odds ratios adjusted for potential confounders for ALT elevation were determined across exposure quartiles for 17 pollutant subclasses comprised of 111 individual pollutants. The overall prevalence of ALT elevation was 37.6%. Heavy metal and organochlorine insecticide subclasses were associated with dose-dependent increased adjusted odds ratios for ALT elevation of 1.6 (95% CI 1.2-2.3) and 3.5 (95% CI 2.3-5.5) respectively, for the highest vs. lowest exposure quartiles (ptrend < 0.01). Within these subclasses, increasing whole blood levels of lead and mercury, and lipid-adjusted serum levels of dieldrin, and the chlordane metabolites, heptachlor epoxide, and trans-nonachlor, were associated with increased odds ratios for ALT elevation. In conclusion, organochlorine insecticide, lead, and mercury exposures were associated with ALT elevation and suspected NAFLD in adult NHANES 2003-2004.
Collapse
Affiliation(s)
- Banrida Wahlang
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Louisville, Louisville, 40202, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Savitri Appana
- School of Public Health, University of Louisville, Department of Bioinformatics and Biostatistics, Louisville, 40202, KY, USA
- Department of Biostatistics, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, 77030, TX, USA
| | - K Cameron Falkner
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Louisville, Louisville, 40202, USA
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville School of Medicine, Kosair Charities Clinical and Translational Research Building 505 S. Hancock St., Louisville, 40202, KY, USA
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Louisville, Louisville, 40202, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville School of Medicine, Kosair Charities Clinical and Translational Research Building 505 S. Hancock St., Louisville, 40202, KY, USA
- The Robley Rex Veterans Affairs Medical Center, 800 Zorn Ave, Louisville, KY, USA
| | - Guy Brock
- School of Public Health, University of Louisville, Department of Bioinformatics and Biostatistics, Louisville, 40202, KY, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, 43210, OH, USA
| | - Matthew C Cave
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Louisville, Louisville, 40202, USA.
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA.
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville School of Medicine, Kosair Charities Clinical and Translational Research Building 505 S. Hancock St., Louisville, 40202, KY, USA.
- The Robley Rex Veterans Affairs Medical Center, 800 Zorn Ave, Louisville, KY, USA.
| |
Collapse
|
61
|
Jin R, McConnell R, Catherine C, Xu S, Walker DI, Stratakis N, Jones DP, Miller GW, Peng C, Conti DV, Vos MB, Chatzi L. Perfluoroalkyl substances and severity of nonalcoholic fatty liver in Children: An untargeted metabolomics approach. ENVIRONMENT INTERNATIONAL 2020; 134:105220. [PMID: 31744629 PMCID: PMC6944061 DOI: 10.1016/j.envint.2019.105220] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 05/08/2023]
Abstract
BACKGROUND Toxicant-associated steatohepatitis has been described in adults but less is known regarding the role of toxicants in liver disease of children. Perfluoroalkyl substances (PFAS) cause hepatic steatosis in rodents, but few previous studies have examined PFAS effects on severity of liver injury in children. OBJECTIVES We aimed to examine the relationship of PFAS to histologic severity of nonalcoholic fatty liver disease (NAFLD) in children. METHODS Seventy-four children with physician-diagnosed NAFLD were recruited from Children's Healthcare of Atlanta between 2007 and 2015. Biopsy-based liver histological features were scored for steatosis, lobular and portal inflammation, ballooning, and fibrosis. Plasma concentrations of perfluorooctanoic acid (PFOA), perfluorooctane sulfonate (PFOS) and perfluorohexane sulfonic acid (PFHxS), and untargeted plasma metabolomic profiling, were determined using liquid chromatography with high-resolution mass spectrometry. A metabolome-wide association study coupled with pathway enrichment analysis was performed to evaluate metabolic dysregulation associated with PFAS. A structural integrated analysis was applied to identify latent clusters of children with more severe form of NAFLD based on their PFAS levels and metabolite pattern. RESULTS Patients were 7-19 years old, mostly boys (71%), Hispanic (51%), and obese (85%). The odds of having nonalcoholic steatohepatitis (NASH), compared to children with steatosis alone, was significantly increased with each interquartile range (IQR) increase of PFOS (OR: 3.32, 95% CI: 1.40-7.87) and PFHxS (OR: 4.18, 95% CI: 1.64-10.7). Each IQR increase of PFHxS was associated with increased odds for liver fibrosis (OR: 4.44, 95% CI: 1.34-14.8), lobular inflammation (OR: 2.87, 95% CI: 1.12-7.31), and higher NAFLD activity score (β coefficient 0.46; 95% CI: 0.03, 0.89). A novel integrative analysis identified a cluster of children with NASH, characterized by increased PFAS levels and altered metabolite patterns including higher plasma levels of phosphoethanolamine, tyrosine, phenylalanine, aspartate and creatine, and decreased plasma levels of betaine. CONCLUSIONS Ηigher PFAS exposure was associated with more severe disease in children with NAFLD. PFAS may be an important toxicant contributing to NAFLD progression; however larger, longitudinal studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Ran Jin
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Rob McConnell
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Cioffi Catherine
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, USA.
| | - Shujing Xu
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Douglas I Walker
- Clinical Biomarkers Laboratory, Division of Pulmonary Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA; Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | - Nikos Stratakis
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA.
| | - Gary W Miller
- Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | - Cheng Peng
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| | - David V Conti
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Miriam B Vos
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Leda Chatzi
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
62
|
Kim JJ, Kumar S, Kumar V, Lee YM, Kim YS, Kumar V. Bisphenols as a Legacy Pollutant, and Their Effects on Organ Vulnerability. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 17:E112. [PMID: 31877889 PMCID: PMC6982222 DOI: 10.3390/ijerph17010112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022]
Abstract
Bisphenols are widely used in the synthesis of polycarbonate plastics, epoxy resins, and thermal paper, which are used in manufacturing items of daily use. Packaged foods and drinks are the main sources of exposure to bisphenols. These chemicals affect humans and animals by disrupting the estrogen, androgen, progesterone, thyroid, and aryl hydrocarbon receptor functions. Bisphenols exert numerous harmful effects because of their interaction with receptors, reactive oxygen species (ROS) formation, lipid peroxidation, mitochondrial dysfunction, and cell signal alterations. Both cohort and case-control studies have determined an association between bisphenol exposure and increased risk of cardiovascular diseases, neurological disorders, reproductive abnormalities, obesity, and diabetes. Prenatal exposure to bisphenols results in developmental disorders in animals. These chemicals also affect the immune cells and play a significant role in initiating the inflammatory response. Exposure to bisphenols exhibit age, gender, and dose-dependent effects. Even at low concentrations, bisphenols exert toxicity, and hence deserve a critical assessment of their uses. Since bisphenols have a global influence on human health, the need to discover the underlying pathways involved in all disease conditions is essential. Furthermore, it is important to promote the use of alternatives for bisphenols, thereby restricting their uses.
Collapse
Affiliation(s)
- Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea; (J.-J.K.); (Y.-M.L.); (Y.-S.K.)
| | - Surendra Kumar
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Vinay Kumar
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh 758307, Vietnam;
| | - Yun-Mi Lee
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea; (J.-J.K.); (Y.-M.L.); (Y.-S.K.)
| | - You-Sam Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea; (J.-J.K.); (Y.-M.L.); (Y.-S.K.)
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea; (J.-J.K.); (Y.-M.L.); (Y.-S.K.)
| |
Collapse
|
63
|
Olsvik P, Hammer S, Sanden M, Søfteland L. Chlorpyrifos-induced dysfunction of lipid metabolism is not restored by supplementation of polyunsaturated fatty acids EPA and ARA in Atlantic salmon liver cells. Toxicol In Vitro 2019; 61:104655. [DOI: 10.1016/j.tiv.2019.104655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/06/2019] [Accepted: 09/15/2019] [Indexed: 12/22/2022]
|
64
|
Abedini JA, Handa S, Edwards S, Chorley B, El-Masri H. Identification of differentially expressed genes and networks related to hepatic lipid dysfunction. Toxicol Appl Pharmacol 2019; 382:114757. [DOI: 10.1016/j.taap.2019.114757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 12/20/2022]
|
65
|
AbdulHameed MDM, Pannala VR, Wallqvist A. Mining Public Toxicogenomic Data Reveals Insights and Challenges in Delineating Liver Steatosis Adverse Outcome Pathways. Front Genet 2019; 10:1007. [PMID: 31681434 PMCID: PMC6813744 DOI: 10.3389/fgene.2019.01007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/23/2019] [Indexed: 12/19/2022] Open
Abstract
Exposure to chemicals contributes to the development and progression of fatty liver, or steatosis, a process characterized by abnormal accumulation of lipids within liver cells. However, lack of knowledge on how chemicals cause steatosis has prevented any large-scale assessment of the 80,000+ chemicals in current use. To address this gap, we mined a large, publicly available toxicogenomic dataset associated with 18 known steatogenic chemicals to assess responses across assays (in vitro and in vivo) and species (i.e., rats and humans). We identified genes that were differentially expressed (DEGs) in rat in vivo, rat in vitro, and human in vitro studies in which rats or in vitro primary cell lines were exposed to the chemicals at different doses and durations. Using these DEGs, we performed pathway enrichment analysis, analyzed the molecular initiating events (MIEs) of the steatosis adverse outcome pathway (AOP), and predicted metabolite changes using metabolic network analysis. Genes indicative of oxidative stress were among the DEGs most frequently observed in the rat in vivo studies. Nox4, a pro-fibrotic gene, was down-regulated across these chemical exposure conditions. We identified eight genes (Cyp1a1, Egr1, Ccnb1, Gdf15, Cdk1, Pdk4, Ccna2, and Ns5atp9) and one pathway (retinol metabolism), associated with steatogenic chemicals and whose response was conserved across the three in vitro and in vivo systems. Similarly, we found the predicted metabolite changes, such as increases of saturated and unsaturated fatty acids, conserved across the three systems. Analysis of the target genes associated with the MIEs of the current steatosis AOP did not provide a clear association between these 18 chemicals and the MIEs, underlining the multi-factorial nature of this disease. Notably, our overall analysis implicated mitochondrial toxicity as an important and overlooked MIE for chemical-induced steatosis. The integrated toxicogenomics approach to identify genes, pathways, and metabolites based on known steatogenic chemicals, provide an important mean to assess development of AOPs and gauging the relevance of new testing strategies.
Collapse
Affiliation(s)
- Mohamed Diwan M AbdulHameed
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD, United States.,The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Venkat R Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD, United States.,The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD, United States
| |
Collapse
|
66
|
Tsatsakis A, Docea AO, Constantin C, Calina D, Zlatian O, Nikolouzakis TK, Stivaktakis PD, Kalogeraki A, Liesivuori J, Tzanakakis G, Neagu M. Genotoxic, cytotoxic, and cytopathological effects in rats exposed for 18 months to a mixture of 13 chemicals in doses below NOAEL levels. Toxicol Lett 2019; 316:154-170. [PMID: 31521832 DOI: 10.1016/j.toxlet.2019.09.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 12/13/2022]
Abstract
The present study investigates the genotoxic and cytotoxic effects of long term exposure to low doses of a mixture consisting of methomyl, triadimefon, dimethoate, glyphosate, carbaryl, methyl parathion, aspartame, sodium benzoate, EDTA, ethylparaben, buthylparaben, bisphenol A and acacia gum in rats. Four groups of ten Sprangue Dawley rats (5 males and 5 females per group) were exposed for 18 months to the mixture in doses of 0xNOAEL, 0.0025xNOAEL, 0.01xNOAEL and 0.05xNOAEL (mg/kg bw/day). After 18 months of exposure, the rats were sacrificed and their organs were harvested. Micronuclei frequency was evaluated in bone marrow erythrocytes whereas the organs were cytopathologically examined by the touch preparation technique. The exposure to the mixture caused a genotoxic effect identified only in females. Cytopathological examination showed specific alterations of tissue organization in a tissue-type dependent manner. The observed effects were dose-dependent and correlated to various tissue parameters. Specifically, testes samples revealed degenerative and cellularity disorders, liver hepatocytes exhibited decreased glycogen deposition whereas degenerative changes were present in gastric cells. Lung tissue presented increased inflammatory cells infiltration and alveolar macrophages with enhanced phagocytic activity, whereas brain tissue exhibited changes in glial and astrocyte cells' numbers. In conclusion, exposure to very low doses of the tested mixture for 18 months induces genotoxic effects as well as monotonic cytotoxic effects in a tissue-dependent manner.
Collapse
Affiliation(s)
- Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece; Spin-Off Toxplus S.A., 71601, Heraklion, Greece.
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy, Faculty of Pharmacy, Craiova, 200349, Romania.
| | - Carolina Constantin
- Department of Immunology, Victor Babes National Institute of Pathology, Bucharest, Romania; Department of Pathology Dept. Colentina Clinical Hospital, Bucharest, Romania.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy, Faculty of Pharmacy, Craiova, 200349, Romania.
| | - Ovidiu Zlatian
- Department of Microbiology, University of Medicine and Pharmacy, Faculty of Pharmacy, Craiova, 200349, Romania.
| | | | - Polychronis D Stivaktakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece.
| | - Alexandra Kalogeraki
- Department of Pathology-Cytopathology, Medical School, University of Crete, Heraklion, Crete, Greece.
| | | | - George Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71003, Heraklion, Greece.
| | - Monica Neagu
- Department of Immunology, Victor Babes National Institute of Pathology, Bucharest, Romania; Department of Pathology Dept. Colentina Clinical Hospital, Bucharest, Romania.
| |
Collapse
|
67
|
Ochiai M, Iida M, Agusa T, Takaguchi K, Fujii S, Nomiyama K, Iwata H. Effects of 4-Hydroxy-2,3,3',4',5-Pentachlorobiphenyl (4-OH-CB107) on Liver Transcriptome in Rats: Implication in the Disruption of Circadian Rhythm and Fatty Acid Metabolism. Toxicol Sci 2019; 165:118-130. [PMID: 29788408 DOI: 10.1093/toxsci/kfy123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) and their hydroxylated metabolites (OH-PCBs) have been detected in tissues of both wild animals and humans. Several previous studies have suggested adverse effects of OH-PCBs on the endocrine and nervous systems in mammals. However, there have been no studies on transcriptome analysis of the effects of OH-PCBs, and thus, the whole picture and mechanisms underlying the adverse effects induced by OH-PCBs are still poorly understood. We therefore investigated the mRNA expression profile in the liver of adult male Wistar rats treated with 4-hydroxy-2,3,3',4',5-pentachlorobiphenyl (4-OH-CB107) to explore the genes responsive to OH-PCBs and to understand the potential effects of the chemical. Next-generation RNA sequencing analysis revealed changes in the expression of genes involved in the circadian rhythm and fatty acid metabolism, such as nuclear receptor subfamily 1, group D, member 1, aryl hydrocarbon receptor nuclear translocator-like protein 1, cryptochrome circadian clock 1, and enoyl-CoA hydratase and 3-hydroxyacyl-CoA dehydrogenase, in 4-OH-CB107-treated rats. In addition, biochemical analysis of the plasma revealed a dose-dependent increase in the leucine aminopeptidase, indicating the onset of liver damage. These results suggest that OH-PCB exposure may induce liver injury as well as disrupt the circadian rhythm and peroxisome proliferator-activated receptor-related fatty acid metabolism.
Collapse
Affiliation(s)
- Mari Ochiai
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Midori Iida
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime 790-8577, Japan
- Department of Bioscience and Bioinformatics, Kyusyu Institute of Technology, Iizuka, Fukuoka 820-0067, Japan
| | - Tetsuro Agusa
- Graduate School of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, Kumamoto 862-8502, Japan
| | - Kohki Takaguchi
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Satoshi Fujii
- Department of Bioscience and Bioinformatics, Kyusyu Institute of Technology, Iizuka, Fukuoka 820-0067, Japan
| | - Kei Nomiyama
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime 790-8577, Japan
| |
Collapse
|
68
|
Wahlang B, Jin J, Beier JI, Hardesty JE, Daly EF, Schnegelberger RD, Falkner KC, Prough RA, Kirpich IA, Cave MC. Mechanisms of Environmental Contributions to Fatty Liver Disease. Curr Environ Health Rep 2019; 6:80-94. [PMID: 31134516 PMCID: PMC6698418 DOI: 10.1007/s40572-019-00232-w] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Fatty liver disease (FLD) affects over 25% of the global population and may lead to liver-related mortality due to cirrhosis and liver cancer. FLD caused by occupational and environmental chemical exposures is termed "toxicant-associated steatohepatitis" (TASH). The current review addresses the scientific progress made in the mechanistic understanding of TASH since its initial description in 2010. RECENT FINDINGS Recently discovered modes of actions for volatile organic compounds and persistent organic pollutants include the following: (i) the endocrine-, metabolism-, and signaling-disrupting chemical hypotheses; (ii) chemical-nutrient interactions and the "two-hit" hypothesis. These key hypotheses were then reviewed in the context of the steatosis adverse outcome pathway (AOP) proposed by the US Environmental Protection Agency. The conceptual understanding of the contribution of environmental exposures to FLD has progressed significantly. However, because this is a new research area, more studies including mechanistic human data are required to address current knowledge gaps.
Collapse
Affiliation(s)
- Banrida Wahlang
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY, 40202, USA
| | - Jian Jin
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Juliane I Beier
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Josiah E Hardesty
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Erica F Daly
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Regina D Schnegelberger
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - K Cameron Falkner
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Irina A Kirpich
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Hepatobiology & Toxicology COBRE Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY, 40202, USA
| | - Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY, 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Hepatobiology & Toxicology COBRE Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY, 40202, USA.
- The Robley Rex Veterans Affairs Medical Center, Louisville, KY, 40206, USA.
- The Jewish Hospital Liver Transplant Program, Louisville, KY, 40202, USA.
- Kosair Charities Clinical & Translational Research Building, 505 South Hancock Street, Louisville, KY, 40202, USA.
| |
Collapse
|
69
|
DNA Hydroxymethylation at the Interface of the Environment and Nonalcoholic Fatty Liver Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16152791. [PMID: 31387232 PMCID: PMC6695744 DOI: 10.3390/ijerph16152791] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 12/25/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent forms of chronic liver disorders among adults, children, and adolescents, and a growing epidemic, worldwide. Notwithstanding the known susceptibility factors for NAFLD, i.e., obesity and metabolic syndrome, the exact cause(s) of this disease and the underlying mechanisms of its initiation and progression are not fully elucidated. NAFLD is a multi-faceted disease with metabolic, genetic, epigenetic, and environmental determinants. Accumulating evidence shows that exposure to environmental toxicants contributes to the development of NAFLD by promoting mitochondrial dysfunction and generating reactive oxygen species in the liver. Imbalances in the redox state of the cells are known to cause alterations in the patterns of 5-hydroxymethylcytosine (5hmC), the oxidative product of 5-methylcytosine (5mC), thereby influencing gene regulation. The 5hmC-mediated deregulation of genes involved in hepatic metabolism is an emerging area of research in NAFLD. This review summarizes our current knowledge on the interactive role of xenobiotic exposure and DNA hydroxymethylation in the pathogenesis of fatty liver disease. Increasing the mechanistic knowledge of NAFLD initiation and progression is crucial for the development of new and effective strategies for prevention and treatment of this disease.
Collapse
|
70
|
van Meteren N, Lagadic-Gossmann D, Chevanne M, Gallais I, Gobart D, Burel A, Bucher S, Grova N, Fromenty B, Appenzeller BMR, Chevance S, Gauffre F, Le Ferrec E, Sergent O. Polycyclic aromatic hydrocarbons can trigger hepatocyte release of extracellular vesicles by various mechanisms of action depending on their affinity for the aryl hydrocarbon receptor. Toxicol Sci 2019; 171:443-462. [PMID: 31368503 DOI: 10.1093/toxsci/kfz157] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane enclosed nanostructures released by cells into the extracellular environment. As major actors of physiological intercellular communication, they have been shown to be pathogenic mediators of several liver diseases. EVs also appear to be potential actors of drug-induced liver injury, but nothing is known concerning environmental pollutants. We aimed to study the impact of polycyclic aromatic hydrocarbons (PAHs), major contaminants, on hepatocyte-derived EV production, with a special focus on hepatocyte death. Three PAHs were selected, based on their presence in food and their affinity for the aryl hydrocarbon receptor (AhR): benzo(a)pyrene (BP), dibenzo(a,h)anthracene (DBA), and pyrene (PYR). Treatment of primary rat and WIF-B9 hepatocytes by all three PAHs increased the release of EVs, mainly comprised of exosomes, in parallel with modifying exosome protein marker expression and inducing apoptosis. Moreover, PAH treatment of rodents for three months also led to increased EV levels in plasma. The EV release involved CYP metabolism and the activation of the transcription factor, the AhR, for BP and DBA and another transcription factor, the constitutive androstane receptor (CAR), for PYR. Furthermore, all PAHs increased cholesterol levels in EVs but only BP and DBA were able to reduce the cholesterol content of total cell membranes. All cholesterol changes very likely participated in the increase in EV release and cell death. Finally, we studied changes in cell membrane fluidity caused by BP and DBA due to cholesterol depletion. Our data showed increased cell membrane fluidity, which contributed to hepatocyte EV release and cell death.
Collapse
Affiliation(s)
- Nettie van Meteren
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Dimitri Gobart
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Agnès Burel
- Univ Rennes, Biosit - UMS 3480, US_S 018, F-35000 Rennes, France
| | - Simon Bucher
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer)-UMR_S1241, UMR_A 1341, F-35000 Rennes, France
| | - Nathalie Grova
- Department of Infection and Immunity, Luxembourg Institute of Health, Immune Endocrine Epigenetics Research Group, L-4354 Esch-sur-Alzette, Luxembourg
- Calbinotox, Faculty of Science and Technology, Lorraine University, F-54506 Vandoeuvre-les-Nancy, France
| | - Bernard Fromenty
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer)-UMR_S1241, UMR_A 1341, F-35000 Rennes, France
| | - Brice M R Appenzeller
- Human Biomonitoring Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Soizic Chevance
- Univ Rennes, CNRS, ISCR (Institut des sciences chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Fabienne Gauffre
- Univ Rennes, CNRS, ISCR (Institut des sciences chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Eric Le Ferrec
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
71
|
Arman T, Lynch KD, Montonye ML, Goedken M, Clarke JD. Sub-Chronic Microcystin-LR Liver Toxicity in Preexisting Diet-Induced Nonalcoholic Steatohepatitis in Rats. Toxins (Basel) 2019; 11:E398. [PMID: 31323923 PMCID: PMC6669744 DOI: 10.3390/toxins11070398] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023] Open
Abstract
Microcystin-LR (MCLR) is a hepatotoxic cyanotoxin reported to cause a phenotype similar to nonalcoholic steatohepatitis (NASH). NASH is a common progressive liver disease that advances in severity due to exogenous stressors such as poor diet and toxicant exposure. Our objective was to determine how sub-chronic MCLR toxicity affects preexisting diet-induced NASH. Sprague-Dawley rats were fed one of three diets for 10 weeks: control, methionine and choline deficient (MCD), or high fat/high cholesterol (HFHC). After six weeks of diet, animals received vehicle, 10 µg/kg, or 30 µg/kg MCLR via intraperitoneal injection every other day for the final 4 weeks. Incidence and severity scoring of histopathology endpoints suggested that MCLR toxicity drove NASH to a less fatty and more fibrotic state. In general, expression of genes involved in de novo lipogenesis and fatty acid esterification were altered in favor of decreased steatosis. The higher MCLR dose increased expression of genes involved in fibrosis and inflammation in the control and HFHC groups. These data suggest MCLR toxicity in the context of preexisting NASH may drive the liver to a more severe phenotype that resembles burnt-out NASH.
Collapse
Affiliation(s)
- Tarana Arman
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Katherine D Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Michelle L Montonye
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Michael Goedken
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08901, USA
| | - John D Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
72
|
Abstract
Polychlorinated biphenyls (PCBs) have been associated with abnormal liver enzymes and suspected nonalcoholic fatty liver disease (NAFLD) in cohort studies. NAFLD affects greater than 25% of the global population and may result in liver-related mortality. Both dioxin-like and non-dioxin-like PCBs have been associated with NAFLD, but their effects and mechanisms differ. Dioxin-like PCBs altered the gut:liver axis and microbiome and caused hepatic steatosis by disrupting hepatic lipid metabolism. In contrast, NDL PCBs reduced the liver's protective responses to promote diet-induced NAFLD. Mechanisms included the disruption of phosphoprotein signaling resulting in altered nuclear receptor function.
Collapse
|
73
|
Thomas RS, Bahadori T, Buckley TJ, Cowden J, Deisenroth C, Dionisio KL, Frithsen JB, Grulke CM, Gwinn MR, Harrill JA, Higuchi M, Houck KA, Hughes MF, Hunter ES, Isaacs KK, Judson RS, Knudsen TB, Lambert JC, Linnenbrink M, Martin TM, Newton SR, Padilla S, Patlewicz G, Paul-Friedman K, Phillips KA, Richard AM, Sams R, Shafer TJ, Setzer RW, Shah I, Simmons JE, Simmons SO, Singh A, Sobus JR, Strynar M, Swank A, Tornero-Valez R, Ulrich EM, Villeneuve DL, Wambaugh JF, Wetmore BA, Williams AJ. The Next Generation Blueprint of Computational Toxicology at the U.S. Environmental Protection Agency. Toxicol Sci 2019; 169:317-332. [PMID: 30835285 PMCID: PMC6542711 DOI: 10.1093/toxsci/kfz058] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The U.S. Environmental Protection Agency (EPA) is faced with the challenge of efficiently and credibly evaluating chemical safety often with limited or no available toxicity data. The expanding number of chemicals found in commerce and the environment, coupled with time and resource requirements for traditional toxicity testing and exposure characterization, continue to underscore the need for new approaches. In 2005, EPA charted a new course to address this challenge by embracing computational toxicology (CompTox) and investing in the technologies and capabilities to push the field forward. The return on this investment has been demonstrated through results and applications across a range of human and environmental health problems, as well as initial application to regulatory decision-making within programs such as the EPA's Endocrine Disruptor Screening Program. The CompTox initiative at EPA is more than a decade old. This manuscript presents a blueprint to guide the strategic and operational direction over the next 5 years. The primary goal is to obtain broader acceptance of the CompTox approaches for application to higher tier regulatory decisions, such as chemical assessments. To achieve this goal, the blueprint expands and refines the use of high-throughput and computational modeling approaches to transform the components in chemical risk assessment, while systematically addressing key challenges that have hindered progress. In addition, the blueprint outlines additional investments in cross-cutting efforts to characterize uncertainty and variability, develop software and information technology tools, provide outreach and training, and establish scientific confidence for application to different public health and environmental regulatory decisions.
Collapse
Affiliation(s)
- Russell S. Thomas
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Tina Bahadori
- National Center for Environmental Assessment, Office of Research and Development, US Environmental Protection Agency
| | - Timothy J. Buckley
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - John Cowden
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Chad Deisenroth
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Kathie L. Dionisio
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Jeffrey B. Frithsen
- Chemical Safety for Sustainability National Research Program, Office of Research and Development, US Environmental Protection Agency
| | - Christopher M. Grulke
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Maureen R. Gwinn
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Joshua A. Harrill
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Mark Higuchi
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Keith A. Houck
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Michael F. Hughes
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - E. Sidney Hunter
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Kristin K. Isaacs
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Richard S. Judson
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Thomas B. Knudsen
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Jason C. Lambert
- National Center for Environmental Assessment, Office of Research and Development, US Environmental Protection Agency
| | - Monica Linnenbrink
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Todd M. Martin
- National Risk Management Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Seth R. Newton
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Stephanie Padilla
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Grace Patlewicz
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Katie Paul-Friedman
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Katherine A. Phillips
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Ann M. Richard
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Reeder Sams
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Timothy J. Shafer
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - R. Woodrow Setzer
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Imran Shah
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Jane E. Simmons
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Steven O. Simmons
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Amar Singh
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Jon R. Sobus
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Mark Strynar
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Adam Swank
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Rogelio Tornero-Valez
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Elin M. Ulrich
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Daniel L Villeneuve
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - John F. Wambaugh
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| | - Barbara A. Wetmore
- National Exposure Research Laboratory, Office of Research and Development, US Environmental Protection Agency
| | - Antony J. Williams
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency
| |
Collapse
|
74
|
Knebel C, Buhrke T, Süssmuth R, Lampen A, Marx-Stoelting P, Braeuning A. Pregnane X receptor mediates steatotic effects of propiconazole and tebuconazole in human liver cell lines. Arch Toxicol 2019; 93:1311-1322. [PMID: 30989312 DOI: 10.1007/s00204-019-02445-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022]
Abstract
Triazoles are commonly used fungicides which show liver toxicity in rodent studies. While hepatocellular hypertrophy is the most prominent finding, some triazoles have also been reported to cause hepatocellular steatosis. The aim of our study was to elucidate molecular mechanisms of triazole-mediated steatosis. Therefore, we used the two triazoles propiconazole (Pi) and tebuconazole (Te) as test compounds in in vitro assays using the human hepatocarcinoma cell lines HepG2 and HepaRG. Triglyceride accumulation was measured using the Adipored assay and by a gas-chromatographic method. Reporter gene analyses were used to assess the ability of Pi and Te to activate nuclear receptors, which are described as the molecular initiators in the adverse outcome pathway (AOP) for liver steatosis. The expression of steatosis-associated genes was investigated by RT-PCR. Mechanistic analyses of triazole-mediated steatosis were performed using HepaRG subclones that are deficient in different nuclear receptors. Pi and Te both interacted with the constitutive androstane receptor (CAR), the peroxisome proliferator-activated receptor alpha (PPARα), and the pregnane X receptor (PXR). Both compounds induced expression of steatosis-related genes and cellular triglyceride accumulation. The knockout of PXR in HepaRG cells, but not the CAR knockout, abolished triazole-induced triglyceride accumulation, thus underlining the crucial role of PXR in hepatic steatosis resulting from exposure to these fungicides. In conclusion, our findings provide new insight into the molecular mechanisms of steatosis induction by triazole fungicides and identify PXR as a critical mediator of this process.
Collapse
Affiliation(s)
- Constanze Knebel
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Thorsten Buhrke
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Roderich Süssmuth
- Institute of Chemistry, Technical University Berlin, Straße des 17.Juni 124, 10623, Berlin, Germany
| | - Alfonso Lampen
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- Department Pesticides Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| |
Collapse
|
75
|
Clarke JD, Dzierlenga A, Arman T, Toth E, Li H, Lynch KD, Tian DD, Goedken M, Paine MF, Cherrington N. Nonalcoholic fatty liver disease alters microcystin-LR toxicokinetics and acute toxicity. Toxicon 2019; 162:1-8. [PMID: 30849452 PMCID: PMC6447445 DOI: 10.1016/j.toxicon.2019.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 02/27/2019] [Accepted: 03/03/2019] [Indexed: 02/08/2023]
Abstract
Microcystin-LR (MCLR) is a cyanotoxin produced by blue-green algae that causes liver and kidney toxicities. MCLR toxicity is dependent on cellular uptake through the organic anion transporting polypeptide (OATP) transporters. Nonalcoholic fatty liver disease (NAFLD) progresses through multiple stages, alters expression of hepatic OATPs, and is associated with chronic kidney disease. The purpose of this study was to determine whether NAFLD increases systemic exposure to MCLR and influences acute liver and kidney toxicities. Rats were fed a control diet or two dietary models of NAFLD; methionine and choline deficient (MCD) or high fat/high cholesterol (HFHC). Two studies were performed in these groups: 1) a single dose intravenous toxicokinetic study (20 μg/kg), and 2) a single dose intraperitoneal toxicity study (60 μg/kg). Compared to control rats, plasma MCLR area under the concentration-time curve (AUC) in MCD rats doubled, whereas biliary clearance (Clbil) was unchanged; in contrast, plasma AUC in HFHC rats was unchanged, whereas Clbil approximately doubled. Less MCLR bound to PP2A was observed in the liver of MCD rats. This shift in exposure decreased the severity of liver pathology only in the MCD rats after a single toxic dose of MCLR (60 μg/kg). In contrast, the single toxic dose of MCLR increased hepatic inflammation, plasma cholesterol, proteinuria, and urinary KIM1 in HFHC rats more than MCLR exposed control rats. In conclusion, rodent models of NAFLD alter MCLR toxicokinetics and acute toxicity and may have implications for liver and kidney pathologies in NAFLD patients.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
| | - Anika Dzierlenga
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Tarana Arman
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Erica Toth
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Hui Li
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Katherine D Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Dan-Dan Tian
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Michael Goedken
- Rutgers Translational Sciences, Rutgers University, Piscataway, NJ, 08901, USA
| | - Mary F Paine
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Nathan Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
76
|
Filis P, Walker N, Robertson L, Eaton-Turner E, Ramona L, Bellingham M, Amezaga MR, Zhang Z, Mandon-Pepin B, Evans NP, Sharpe RM, Cotinot C, Rees WD, O'Shaughnessy P, Fowler PA. Long-term exposure to chemicals in sewage sludge fertilizer alters liver lipid content in females and cancer marker expression in males. ENVIRONMENT INTERNATIONAL 2019; 124:98-108. [PMID: 30641261 DOI: 10.1016/j.envint.2019.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/01/2018] [Accepted: 01/03/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The increased incidence of diseases, including metabolic syndrome and infertility, may be related to exposure to the mixture of chemicals, which are ubiquitous in the modern environment (environmental chemicals, ECs). Xeno-detoxification occurs within the liver which is also the source of many plasma proteins and growth factors and plays an important role in the regulation of homeostasis. OBJECTIVES The objective of this study was to investigate the effects of ECs on aspects of liver function, in a well characterized ovine model of exposure to a real-life EC mixture. METHODS Four groups of sheep (n = 10-12/sex/treatment) were maintained long-term on control or sewage sludge-fertilized pastures: from conception to culling at 19 months of age in females and from conception to 7 months of age and thereafter in control plots until culling at 19 months of age in males. Environmental chemicals were measured in sheep livers and RNA and protein extracts were assessed for exposure markers. Liver proteins were resolved using 2D differential in-gel electrophoresis and differentially expressed protein spots were identified by liquid chromatography/tandem mass spectroscopy. RESULTS Higher levels of polycyclic aromatic hydrocarbons (PAHs) and lower levels of polychlorinated biphenyls (PCBs) in the livers of control males compared to control females indicated sexually dimorphic EC body burdens. Increased levels of the PAHs Benzo[a]anthracene and chrysene and reduced levels of PCB 153 and PCB 180 were observed in the livers of continuously exposed females. EC exposure affected xenobiotic and detoxification responses and the liver proteome in both sexes and included major plasma-secreted and blood proteins, and metabolic enzymes whose pathway analysis predicted dysregulation of cancer-related pathways and altered lipid dynamics. The latter were confirmed by a reduction in total lipids in female livers and up-regulation of cancer-related transcript markers in male livers respectively by sewage sludge exposure. CONCLUSIONS Our results demonstrate that chronic exposure to ECs causes major physiological changes in the liver, likely to affect multiple systems in the body and which may predispose individuals to increased disease risks.
Collapse
Affiliation(s)
- Panagiotis Filis
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | - Natasha Walker
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Linda Robertson
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Emily Eaton-Turner
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Lauma Ramona
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Michelle Bellingham
- Institute of Biodiversity, Animal Health & Comparative Medicine (IBAHCM), College of Medical, Veterinary & Life Sciences, University of Glasgow, Garscube Campus, Bearsden Rd, Glasgow G61 1QH, UK
| | - Maria R Amezaga
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Zulin Zhang
- The James Hutton Institute, Craigiebuckler, Aberdeen AB15 8QH, UK
| | | | - Neil P Evans
- Institute of Biodiversity, Animal Health & Comparative Medicine (IBAHCM), College of Medical, Veterinary & Life Sciences, University of Glasgow, Garscube Campus, Bearsden Rd, Glasgow G61 1QH, UK
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Corinne Cotinot
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy en Josas, France
| | - William D Rees
- The Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter O'Shaughnessy
- Institute of Biodiversity, Animal Health & Comparative Medicine (IBAHCM), College of Medical, Veterinary & Life Sciences, University of Glasgow, Garscube Campus, Bearsden Rd, Glasgow G61 1QH, UK
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
77
|
Li X, Wang Z, Klaunig JE. The effects of perfluorooctanoate on high fat diet induced non-alcoholic fatty liver disease in mice. Toxicology 2019; 416:1-14. [DOI: 10.1016/j.tox.2019.01.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/21/2019] [Accepted: 01/29/2019] [Indexed: 01/28/2023]
|
78
|
VoPham T. Environmental risk factors for liver cancer and nonalcoholic fatty liver disease. CURR EPIDEMIOL REP 2019; 6:50-66. [PMID: 31080703 DOI: 10.1007/s40471-019-0183-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Purpose of review The objective of this review was to summarize recent epidemiologic research examining the associations between environmental exposures and liver cancer and nonalcoholic fatty liver disease (NAFLD). Recent findings There were 28 liver cancer studies showing positive associations for exposures to aflatoxin, air pollution, polycyclic aromatic hydrocarbons, asbestos, chimney sweeping occupation, and paints; an inverse association for ultraviolet radiation; and null/inconsistent results for organic solvents, pesticides, perfluorooctanoic acid, nuclear radiation, iron foundry occupation, and brick kiln pollution. There were n=5 NAFLD studies showing positive associations for heavy metals, methyl tertiary-butyl ether, and selenium; and no association with trihalomethanes. Summary Evidence suggests that particular environmental exposures may be associated with liver cancer and NAFLD. Future liver cancer studies should examine specific histological subtypes and assess historical environmental exposures. Future NAFLD research should examine incident, biopsy-confirmed cases and the potential role of obesity and/or diabetes in studies of environmental factors and NAFLD.
Collapse
Affiliation(s)
- Trang VoPham
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
79
|
Abstract
Zebrafish (Danio rerio) larvae are a uniquely powerful model system which investigate the effects of toxicant exposure on liver development and function. Manufacturing processes and development of new synthetic compounds increased rapidly since the middle of the twentieth century, resulting in widespread exposure to environmental toxicants. This is compounded by the shift in the global burden of disease from infectious agents to chronic disease, particularly in industrialized nations, which increases the need to investigate the long-term and transgenerational effects of environmental exposures on human health. Zebrafish provide an excellent model to investigate the mechanisms of action of environmental pollutants given their large-scale embryo production and rapid development, which allow for short-term assessment of toxicity in a whole animal system. Here we describe methods for the use of zebrafish to study hepatotoxicity and liver disease induced by chemical toxicants. Many of the genetic, molecular, and cellular processes are conserved between zebrafish and mammals, enabling translation to human populations and diseases.
Collapse
Affiliation(s)
- Kathryn Bambino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua Morrison
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaime Chu
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
80
|
Papalou O, Kandaraki EA, Papadakis G, Diamanti-Kandarakis E. Endocrine Disrupting Chemicals: An Occult Mediator of Metabolic Disease. Front Endocrinol (Lausanne) 2019; 10:112. [PMID: 30881345 PMCID: PMC6406073 DOI: 10.3389/fendo.2019.00112] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs), a heterogeneous group of exogenous chemicals that can interfere with any aspect of endogenous hormones, represent an emerging global threat for human metabolism. There is now considerable evidence that the observed upsurge of metabolic disease cannot be fully attributed to increased caloric intake, physical inactivity, sleep deficit, and ageing. Among environmental factors implicated in the global deterioration of metabolic health, EDCs have drawn the biggest attention of scientific community, and not unjustifiably. EDCs unleash a coordinated attack toward multiple components of human metabolism, including crucial, metabolically-active organs such as hypothalamus, adipose tissue, pancreatic beta cells, skeletal muscle, and liver. Specifically, EDCs' impact during critical developmental windows can promote the disruption of individual or multiple systems involved in metabolism, via inducing epigenetic changes that can permanently alter the epigenome in the germline, enabling changes to be transmitted to the subsequent generations. The clear effect of this multifaceted attack is the manifestation of metabolic disease, clinically expressed as obesity, metabolic syndrome, diabetes mellitus, and non-alcoholic fatty liver disease. Although limitations of EDCs research do exist, there is no doubt that EDCs constitute a crucial parameter of the global deterioration of metabolic health we currently encounter.
Collapse
Affiliation(s)
- Olga Papalou
- Department of Endocrinology & Diabetes, Hygeia Hospital, Athens, Greece
| | | | | | - Evanthia Diamanti-Kandarakis
- Department of Endocrinology & Diabetes, Hygeia Hospital, Athens, Greece
- *Correspondence: Evanthia Diamanti-Kandarakis
| |
Collapse
|
81
|
Akinmoladun AC, Oladejo CO, Josiah SS, Famusiwa CD, Ojo OB, Olaleye MT. Catechin, quercetin and taxifolin improve redox and biochemical imbalances in rotenone-induced hepatocellular dysfunction: Relevance for therapy in pesticide-induced liver toxicity? PATHOPHYSIOLOGY 2018; 25:365-371. [DOI: 10.1016/j.pathophys.2018.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/03/2018] [Accepted: 07/07/2018] [Indexed: 12/22/2022] Open
|
82
|
Nelms MD, Mellor CL, Enoch SJ, Judson RS, Patlewicz G, Richard AM, Madden JM, Cronin MTD, Edwards SW. A Mechanistic Framework for Integrating Chemical Structure and High-Throughput Screening Results to Improve Toxicity Predictions. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 8:1-12. [PMID: 36779220 PMCID: PMC9910356 DOI: 10.1016/j.comtox.2018.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Adverse Outcome Pathways (AOPs) establish a connection between a molecular initiating event (MIE) and an adverse outcome. Detailed understanding of the MIE provides the ideal data for determining chemical properties required to elicit the MIE. This study utilized high-throughput screening data from the ToxCast program, coupled with chemical structural information, to generate chemical clusters using three similarity methods pertaining to nine MIEs within an AOP network for hepatic steatosis. Three case studies demonstrate the utility of the mechanistic information held by the MIE for integrating biological and chemical data. Evaluation of the chemical clusters activating the glucocorticoid receptor identified activity differences in chemicals within a cluster. Comparison of the estrogen receptor results with previous work showed that bioactivity data and structural alerts can be combined to improve predictions in a customizable way where bioactivity data are limited. The aryl hydrocarbon receptor (AHR) highlighted that while structural data can be used to offset limited data for new screening efforts, not all ToxCast targets have sufficient data to define robust chemical clusters. In this context, an alternative to additional receptor assays is proposed where assays for proximal key events downstream of AHR activation could be used to enhance confidence in active calls. These case studies illustrate how the AOP framework can support an iterative process whereby in vitro toxicity testing and chemical structure can be combined to improve toxicity predictions. In vitro assays can inform the development of structural alerts linking chemical structure to toxicity. Consequently, structurally related chemical groups can facilitate identification of assays that would be informative for a specific MIE. Together, these activities form a virtuous cycle where the mechanistic basis for the in vitro results and the breadth of the structural alerts continually improve over time to better predict activity of chemicals for which limited toxicity data exist.
Collapse
Affiliation(s)
- Mark D. Nelms
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA,Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory (NHEERL), Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27709, USA
| | - Claire L. Mellor
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Steven J. Enoch
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Richard S. Judson
- National Center for Computational Toxicology (NCCT), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27709, USA
| | - Grace Patlewicz
- National Center for Computational Toxicology (NCCT), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27709, USA
| | - Ann M. Richard
- National Center for Computational Toxicology (NCCT), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27709, USA
| | - Judith M. Madden
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Mark T. D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Stephen W. Edwards
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory (NHEERL), Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
83
|
Huck I, Beggs K, Apte U. Paradoxical Protective Effect of Perfluorooctanesulfonic Acid Against High-Fat Diet-Induced Hepatic Steatosis in Mice. Int J Toxicol 2018; 37:383-392. [PMID: 30134762 PMCID: PMC6150807 DOI: 10.1177/1091581818790934] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Perfluorooctanesulfonic acid (PFOS) is a persistent organic pollutant with worldwide bioaccumulation due to a very long half-life. Perfluorooctanesulfonic acid exposure results in significant hepatic effects including steatosis, proliferation, hepatomegaly, and in rodents, carcinogenesis. The objective of this study was to determine whether PFOS exposure exacerbates nonalcoholic fatty liver disease and nonalcoholic steatohepatitis pathogenesis. Eight-week-old male C57BL/6 J mice (n = 5 per group) were fed ad libitum normal chow diet (ND) alone, 60% high-fat diet (HFD) alone, ND + PFOS, and HFD + PFOS (0.0001% w/w (1 mg/kg) of PFOS) for 6 weeks. Both HFD alone and the ND + PFOS treatment induced significant adiposity and hepatomegaly, but the HFD + PFOS treatment showed a marked protection. Oil Red O staining and quantitative analysis of hepatic lipid content revealed increased hepatic steatosis in ND + PFOS and in HFD alone fed mice, which was prevented in HFD + PFOS treatment. Further studies revealed that ND + PFOS treatment significantly affected expression of lipid trafficking genes to favor steatosis, but these changes were absent in HFD + PFOS group. Specifically, expression of CD36, the major lipid importer in the cells, and peroxisome proliferator-activated receptor gamma (PPARγ), its major regulator, were induced in HFD + no treatment (NT) and ND + PFOS-fed mice but remained unchanged in HFD + PFOS mice. In conclusion, these data indicate that coadministration of PFOS with HFD mitigates steatosis and hepatomegaly induced by HFD and that by PFOS fed in ND diet via regulation of cellular lipid import machinery. These findings suggest dietary lipid content be considered when performing risk management of PFOS in humans and the elucidation of PFOS-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ian Huck
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kevin Beggs
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Udayan Apte
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
84
|
Mesnage R, Biserni M, Wozniak E, Xenakis T, Mein CA, Antoniou MN. Comparison of transcriptome responses to glyphosate, isoxaflutole, quizalofop-p-ethyl and mesotrione in the HepaRG cell line. Toxicol Rep 2018; 5:819-826. [PMID: 30128299 PMCID: PMC6098220 DOI: 10.1016/j.toxrep.2018.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/09/2018] [Indexed: 01/27/2023] Open
Abstract
Use and thus exposure to quizalofop-p-ethyl, isoxaflutole, mesotrione and glyphosate, which are declared as active principles in commercial formulations of herbicides, is predicted to rapidly increase in coming years in an effort to overcome the wide-spread appearance of glyphosate-resistant weeds, especially in fields where glyphosate-tolerant genetically modified crops are cultivated in the USA. Thus, there is an urgent need for an evaluation of metabolic effects of new pesticide ingredients used to replace glyphosate. As the liver is a primary target of chemical pollutant toxicity, we have used the HepaRG human liver cell line as a model system to assess the toxicological insult from quizalofop-p-ethyl, isoxaflutole, mesotrione and glyphosate by determining alterations in the transcriptome caused by exposure to three concentrations of each of these compounds, including a low environmentally relevant dose. RNA-seq data were analysed with HISAT2, StringTie and Ballgown. Quizalofop-p-ethyl was found to be the most toxic of the pesticide ingredients tested, causing alterations in gene expression that are associated with pathways involved in fatty acid degradation and response to alcoholism. Isoxaflutole was less toxic, but caused detectable changes in retinol metabolism and in the PPAR signalling pathway at a concentration of 1 mM. ToxCast data analysis revealed that isoxaflutole activated PPAR gamma receptor and pregnane X responsive elements in reporter gene assays. Glyphosate and mesotrione caused subtle changes in transcriptome profiles, with too few genes altered in their function to allow a reliable pathway analysis. In order to explore the effects of glyphosate in greater depth and detail, we undertook a global metabolome profiling. This revealed a decrease in free long chain fatty acids and polyunsaturated fatty acid levels at the lowest concentration (0.06 μM) of glyphosate, although no effects were detected at the two higher concentrations tested, perhaps suggesting a non-linear dose response. This surprising result will need to be confirmed by additional studies. Overall, our findings contribute to filling the knowledge gap regarding metabolic toxicity that can potentially arise from exposure to these four herbicide active principles.
Collapse
Affiliation(s)
- Robin Mesnage
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, Guy's Hospital, 8th Floor, Tower Wing, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Martina Biserni
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, Guy's Hospital, 8th Floor, Tower Wing, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Eva Wozniak
- Genome Centre, Barts and the London School of Medicine and Dentistry, John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Theodoros Xenakis
- Genome Centre, Barts and the London School of Medicine and Dentistry, John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Charles A. Mein
- Genome Centre, Barts and the London School of Medicine and Dentistry, John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Michael N. Antoniou
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, Guy's Hospital, 8th Floor, Tower Wing, Great Maze Pond, London SE1 9RT, United Kingdom
| |
Collapse
|
85
|
Severe Alcoholic Hepatitis in a Teetotaler. Am J Gastroenterol 2018; 113:1260-1261. [PMID: 29887602 DOI: 10.1038/s41395-018-0154-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 05/11/2018] [Indexed: 12/11/2022]
|
86
|
Luckert C, Braeuning A, de Sousa G, Durinck S, Katsanou ES, Konstantinidou P, Machera K, Milani ES, Peijnenburg AACM, Rahmani R, Rajkovic A, Rijkers D, Spyropoulou A, Stamou M, Stoopen G, Sturla S, Wollscheid B, Zucchini-Pascal N, Lampen A. Adverse Outcome Pathway-Driven Analysis of Liver Steatosis in Vitro: A Case Study with Cyproconazole. Chem Res Toxicol 2018; 31:784-798. [DOI: 10.1021/acs.chemrestox.8b00112] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Claudia Luckert
- Department Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| | - Georges de Sousa
- INRA Unit 1331, TOXALIM, French National Institute for Agricultural Research, 180 chemin de Tournefeuille - BP.93173 F-31027 TOULOUSE cedex 3, France
| | - Sigrid Durinck
- Faculty of Bioscience Engineering, Department of Food Technology, Food Safety and Health, Ghent University, 9000 Ghent, Belgium
| | | | | | | | | | | | - Roger Rahmani
- INRA Unit 1331, TOXALIM, French National Institute for Agricultural Research, 180 chemin de Tournefeuille - BP.93173 F-31027 TOULOUSE cedex 3, France
| | - Andreja Rajkovic
- Faculty of Bioscience Engineering, Department of Food Technology, Food Safety and Health, Ghent University, 9000 Ghent, Belgium
| | - Deborah Rijkers
- RIKILT Wageningen University and Research, 6708 WB Wageningen, The Netherlands
| | | | | | - Geert Stoopen
- RIKILT Wageningen University and Research, 6708 WB Wageningen, The Netherlands
| | | | | | - Nathalie Zucchini-Pascal
- INRA Unit 1331, TOXALIM, French National Institute for Agricultural Research, 180 chemin de Tournefeuille - BP.93173 F-31027 TOULOUSE cedex 3, France
| | - Alfonso Lampen
- Department Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| |
Collapse
|
87
|
Clair HB, Pinkston CM, Rai SN, Pavuk M, Dutton ND, Brock GN, Prough RA, Falkner KC, McClain CJ, Cave MC. Liver Disease in a Residential Cohort With Elevated Polychlorinated Biphenyl Exposures. Toxicol Sci 2018; 164:39-49. [PMID: 29684222 PMCID: PMC6016643 DOI: 10.1093/toxsci/kfy076] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endocrine and metabolism disrupting chemicals (EDCs/MDCs) have been associated with environmental liver diseases including toxicant-associated steatohepatitis (TASH). TASH has previously been characterized by hepatocellular necrosis, disrupted intermediary metabolism, and liver inflammation. Polychlorinated biphenyls (PCBs) are environmental EDCs/MDCs associated with the genesis and progression of steatohepatitis in animal models and human liver injury in epidemiology studies. The cross-sectional Anniston Community Health Survey (ACHS) investigates ortho-substituted PCB exposures and health effects near a former PCB manufacturing complex. The rates of obesity, diabetes, and dyslipidemia were previously determined to be high in ACHS. In this study, 738 ACHS participants were categorized by liver disease status using the serum cytokeratin 18 biomarker. Associations between PCB exposures and mechanistic biomarkers of intermediary metabolism, inflammation, and hepatocyte death were determined. The liver disease prevalence was high (60.2%), and 80.7% of these individuals were categorized as having TASH. Sex and race/ethnicity differences were noted. TASH was associated with increased exposures to specific PCB congeners, insulin resistance, dyslipidemia, proinflammatory cytokines, and liver necrosis. These findings are consistent with PCB-related steatohepatitis. ΣPCBs was inversely associated with insulin resistance/production, leptin, and hepatocyte apoptosis, while other adipocytokines were increased. This is possibly the largest environmental liver disease study applying mechanistic biomarkers ever performed and the most comprehensive analysis of PCBs and adipocytokines. It provides insight into the mechanisms of PCB-related endocrine and metabolic disruption in liver disease and diabetes. In the future, associations between additional exposures and liver disease biomarkers will be evaluated in the ACHS and follow-up ACHS-II studies.
Collapse
Affiliation(s)
- Heather B Clair
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Christina M Pinkston
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences
- Biostatistics Shared Facility, James Graham Brown Cancer Center
- Hepatobiology and Toxicology COBRE Program, University of Louisville, Louisville, Kentucky 40202
| | - Shesh N Rai
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences
- Biostatistics Shared Facility, James Graham Brown Cancer Center
- Hepatobiology and Toxicology COBRE Program, University of Louisville, Louisville, Kentucky 40202
| | - Marian Pavuk
- Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, Georgia 30341
| | - Nina D Dutton
- Oak Ridge Institute for Science and Education (ORISE) Research Participant, ATSDR, Atlanta, Georgia 30341
| | - Guy N Brock
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Keith Cameron Falkner
- Hepatobiology and Toxicology COBRE Program, University of Louisville, Louisville, Kentucky 40202
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky 40202
- University of Louisville Alcohol Research Center, Louisville, Kentucky
| | - Craig J McClain
- Hepatobiology and Toxicology COBRE Program, University of Louisville, Louisville, Kentucky 40202
- Oak Ridge Institute for Science and Education (ORISE) Research Participant, ATSDR, Atlanta, Georgia 30341
- University of Louisville Alcohol Research Center, Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky 40202
- The Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky 40206
- The KentuckyOne Health Jewish Hospital Liver Transplant Program, Louisville, Kentucky 40202
| | - Matthew C Cave
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202
- Hepatobiology and Toxicology COBRE Program, University of Louisville, Louisville, Kentucky 40202
- Oak Ridge Institute for Science and Education (ORISE) Research Participant, ATSDR, Atlanta, Georgia 30341
- University of Louisville Alcohol Research Center, Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky 40202
- The Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky 40206
- The KentuckyOne Health Jewish Hospital Liver Transplant Program, Louisville, Kentucky 40202
| |
Collapse
|
88
|
Klaunig JE, Li X, Wang Z. Role of xenobiotics in the induction and progression of fatty liver disease. Toxicol Res (Camb) 2018; 7:664-680. [PMID: 30090613 PMCID: PMC6062016 DOI: 10.1039/c7tx00326a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease is a major cause of chronic liver pathology in humans. Fatty liver disease involves the accumulation of hepatocellular fat in hepatocytes that can progress to hepatitis. Steatohepatitis is categorized into alcoholic (ASH) or non-alcoholic (NASH) steatohepatitis based on the etiology of the insult. Both pathologies involve an initial steatosis followed by a progressive inflammation of the liver and eventual hepatic fibrosis (steatohepatitis) and cirrhosis. The involvement of pharmaceuticals and other chemicals in the initiation and progression of fatty liver disease has received increased study. This review will examine not only how xenobiotics initiate hepatic steatosis and steatohepatitis but also how the presence of fatty liver may modify the metabolism and pathologic effects of xenobiotics. The feeding of a high fat diet results in changes in the expression of nuclear receptors that are involved in adaptive and adverse liver effects following xenobiotic exposure. High fat diets also modulate cellular and molecular pathways involved in inflammation, metabolism, oxidative phosphorylation and cell growth. Understanding the role of hepatic steatosis and steatohepatitis on the sequelae of toxic and pathologic changes seen following xenobiotic exposure has importance in defining proper and meaningful human risk characterization of the drugs and other chemical agents.
Collapse
Affiliation(s)
- James E Klaunig
- Indiana University , School of Public Health , Bloomington , Indiana , USA .
| | - Xilin Li
- Indiana University , School of Public Health , Bloomington , Indiana , USA .
| | - Zemin Wang
- Indiana University , School of Public Health , Bloomington , Indiana , USA .
| |
Collapse
|
89
|
Bonvallot N, Canlet C, Blas-Y-Estrada F, Gautier R, Tremblay-Franco M, Chevolleau S, Cordier S, Cravedi JP. Metabolome disruption of pregnant rats and their offspring resulting from repeated exposure to a pesticide mixture representative of environmental contamination in Brittany. PLoS One 2018; 13:e0198448. [PMID: 29924815 PMCID: PMC6010212 DOI: 10.1371/journal.pone.0198448] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 05/18/2018] [Indexed: 11/19/2022] Open
Abstract
The use of pesticides exposes humans to numerous harmful molecules. Exposure in early-life may be responsible for adverse effects in later life. This study aimed to assess the metabolic modifications induced in pregnant rats and their offspring by a pesticide mixture representative of human exposure. Ten pregnant rats were exposed to a mixture of eight pesticides: acetochlor (246 μg/kg bw/d) + bromoxynil (12 μg/kg bw/d) + carbofuran (22.5 μg/kg bw/d) + chlormequat (35 μg/kg bw/d) + ethephon (22.5 μg/kg bw/d) + fenpropimorph (15.5 μg/kg bw/d) + glyphosate (12 μg/kg bw/d) + imidacloprid (12.5 μg/kg bw/d) representing the main environmental pesticide exposure in Brittany (France) in 2004. Another group of 10 pregnant rats served as controls. Females were fed ad libitum from early pregnancy, which is from gestational day (GD) 4 to GD 21. Urine samples were collected at GD 15. At the end of the exposure, mothers and pups were euthanized and blood, liver, and brain samples collected. 1H NMR-based metabolomics and GC-FID analyses were performed and PCA and PLS-DA used to discriminate between control and exposed groups. Metabolites for which the levels were significantly modified were then identified using the Kruskal-Wallis test, and p-values were adjusted for multiple testing correction using the False Discovery Rate. The metabolomics analysis revealed many differences between dams of the two groups, especially in the plasma, liver and brain. The modified metabolites are involved in TCA cycle, energy production and storage, lipid and carbohydrate metabolism, and amino-acid metabolism. These modifications suggest that the pesticide mixture may induce oxidative stress associated with mitochondrial dysfunction and the impairment of glucose and lipid metabolism. These observations may reflect liver dysfunction with increased relative liver weight and total lipid content. Similar findings were observed for glucose and energy metabolism in the liver of the offspring, and oxidative stress was also suggested in the brains of male offspring.
Collapse
Affiliation(s)
- Nathalie Bonvallot
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, Rennes, France
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| | - Cécile Canlet
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| | - Florence Blas-Y-Estrada
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| | - Roselyne Gautier
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| | - Marie Tremblay-Franco
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| | - Sylvie Chevolleau
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| | - Sylvaine Cordier
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, Rennes, France
| | - Jean-Pierre Cravedi
- INRA UMR 1331 Toxalim, University of Toulouse, INP, ENVT, EIP, UPS, UMR1331, Toulouse, France
| |
Collapse
|
90
|
Integrated transcriptomics and metabolomics reveal signatures of lipid metabolism dysregulation in HepaRG liver cells exposed to PCB 126. Arch Toxicol 2018; 92:2533-2547. [PMID: 29947894 PMCID: PMC6063328 DOI: 10.1007/s00204-018-2235-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022]
Abstract
Chemical pollutant exposure is a risk factor contributing to the growing epidemic of non-alcoholic fatty liver disease (NAFLD) affecting human populations that consume a western diet. Although it is recognized that intoxication by chemical pollutants can lead to NAFLD, there is limited information available regarding the mechanism by which typical environmental levels of exposure can contribute to the onset of this disease. Here, we describe the alterations in gene expression profiles and metabolite levels in the human HepaRG liver cell line, a validated model for cellular steatosis, exposed to the polychlorinated biphenyl (PCB) 126, one of the most potent chemical pollutants that can induce NAFLD. Sparse partial least squares classification of the molecular profiles revealed that exposure to PCB 126 provoked a decrease in polyunsaturated fatty acids as well as an increase in sphingolipid levels, concomitant with a decrease in the activity of genes involved in lipid metabolism. This was associated with an increased oxidative stress reflected by marked disturbances in taurine metabolism. A gene ontology analysis showed hallmarks of an activation of the AhR receptor by dioxin-like compounds. These changes in metabolome and transcriptome profiles were observed even at the lowest concentration (100 pM) of PCB 126 tested. A decrease in docosatrienoate levels was the most sensitive biomarker. Overall, our integrated multi-omics analysis provides mechanistic insight into how this class of chemical pollutant can cause NAFLD. Our study lays the foundation for the development of molecular signatures of toxic effects of chemicals causing fatty liver diseases to move away from a chemical risk assessment based on in vivo animal experiments.
Collapse
|
91
|
Lukowicz C, Ellero-Simatos S, Régnier M, Polizzi A, Lasserre F, Montagner A, Lippi Y, Jamin EL, Martin JF, Naylies C, Canlet C, Debrauwer L, Bertrand-Michel J, Al Saati T, Théodorou V, Loiseau N, Mselli-Lakhal L, Guillou H, Gamet-Payrastre L. Metabolic Effects of a Chronic Dietary Exposure to a Low-Dose Pesticide Cocktail in Mice: Sexual Dimorphism and Role of the Constitutive Androstane Receptor. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:067007. [PMID: 29950287 PMCID: PMC6084886 DOI: 10.1289/ehp2877] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 04/27/2018] [Accepted: 04/28/2018] [Indexed: 05/03/2023]
Abstract
BACKGROUND Epidemiological evidence suggests a link between pesticide exposure and the development of metabolic diseases. However, most experimental studies have evaluated the metabolic effects of pesticides using individual molecules, often at nonrelevant doses or in combination with other risk factors such as high-fat diets. OBJECTIVES We aimed to evaluate, in mice, the metabolic consequences of chronic dietary exposure to a pesticide mixture at nontoxic doses, relevant to consumers' risk assessment. METHODS A mixture of six pesticides commonly used in France, i.e., boscalid, captan, chlorpyrifos, thiofanate, thiacloprid, and ziram, was incorporated in a standard chow at doses exposing mice to the tolerable daily intake (TDI) of each pesticide. Wild-type (WT) and constitutive androstane receptor-deficient (CAR-/-) male and female mice were exposed for 52 wk. We assessed metabolic parameters [body weight (BW), food and water consumption, glucose tolerance, urinary metabolome] throughout the experiment. At the end of the experiment, we evaluated liver metabolism (histology, transcriptomics, metabolomics, lipidomics) and pesticide detoxification using liquid chromatography-mass spectrometry (LC-MS). RESULTS Compared to those fed control chow, WT male mice fed pesticide chow had greater BW gain and more adiposity. Moreover, these WT males fed pesticide chow exhibited characteristics of hepatic steatosis and glucose intolerance, which were not observed in those fed control chow. WT exposed female mice exhibited fasting hyperglycemia, higher reduced glutathione (GSH):oxidized glutathione (GSSG) liver ratio and perturbations of gut microbiota-related urinary metabolites compared to WT mice fed control chow. When we performed these experiments on CAR-/- mice, pesticide-exposed CAR-/- males did not exhibit BW gain or changes in glucose metabolism compared to the CAR-/- males fed control chow. Moreover, CAR-/- females fed pesticide chow exhibited pesticide toxicity with higher BWs and mortality rate compared to the CAR-/- females fed control chow. CONCLUSIONS To our knowledge, we are the first to demonstrate a sexually dimorphic obesogenic and diabetogenic effect of chronic dietary exposure to a common mixture of pesticides at TDI levels, and to provide evidence for a partial role for CAR in an in vivo mouse model. This raises questions about the relevance of TDI for individual pesticides when present in a mixture. https://doi.org/10.1289/EHP2877.
Collapse
Affiliation(s)
- Céline Lukowicz
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Marion Régnier
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Arnaud Polizzi
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Frédéric Lasserre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Alexandra Montagner
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Yannick Lippi
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Emilien L Jamin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
- Axiom Platform, MetaToul-MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Jean-François Martin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Claire Naylies
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Cécile Canlet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
- Axiom Platform, MetaToul-MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Laurent Debrauwer
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
- Axiom Platform, MetaToul-MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Justine Bertrand-Michel
- Plateforme Lipidomique Inserm/UPS UMR 1048 - I2MC Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Talal Al Saati
- Service d’histopathologie Expérimentale Unité Inserm/UPS/ENVT -US006/CREFRE Inserm, Bât. F, CHU Purpan, Toulouse, France
| | - Vassilia Théodorou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Nicolas Loiseau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Laïla Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
92
|
Bambino K, Zhang C, Austin C, Amarasiriwardena C, Arora M, Chu J, Sadler KC. Inorganic arsenic causes fatty liver and interacts with ethanol to cause alcoholic liver disease in zebrafish. Dis Model Mech 2018; 11:dmm.031575. [PMID: 29361514 PMCID: PMC5894941 DOI: 10.1242/dmm.031575] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
The rapid increase in fatty liver disease (FLD) incidence is attributed largely to genetic and lifestyle factors; however, environmental toxicants are a frequently overlooked factor that can modify the effects of more common causes of FLD. Chronic exposure to inorganic arsenic (iAs) is associated with liver disease in humans and animal models, but neither the mechanism of action nor the combinatorial interaction with other disease-causing factors has been fully investigated. Here, we examined the contribution of iAs to FLD using zebrafish and tested the interaction with ethanol to cause alcoholic liver disease (ALD). We report that zebrafish exposed to iAs throughout development developed specific phenotypes beginning at 4 days post-fertilization (dpf), including the development of FLD in over 50% of larvae by 5 dpf. Comparative transcriptomic analysis of livers from larvae exposed to either iAs or ethanol revealed the oxidative stress response and the unfolded protein response (UPR) caused by endoplasmic reticulum (ER) stress as common pathways in both these models of FLD, suggesting that they target similar cellular processes. This was confirmed by our finding that arsenic is synthetically lethal with both ethanol and a well-characterized ER-stress-inducing agent (tunicamycin), suggesting that these exposures work together through UPR activation to cause iAs toxicity. Most significantly, combined exposure to sub-toxic concentrations of iAs and ethanol potentiated the expression of UPR-associated genes, cooperated to induce FLD, reduced the expression of as3mt, which encodes an arsenic-metabolizing enzyme, and significantly increased the concentration of iAs in the liver. This demonstrates that iAs exposure is sufficient to cause FLD and that low doses of iAs can potentiate the effects of ethanol to cause liver disease. This article has an associated First Person interview with the first author of the paper. Summary: Using zebrafish, the authors show that exposure to a common environmental contaminant, inorganic arsenic, increases the risk of alcoholic liver disease.
Collapse
Affiliation(s)
- Kathryn Bambino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chi Zhang
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| | - Christine Austin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jaime Chu
- Department of Pediatrics, Division of Pediatric Hepatology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| |
Collapse
|
93
|
Treviño LS, Katz TA. Endocrine Disruptors and Developmental Origins of Nonalcoholic Fatty Liver Disease. Endocrinology 2018; 159:20-31. [PMID: 29126168 PMCID: PMC5761605 DOI: 10.1210/en.2017-00887] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing epidemic worldwide, particularly in countries that consume a Western diet, and can lead to life-threatening conditions such as cirrhosis and hepatocellular carcinoma. With increasing prevalence of NAFLD in both children and adults, an understanding of the factors that promote NAFLD development and progression is crucial. Environmental agents, including endocrine-disrupting chemicals (EDCs), which have been linked to other diseases, may play a role in NAFLD development. Increasing evidence supports a developmental origin of liver disease, and early-life exposure to EDCs could represent one risk factor for the development of NAFLD later in life. Rodent studies provide the strongest evidence for this link, but further studies are needed to define whether there is a causal link between early-life EDC exposure and NAFLD development in humans. Elucidating the molecular mechanisms underlying development of NAFLD in the context of developmental EDC exposures may identify biomarkers for people at risk, as well as potential intervention and/or therapeutic opportunities for the disease.
Collapse
Affiliation(s)
- Lindsey S. Treviño
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Tiffany A. Katz
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
94
|
Valvi D, Oulhote Y, Weihe P, Dalgård C, Bjerve KS, Steuerwald U, Grandjean P. Gestational diabetes and offspring birth size at elevated environmental pollutant exposures. ENVIRONMENT INTERNATIONAL 2017; 107:205-215. [PMID: 28753482 PMCID: PMC5584560 DOI: 10.1016/j.envint.2017.07.016] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 05/17/2023]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is associated with increased availability of glucose and macronutrients in fetal circulation and macrosomia. Therefore, the role of GDM in the association between metabolism-disrupting chemicals and birth size deserves attention. OBJECTIVE We examined whether GDM may mediate or modify the associations between maternal environmental pollutant exposures and offspring birth size measures. METHODS We analyzed 604 Faroese pregnant women and their offsprings born in 1997-2000. Maternal pregnancy serum concentrations of organochlorine compounds (OCs: polychlorinated biphenyl (PCB) congeners and dichlorodiphenyldichloroethylene (DDE)), and five perfluoroalkyl substances (PFASs), and hair and cord blood mercury concentrations were measured. We used regression (single-pollutants) and structural equation models (SEMs) (multiple-pollutant analyses using latent constructs of OCs, PFASs and mercury) to estimate the associations with GDM and birth size measures, accounting for mediation and/or effect modification by GDM. RESULTS Serum-DDE and hair-mercury concentrations were associated with GDM (adjusted OR per concentration doubling: 1.29; 95% CI: 0.94, 1.77 for DDE, and 0.79; 95% CI: 0.62, 0.99 for mercury), but in multiple pollutant-adjusted SEMs only a positive association between OC exposure and GDM remained significant (change in GDM odds per OC doubling: 0.45; 95% CI: 0.05, 0.86). PCB and overall OC exposure were positively associated with head circumference (SEM; mean change per OC doubling: 0.13cm; 95% CI, 0.01. 0.25). Overall PFAS exposure was inversely associated with birth weight (SEM; mean change per PFAS doubling: -169g; 95% CI: -359, 21), and for many single-PFASs we found a pattern of inverse associations with birth weight and head circumference in boys, and positive or null associations in girls. None of the environmental pollutants was associated with offspring length. GDM neither modified nor mediated the associations with birth size measures. CONCLUSIONS We found associations with GDM and offspring birth size to be specific to the environmental pollutant or pollutant group. Associations with birth size measures appear to be independent of GDM occurrence.
Collapse
Affiliation(s)
- Damaskini Valvi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| | - Youssef Oulhote
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Pal Weihe
- Department of Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark; Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
| | - Christine Dalgård
- Department of Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Kristian S Bjerve
- Department of Medical Biochemistry, St. Olays Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Laboratory Medicine, Children's and Women's Health, NTNU, Trondheim, Norway
| | - Ulrike Steuerwald
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
| | - Philippe Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
95
|
Hepatoprotective Effect of Microemulsion-Based System of Prunus Cerasus Kernel Extract on CCL4-induced Liver Damage in Mice. Jundishapur J Nat Pharm Prod 2017. [DOI: 10.5812/jjnpp.14282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
96
|
Shimpi PC, More VR, Paranjpe M, Donepudi AC, Goodrich JM, Dolinoy DC, Rubin B, Slitt AL. Hepatic Lipid Accumulation and Nrf2 Expression following Perinatal and Peripubertal Exposure to Bisphenol A in a Mouse Model of Nonalcoholic Liver Disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:087005. [PMID: 28796629 PMCID: PMC5783659 DOI: 10.1289/ehp664] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 05/03/2023]
Abstract
BACKGROUND Exposure to chemicals during critical windows of development may re-program liver for increased risk of nonalcoholic fatty liver disease (NAFLD). Bisphenol A (BPA), a plastics component, has been described to impart adverse effects during gestational and lactational exposure. Our work has pointed to nuclear factor E2-related factor 2 (Nrf2) being a modulator of hepatic lipid accumulation in models of NAFLD. OBJECTIVES To determine if chemical exposure can prime liver for steatosis via modulation of NRF2 and epigenetic mechanisms. METHODS Utilizing BPA as a model exposure, pregnant CD-1 mice were administered 25μg/kg/day BPA via osmotic minipumps from gestational day 8 through postnatal day (PND)16. The offspring were weaned on PND21 and exposed to same dose of BPA via their drinking water through PND35. Tissues were collected from pups at week 5 (W5), and their littermates at week 39 (W39). RESULTS BPA increased hepatic lipid content concomitant with increased Nrf2 and pro-lipogenic enzyme expression at W5 and W39 in female offspring. BPA exposure increased Nrf2 binding to a putative antioxidant response element consensus sequence in the sterol regulatory-element binding protein-1c (Srebp-1c) promoter. Known Nrf2 activators increased SREBP-1C promoter reporter activity in HepG2 cells. Methylated DNA immunoprecipitation-PCR and pyrosequencing revealed that developmental BPA exposure induced hypomethylation of the Nrf2 and Srebp-1c promoters in livers of W5 mice, which was more prominent in W39 mice than in others. CONCLUSION Exposure to a xenobiotic during early development induced persistent fat accumulation via hypomethylation of lipogenic genes. Moreover, increased Nrf2 recruitment to the Srebp-1c promoter in livers of BPA-exposed mice was observed. Overall, the underlying mechanisms described a broader impact beyond BPA exposure and can be applied to understand other models of NAFLD. https://doi.org/10.1289/EHP664.
Collapse
Affiliation(s)
- Prajakta C Shimpi
- Biomedical and Pharmaceutical Sciences, University of Rhode Island , Kingston, Rhode Island, USA
| | - Vijay R More
- Biomedical and Pharmaceutical Sciences, University of Rhode Island , Kingston, Rhode Island, USA
| | - Maneesha Paranjpe
- Sackler School of Graduate Biomedical Sciences, Tufts University , Boston, Massachusetts, USA
| | - Ajay C Donepudi
- Biomedical and Pharmaceutical Sciences, University of Rhode Island , Kingston, Rhode Island, USA
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health , Ann Arbor, Michigan, USA
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health , Ann Arbor, Michigan, USA
| | - Beverly Rubin
- Sackler School of Graduate Biomedical Sciences, Tufts University , Boston, Massachusetts, USA
| | - Angela L Slitt
- Biomedical and Pharmaceutical Sciences, University of Rhode Island , Kingston, Rhode Island, USA
| |
Collapse
|
97
|
Abstract
A growing epidemic of nonalcoholic fatty liver disease (NAFLD) is paralleling the increase in the incidence of obesity and diabetes mellitus in countries that consume a Western diet. As NAFLD can lead to life-threatening conditions such as cirrhosis and hepatocellular carcinoma, an understanding of the factors that trigger its development and pathological progression is needed. Although by definition this disease is not associated with alcohol consumption, exposure to environmental agents that have been linked to other diseases might have a role in the development of NAFLD. Here, we focus on one class of these agents, endocrine-disrupting chemicals (EDCs), and their potential to influence the initiation and progression of a cascade of pathological conditions associated with hepatic steatosis (fatty liver). Experimental studies have revealed several potential mechanisms by which EDC exposure might contribute to disease pathogenesis, including the modulation of nuclear hormone receptor function and the alteration of the epigenome. However, many questions remain to be addressed about the causal link between acute and chronic EDC exposure and the development of NAFLD in humans. Future studies that address these questions hold promise not only for understanding the linkage between EDC exposure and liver disease but also for elucidating the molecular mechanisms that underpin NAFLD, which in turn could facilitate the development of new prevention and treatment opportunities.
Collapse
Affiliation(s)
- Charles E Foulds
- Department of Molecular and Cellular Biology, Baylor College of Medicine
- Center for Precision Environmental Health, Baylor College of Medicine
| | - Lindsey S Treviño
- Department of Molecular and Cellular Biology, Baylor College of Medicine
- Center for Precision Environmental Health, Baylor College of Medicine
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine
- Dan L. Duncan Cancer Center, Baylor College of Medicine
| | - Cheryl L Walker
- Department of Molecular and Cellular Biology, Baylor College of Medicine
- Center for Precision Environmental Health, Baylor College of Medicine
- Dan L. Duncan Cancer Center, Baylor College of Medicine
- Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030, USA
| |
Collapse
|
98
|
Hardesty JE, Wahlang B, Falkner KC, Clair HB, Clark BJ, Ceresa BP, Prough RA, Cave MC. Polychlorinated biphenyls disrupt hepatic epidermal growth factor receptor signaling. Xenobiotica 2017; 47:807-820. [PMID: 27458090 DOI: 10.1080/00498254.2016.1217572] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
1. Polychlorinated biphenyls (PCBs) are persistent environmental pollutants that disrupt hepatic xenobiotic and intermediary metabolism, leading to metabolic syndrome and nonalcoholic steatohepatitis (NASH). 2. Since phenobarbital indirectly activates Constitutive Androstane Receptor (CAR) by antagonizing growth factor binding to the epidermal growth factor receptor (EGFR), we hypothesized that PCBs may also diminish EGFR signaling. 3. The effects of the PCB mixture Aroclor 1260 on the protein phosphorylation cascade triggered by EGFR activation were determined in murine (in vitro and in vivo) and human models (in vitro). EGFR tyrosine residue phosphorylation was decreased by PCBs in all models tested. 4. The IC50 values for Aroclor 1260 concentrations that decreased Y1173 phosphorylation of EGFR were similar in murine AML-12 and human HepG2 cells (∼2-4 μg/mL). Both dioxin and non-dioxin-like PCB congeners decreased EGFR phosphorylation in cell culture. 5. PCB treatment reduced phosphorylation of downstream EGFR effectors including Akt and mTOR, as well as other phosphoprotein targets including STAT3 and c-RAF in vivo. 6. PCBs diminish EGFR signaling in human and murine hepatocyte models and may dysregulate critical phosphoprotein regulators of energy metabolism and nutrition, providing a new mechanism of action in environmental diseases.
Collapse
Affiliation(s)
- Josiah E Hardesty
- a Department of Biochemistry and Molecular Genetics , University of Louisville School of Medicine , Louisville , KY , USA
| | - Banrida Wahlang
- b University of Kentucky Superfund Research Center, University of Kentucky , Lexington , KY , USA
| | - K Cameron Falkner
- c Department of Medicine , Division of Gastroenterology, Hepatology and Nutrition , and
| | - Heather B Clair
- a Department of Biochemistry and Molecular Genetics , University of Louisville School of Medicine , Louisville , KY , USA
| | - Barbara J Clark
- a Department of Biochemistry and Molecular Genetics , University of Louisville School of Medicine , Louisville , KY , USA
| | - Brian P Ceresa
- d Department of Pharmacology and Toxicology , University of Louisville School of Medicine , Louisville , KY , USA
| | - Russell A Prough
- a Department of Biochemistry and Molecular Genetics , University of Louisville School of Medicine , Louisville , KY , USA
| | - Matthew C Cave
- a Department of Biochemistry and Molecular Genetics , University of Louisville School of Medicine , Louisville , KY , USA.,c Department of Medicine , Division of Gastroenterology, Hepatology and Nutrition , and.,d Department of Pharmacology and Toxicology , University of Louisville School of Medicine , Louisville , KY , USA.,e The Robley Rex Veterans Affairs Medical Center , Louisville , KY , USA , and.,f The Kentucky One Health Jewish Hospital Liver Transplant Program , Louisville , KY , USA
| |
Collapse
|
99
|
Heindel JJ, Blumberg B, Cave M, Machtinger R, Mantovani A, Mendez MA, Nadal A, Palanza P, Panzica G, Sargis R, Vandenberg LN, Vom Saal F. Metabolism disrupting chemicals and metabolic disorders. Reprod Toxicol 2017; 68:3-33. [PMID: 27760374 PMCID: PMC5365353 DOI: 10.1016/j.reprotox.2016.10.001] [Citation(s) in RCA: 646] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/04/2016] [Accepted: 10/13/2016] [Indexed: 01/09/2023]
Abstract
The recent epidemics of metabolic diseases, obesity, type 2 diabetes(T2D), liver lipid disorders and metabolic syndrome have largely been attributed to genetic background and changes in diet, exercise and aging. However, there is now considerable evidence that other environmental factors may contribute to the rapid increase in the incidence of these metabolic diseases. This review will examine changes to the incidence of obesity, T2D and non-alcoholic fatty liver disease (NAFLD), the contribution of genetics to these disorders and describe the role of the endocrine system in these metabolic disorders. It will then specifically focus on the role of endocrine disrupting chemicals (EDCs) in the etiology of obesity, T2D and NAFLD while finally integrating the information on EDCs on multiple metabolic disorders that could lead to metabolic syndrome. We will specifically examine evidence linking EDC exposures during critical periods of development with metabolic diseases that manifest later in life and across generations.
Collapse
Affiliation(s)
- Jerrold J Heindel
- National Institute of Environmental Health Sciences, Division of Extramural Research and Training Research Triangle Park, NC, USA.
| | - Bruce Blumberg
- University of California, Department of Developmental and Cell Biology, Irvine CA, USA
| | - Mathew Cave
- University of Louisville, Division of Gastroenterology, Hepatology and Nutrition, Louisville KY, USA
| | | | | | - Michelle A Mendez
- University of North Carolina at Chapel Hill, School of Public Health, Chapel Hill NC, USA
| | - Angel Nadal
- Institute of Bioengineering and CIBERDEM, Miguel Hernandez University of Elche, Elche, Alicante, Spain
| | - Paola Palanza
- University of Parma, Department of Neurosciences, Parma, Italy
| | - Giancarlo Panzica
- University of Turin, Department of Neuroscience and Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy
| | - Robert Sargis
- University of Chicago, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine Chicago, IL, USA
| | - Laura N Vandenberg
- University of Massachusetts, Department of Environmental Health Sciences, School of Public Health & Health Sciences, Amherst, MA, USA
| | - Frederick Vom Saal
- University of Missouri, Department of Biological Sciences, Columbia, MO, USA
| |
Collapse
|
100
|
Das KP, Wood CR, Lin MT, Starkov AA, Lau C, Wallace KB, Corton JC, Abbott BD. Perfluoroalkyl acids-induced liver steatosis: Effects on genes controlling lipid homeostasis. Toxicology 2016; 378:37-52. [PMID: 28049043 DOI: 10.1016/j.tox.2016.12.007] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/21/2016] [Accepted: 12/29/2016] [Indexed: 02/07/2023]
Abstract
Persistent presence of perfluoroalkyl acids (PFAAs) in the environment is due to their extensive use in industrial and consumer products, and their slow decay. Biochemical tests in rodent demonstrated that these chemicals are potent modifiers of lipid metabolism and cause hepatocellular steatosis. However, the molecular mechanism of PFAAs interference with lipid metabolism remains to be elucidated. Currently, two major hypotheses are that PFAAs interfere with mitochondrial beta-oxidation of fatty acids and/or they affect the transcriptional activity of peroxisome proliferator-activated receptor α (PPARα) in liver. To determine the ability of structurally-diverse PFAAs to cause steatosis, as well as to understand the underlying molecular mechanisms, wild-type (WT) and PPARα-null mice were treated with perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), or perfluorohexane sulfonate (PFHxS), by oral gavage for 7days, and their effects were compared to that of PPARα agonist WY-14643 (WY), which does not cause steatosis. Increases in liver weight and cell size, and decreases in DNA content per mg of liver, were observed for all compounds in WT mice, and were also seen in PPARα-null mice for PFOA, PFNA, and PFHxS, but not for WY. In Oil Red O stained sections, WT liver showed increased lipid accumulation in all treatment groups, whereas in PPARα-null livers, accumulation was observed after PFNA and PFHxS treatment, adding to the burden of steatosis observed in control (untreated) PPARα-null mice. Liver triglyceride (TG) levels were elevated in WT mice by all PFAAs and in PPARα-null mice only by PFNA. In vitro β-oxidation of palmitoyl carnitine by isolated rat liver mitochondria was not inhibited by any of the 7 PFAAs tested. Likewise, neither PFOA nor PFOS inhibited palmitate oxidation by HepG2/C3A human liver cell cultures. Microarray analysis of livers from PFAAs-treated mice indicated that the PFAAs induce the expression of the lipid catabolism genes, as well as those involved in fatty acid and triglyceride synthesis, in WT mice and, to a lesser extent, in PPARα-null mice. These results indicate that most of the PFAAs increase liver TG load and promote steatosis in mice We hypothesize that PFAAs increase steatosis because the balance of fatty acid accumulation/synthesis and oxidation is disrupted to favor accumulation.
Collapse
Affiliation(s)
- Kaberi P Das
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA
| | - Carmen R Wood
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA
| | - Mimi T Lin
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA
| | - Anatoly A Starkov
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Christopher Lau
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA
| | - Kendall B Wallace
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - J Christopher Corton
- Integrated System Toxicology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA
| | - Barbara D Abbott
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA.
| |
Collapse
|