1
|
Rizan C, Rotchell JM, Eng PC, Robaire B, Ciocan C, Kapoor N, Kalra S, Sherman JD. Mitigating the environmental effects of healthcare: the role of the endocrinologist. Nat Rev Endocrinol 2025; 21:344-359. [PMID: 40082727 DOI: 10.1038/s41574-025-01098-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/16/2025]
Abstract
Human health depends on planetary health, and yet healthcare provision can have unintended consequences for the health of the planet. Emissions from the healthcare sector include greenhouse gases, air pollution and plastic pollution, alongside chemical contamination. Chemical pollution resulting in endocrine disruption has been associated with plastics, which are a source of concerning additives such as phthalates, bisphenols, perfluoroalkyl and polyfluoroalkyl substances, and flame retardants (all routinely found in healthcare products). Many endocrine-disrupting chemicals are persistent and ubiquitous in the environment (including water and food sources), with potential secondary harms for human health, including disrupting reproductive, metabolic and thyroid function. Here we review evidence-based strategies for mitigating environmental effects of healthcare delivery. We focus on what endocrinologists can do, including reducing demand for healthcare services through better preventative health, focusing on high-value care and improving sustainability of medical equipment and pharmaceuticals through adopting circular economy principles (including reduce, reuse and, as a last resort, recycle). The specific issue of endocrine-disrupting chemicals might be mitigated through responsible disposal and processing, alongside advocating for the use of alternative materials and replacing additive chemicals with those that have lower toxicity profiles, as well as tighter regulations. We must work to urgently transition to sustainable models of care provision, minimizing negative effects on human and planetary health.
Collapse
Affiliation(s)
- Chantelle Rizan
- Centre for Sustainable Medicine, National University of Singapore, Singapore, Singapore.
- Brighton and Sussex Medical School, Brighton, UK.
| | | | - Pei Chia Eng
- Department of Endocrinology, National University Hospital, Singapore, Singapore
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Bernard Robaire
- Faculty of Medicine and Biomedical Sciences, McGill University, Montreal, Quebec, Canada
| | - Corina Ciocan
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Nitin Kapoor
- Department of Endocrinology, Christian Medical College, Vellore, India
- The Non-Communicable Disease Unit, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Jodi D Sherman
- Yale School of Medicine, Yale University, New Haven, CT, USA
- Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
2
|
Yasuda A, Murase W, Kubota A, Uramaru N, Okuda K, Hakota R, Ikeda A, Kojima H. Effects of di-(2-ethylhexyl) phthalate and its metabolites on transcriptional activity via human nuclear receptors and gene expression in HepaRG cells. Toxicol In Vitro 2024; 101:105943. [PMID: 39341470 DOI: 10.1016/j.tiv.2024.105943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/14/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is widely used as a plasticizer in polyvinyl chloride products. DEHP exposure in humans is of great concern due to its endocrine-disrupting properties. In this study, we characterized the agonistic activities of DEHP and its five metabolites, mono-(2-ethylhexyl) phthalate (MEHP), 5OH-MEHP, 5oxo-MEHP, 5cx-MEPP and 2cx-MMHP against human nuclear receptors, peroxisome proliferator-activated receptor α (PPARα), pregnane X receptor (PXR), and constitutive androstane receptor (CAR) using transactivation assays. In the PPARα assay, the order of the agonistic activity was MEHP >> 5cx-MEPP >5OH-MEHP, 5oxo-MEHP >2cx-MMHP > DEHP, with DEHP significantly inhibiting MEHP-induced PPARα agonistic activity. This finding was compared to the results from in silico docking simulation. In the PXR assay, DEHP showed PXR agonistic activity more potent than that of MEHP, whereas the other metabolites showed little activity. In the CAR assay, none of the tested compounds showed agonistic activity. Moreover, the expression levels of PPARα-, PXR-, and CAR-target genes in HepaRG cells exposed to DEHP or MEHP were investigated using qRT-PCR analysis. As a result, exposure to these compounds significantly upregulated PXR/CAR target genes (CYP3A4 and CYP2B6), but not PPARα target genes (CYP4A11, etc.) in HepaRG cells. Taken together, these results suggest that direct PXR and/or indirect CAR activation by several DEHP metabolites may be involved in the endocrine disruption by altering hormone metabolism.
Collapse
Affiliation(s)
- Ayaka Yasuda
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Wataru Murase
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Atsuhito Kubota
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Naoto Uramaru
- School of Health and Social Services, Center for University-wide Education, Saitama Prefectural University, 820 San-Nomiya, Koshigaya, Saitama 343-8540, Japan; Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Katsuhiro Okuda
- Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa 078-8510, Japan
| | - Ryo Hakota
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Atsuko Ikeda
- Hokkaido University Faculty of Health Sciences, Kita-12, Nishi-5, Kita-ku, Sapporo 060-0812, Japan; Center for Environmental and Health Sciences, Hokkaido University, Kita-12, Nishi-7, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroyuki Kojima
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan; Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan.
| |
Collapse
|
3
|
Miller KL, Liu X, McSwain MG, Jauregui EJ, Langlais PR, Craig ZR. Quantitative label-free proteomic analysis of mouse ovarian antral follicles following oral exposure to a human-relevant mixture of three phthalates. Toxicol Sci 2024; 201:226-239. [PMID: 38995844 PMCID: PMC11424887 DOI: 10.1093/toxsci/kfae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024] Open
Abstract
Dibutyl phthalate (DBP), di-2-ethylhexyl phthalate (DEHP), and benzyl butyl phthalate (BBP) are used in personal and medical care products. In the ovary, antral follicles are essential for steroidogenesis and ovulation. DBP, BBP, and DEHP are known to inhibit mouse antral follicle growth and ovulation in vitro, and associate with decreased antral follicle counts in women. Given that the in vivo effects of a three-phthalate mixture on antral follicles are unknown, we evaluated the effects of a human-relevant mixture of DBP, BBP, and DEHP on ovarian follicles through proteome profiling analysis. Adult CD-1 female mice were fed corn oil (vehicle), or two dose levels of a phthalate mixture based on estimated exposures in general (32 µg/kg/d; PHT 32) and occupationally exposed (500 µg/kg/d; PHT 500) populations for 10 d. Antral follicles (>250 µm) were isolated and subjected to proteome profiling via label-free tandem mass spectrometry. A total of 5,417 antral follicle proteins were detected, of which 194 were differentially abundant between vehicle and PHT 32, and 136 between vehicle and PHT 500. Bioinformatic analysis revealed significantly different responses between the two phthalate doses. Protein abundance differences in the PHT 32 exposure mapped to cytoplasm, mitochondria, and lipid metabolism; whereas those in the PHT 500 exposure mapped to cytoplasm, nucleus, and phosphorylation. When both doses altered proteins mapped to common processes, the associated predicted transcription factors were different. These findings provide novel mechanistic insight into phthalate-associated, ovary-driven reproductive outcomes in women.
Collapse
Affiliation(s)
- Kara L Miller
- Department of Pharmacology & Toxicology, The University of Arizona, Tucson, AZ 85721, United States
| | - Xiaosong Liu
- School of Animal & Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, United States
| | - Maile G McSwain
- Environmental Health Transformative Research Undergraduate Experience, The University of Arizona, Tucson, AZ 85721, United States
| | - Estela J Jauregui
- School of Animal & Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, United States
| | - Paul R Langlais
- Department of Medicine, The University of Arizona, Tucson, AZ 85721, United States
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States
| | - Zelieann R Craig
- School of Animal & Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, United States
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ 85721, United States
| |
Collapse
|
4
|
Zamora Z, Wang S, Chen YW, Diamante G, Yang X. Systematic transcriptome-wide meta-analysis across endocrine disrupting chemicals reveals shared and unique liver pathways, gene networks, and disease associations. ENVIRONMENT INTERNATIONAL 2024; 183:108339. [PMID: 38043319 PMCID: PMC11216742 DOI: 10.1016/j.envint.2023.108339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/03/2023] [Accepted: 11/19/2023] [Indexed: 12/05/2023]
Abstract
Cardiometabolic disorders (CMD) are a growing public health problem across the world. Among the known cardiometabolic risk factors are compounds that induce endocrine and metabolic dysfunctions, such as endocrine disrupting chemicals (EDCs). To date, how EDCs influence molecular programs and cardiometabolic risks has yet to be fully elucidated, especially considering the complexity contributed by species-, chemical-, and dose-specific effects. Moreover, different experimental and analytical methodologies employed by different studies pose challenges when comparing findings across studies. To explore the molecular mechanisms of EDCs in a systematic manner, we established a data-driven computational approach to meta-analyze 30 human, mouse, and rat liver transcriptomic datasets for 4 EDCs, namely bisphenol A (BPA), bis(2-ethylhexyl) phthalate (DEHP), tributyltin (TBT), and perfluorooctanoic acid (PFOA). Our computational pipeline uniformly re-analyzed pre-processed quality-controlled microarray data and raw RNAseq data, derived differentially expressed genes (DEGs) and biological pathways, modeled gene regulatory networks and regulators, and determined CMD associations based on gene overlap analysis. Our approach revealed that DEHP and PFOA shared stable transcriptomic signatures that are enriched for genes associated with CMDs, suggesting similar mechanisms of action such as perturbations of peroxisome proliferator-activated receptor gamma (PPARγ) signaling and liver gene network regulators VNN1 and ACOT2. In contrast, TBT exhibited highly divergent gene signatures, pathways, network regulators, and disease associations from the other EDCs. In addition, we found that the rat, mouse, and human BPA studies showed highly variable transcriptomic patterns, providing molecular support for the variability in BPA responses. Our work offers insights into the commonality and differences in the molecular mechanisms of various EDCs and establishes a streamlined data-driven workflow to compare molecular mechanisms of environmental substances to elucidate the underlying connections between chemical exposure and disease risks.
Collapse
Affiliation(s)
- Zacary Zamora
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Susanna Wang
- Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Yen-Wei Chen
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | - Xia Yang
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| |
Collapse
|
5
|
PPARα Induces the Expression of CAR That Works as a Negative Regulator of PPARα Functions in Mouse Livers. Int J Mol Sci 2023; 24:ijms24043953. [PMID: 36835365 PMCID: PMC9960678 DOI: 10.3390/ijms24043953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor α (PPARα) is a transcription factor that controls the transcription of genes responsible for fatty acid metabolism. We have recently reported a possible drug-drug interaction mechanism via the interaction of PPARα with the xenobiotic nuclear receptor constitutive androstane receptor (CAR). Drug-activated CAR competes with the transcriptional coactivator against PPARα and prevents PPARα-mediated lipid metabolism. In this study, to elucidate the crosstalk between CAR and PPARα, we focused on the influence of PPARα activation on CAR's gene expression and activation. Male C57BL/6N mice (8-12 weeks old, n = 4) were treated with PPARα and CAR activators (fenofibrate and phenobarbital, respectively), and hepatic mRNA levels were determined using quantitative reverse transcription PCR. Reporter assays using the mouse Car promoter were performed in HepG2 cells to determine the PPARα-dependent induction of CAR. CAR KO mice were treated with fenofibrate, and the hepatic mRNA levels of PPARα target genes were determined. Treatment of mice with a PPARα activator increased Car mRNA levels as well as genes related to fatty acid metabolism. In reporter assays, PPARα induced the promoter activity of the Car gene. Mutation of the putative PPARα-binding motif prevented PPARα-dependent induction of reporter activity. In electrophoresis mobility shift assay, PPARα bound to the DR1 motif of the Car promoter. Since CAR has been reported to attenuate PPARα-dependent transcription, CAR was considered a negative feedback protein for PPARα activation. Treatment with fenofibrate induced the mRNA levels of PPARα target genes in Car-null mice more than those in wild-type mice, suggesting that CAR functions as a negative feedback factor for PPARα.
Collapse
|
6
|
Pan S, Guo Y, Yu W, Zhang J, Qiao X, Li L, Xu P, Zhai Y. Constitutive Androstane Receptor Agonist, TCPOBOP: Maternal Exposure Impairs the Growth and Development of Female Offspring in Mice. Int J Mol Sci 2023; 24:2602. [PMID: 36768963 PMCID: PMC9917268 DOI: 10.3390/ijms24032602] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Environmental chemicals, which are known to impact offspring health, have become a public concern. Constitutive activated receptor (CAR) is activated by various environmental chemicals and participates in xenobiotic metabolism. Here, we described the effects of maternal exposure to the CAR-specific ligand 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP, TC) on offspring health outcomes. Maternal TC exposure exhibited a stronger inhibition of body weight in 3-week-old and 8-week-old first-generation (F1) offspring female mice compared to controls. Further, maternal TC exposure obtained a strong increase in hepatic drug-metabolizing enzyme expression in 3-week-old female mice that persisted into 8-week-old adulthood. Interestingly, we observed distorted intestinal morphological features in 8-week-old F1 female mice in the TC-exposed group. Moreover, maternal TC exposure triggered a loss of intestinal barrier integrity by reducing the expression of intestinal tight junction proteins. Accordingly, maternal exposure to TC down-regulated serum triglyceride levels as well as decreased the expression of intestinal lipid uptake and transport marker genes. Mechanistically, maternal TC exposure activated the intestinal inflammatory response and disrupted the antioxidant system in the offspring female mice, thereby impeding the intestinal absorption of nutrients and seriously threatening offspring health. Altogether, these findings highlight that the effects of maternal TC exposure on offspring toxicity could not be ignored.
Collapse
Affiliation(s)
- Shijia Pan
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yuan Guo
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Wen Yu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jia Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xiaoxiao Qiao
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Letong Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Pengfei Xu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
7
|
Pan S, Guo Y, Yu W, Hong F, Qiao X, Zhang J, Xu P, Zhai Y. Environmental chemical TCPOBOP disrupts milk lipid homeostasis during pregnancy and lactation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114463. [PMID: 38321682 DOI: 10.1016/j.ecoenv.2022.114463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 02/08/2024]
Abstract
Humans are exposed to different kinds of environmental contaminants or drugs throughout their lifetimes. The widespread presence of these compounds has raised concerns about the consequent adverse effects on lactating women. The constitutive androstane receptor (CAR, Nr1i3) is known as a xenobiotic sensor for environmental pollution or drugs. In this study, the model environmental chemical 1, 4-bis [2-(3, 5-dichloropyridyloxy)] benzene, TCPOBOP (TC), which is a highly specific agonist of CAR, was used to investigate the effects of exogenous exposure on lactation function and offspring health in mice. The results revealed that TC exposure decreased the proliferation of mammary epithelial cells during pregnancy. This deficiency further compromised lobular-alveolar structures, resulting in alveolar cell apoptosis, as well as premature stoppage of the lactation cycle and aberrant lactation. Furthermore, TC exposure significantly altered the size and number of milk lipid droplets, suggesting that TC exposure inhibits milk lipid synthesis. Additionally, TC exposure interfered with the milk lipid metabolism network, resulting in the inability of TC-exposed mice to efficiently secrete nutrients and feed their offspring. These findings demonstrated that restricted synthesis and secretion of milk lipids would indirectly block mammary gland form and function, which explained the possible reasons for lactation failure and retarded offspring growth.
Collapse
Affiliation(s)
- Shijia Pan
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Yuan Guo
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Wen Yu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Fan Hong
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Xiaoxiao Qiao
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Jia Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Pengfei Xu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
8
|
Liu Q, Wei L, Chen X, Xu Y, Gao X, Zhao J. Three-dimensional (3D) thermal controlled polymer for simplified dispersive liquid-liquid microextraction in phthalic acid easters detection of straw. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Roy N, Alencastro F, Roseman BA, Wilson SR, Delgado ER, May MC, Bhushan B, Bello FM, Jurczak MJ, Shiva S, Locker J, Gingras S, Duncan AW. Dysregulation of Lipid and Glucose Homeostasis in Hepatocyte-Specific SLC25A34 Knockout Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1259-1281. [PMID: 35718058 PMCID: PMC9472157 DOI: 10.1016/j.ajpath.2022.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/18/2022] [Accepted: 06/08/2022] [Indexed: 10/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an epidemic affecting 30% of the US population. It is characterized by insulin resistance, and by defective lipid metabolism and mitochondrial dysfunction in the liver. SLC25A34 is a major repressive target of miR-122, a miR that has a central role in NAFLD and liver cancer. However, little is known about the function of SLC25A34. To investigate SLC25A34 in vitro, mitochondrial respiration and bioenergetics were examined using hepatocytes depleted of Slc25a34 or overexpressing Slc25a34. To test the function of SLC25A34 in vivo, a hepatocyte-specific knockout mouse was generated, and loss of SLC25A34 was assessed in mice maintained on a chow diet and a fast-food diet (FFD), a model for NAFLD. Hepatocytes depleted of Slc25a34 displayed increased mitochondrial biogenesis, lipid synthesis, and ADP/ATP ratio; Slc25a34 overexpression had the opposite effect. In the knockout model on chow diet, SLC25A34 loss modestly affected liver function (altered glucose metabolism was the most pronounced defect). RNA-sequencing revealed changes in metabolic processes, especially fatty acid metabolism. After 2 months on FFD, knockouts had a more severe phenotype, with increased lipid content and impaired glucose tolerance, which was attenuated after longer FFD feeding (6 months). This work thus presents a novel model for studying SLC25A34 in vivo in which SLC25A34 plays a role in mitochondrial respiration and bioenergetics during NAFLD.
Collapse
Affiliation(s)
- Nairita Roy
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Frances Alencastro
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bayley A Roseman
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sierra R Wilson
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Evan R Delgado
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meredith C May
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fiona M Bello
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael J Jurczak
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Departments of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph Locker
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew W Duncan
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
10
|
Leng J, Li H, Niu Y, Chen K, Yuan X, Chen H, Fu Z, Zhang L, Wang F, Chen C, Héroux P, Yang J, Zhu X, Lu W, Xia D, Wu Y. Low-dose mono(2-ethylhexyl) phthalate promotes ovarian cancer development through PPARα-dependent PI3K/Akt/NF-κB pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 790:147990. [PMID: 34380243 DOI: 10.1016/j.scitotenv.2021.147990] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 06/13/2023]
Abstract
The plasticizer di(2-ethylhexyl) phthalate (DEHP) and its hydrolysate mono(2-ethylhexyl) phthalate (MEHP) are major toxicants from plastics, but their association with hormone-dependent cancers has been controversial. We treated the human ovarian cancer cell lines SKOV3 and A2780 with low concentrations of DEHP/MEHP, and found that although no significant effect on cell proliferation was observed, ovarian cancer cell migration, invasion, and epithelial-mesenchymal transition (EMT) were promoted by submicromolar MEHP but not DEHP. Next, ovarian cancer patient data from The Cancer Genome Atlas (TCGA) were obtained and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) supported enrichment and Kaplan-Meier survival analyses, which identified PI3K/Akt pathway as a pivotal signaling pathway in ovarian cancer. We found that 500 nM MEHP treatment significantly increased PIK3CA expression, which could be reversed by the knockdown of peroxisome proliferator-activated receptor alpha (PPARα). Silencing PIK3CA significantly suppressed the MEHP-induced migration, invasion and EMT. In addition, we validated that MEHP treatment promoted phosphorylation of Akt and degradation of IκB-α, thereby activating NF-κB and enhancing NF-κB nuclear translocation. In nude mice, MEHP exposure significantly promoted the metastasis of ovarian cancer xenografts, which could be suppressed by the treatment of PPARα inhibitor GW6471. Our findings showed that low-dose MEHP promoted ovarian cancer progression through activating PI3K/Akt/NF-κB pathway, in a PPARα-dependent manner.
Collapse
Affiliation(s)
- Jing Leng
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyi Li
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Scientific Research Department, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuequn Niu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Yuan
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hanwen Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiqin Fu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lihuan Zhang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Wang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaoyi Chen
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou 310058, Zhejiang, China
| | - Paul Héroux
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Jun Yang
- Department of Public Health, Hangzhou Normal University School of Medicine, Hangzhou, China; Zhejiang Provincial Center for Uterine Cancer Diagnosis and Therapy Research of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinqiang Zhu
- Central Laboratory of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Weiguo Lu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
11
|
Zhou M, Yang J, Li Y. A model for phthalic acid esters' biodegradability and biotoxicity multi-effect pharmacophore and its application in molecular modification. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2021; 56:361-378. [PMID: 33563085 DOI: 10.1080/10934529.2021.1881352] [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: 08/01/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
The objective of this study was to investigate 13 phthalic acid esters (PAEs) with medium or long straight-alkyl-chain, branching or unsaturated side chains, because their structural characteristics make them difficult to biodegrade or highly toxic. A biodegradability and biotoxicity multi-effect pharmacophore model was built using comprehensive evaluation method. The results suggested that introducing hydrophobic groups to the side chains of the PAEs could improve the molecules' biodegradability and biotoxicity effects simultaneously. Thus, 40 target PAE (HEHP, DNOP, DUP) derivatives were designed. Two environmentally friendly PAE derivatives (HEHP-Anthryl and HEHP-Naphthyl) were screened via the test of environmental friendliness and functionality. In addition, the biodegradation and biotoxicity of derivatives were found to have improved as a result of the change in van der Waals forces between molecules and their corresponding proteins. Moreover, the environmental safety of the screened PAE derivatives was confirmed by predicting the toxicity of their intermediates and calculating the energy barrier values for biodegradation and metabolic pathways. This study could provide theoretical guidance for the practical development of environmentally friendly plasticizer.
Collapse
Affiliation(s)
- Mengying Zhou
- MOE Key Laboratory of Resources and Environmental Systems Optimization, North China Electric Power University, Beijing, China
- College of Environmental Science and Engineering, North China Electric Power University, Beijing, China
| | - Jiawen Yang
- MOE Key Laboratory of Resources and Environmental Systems Optimization, North China Electric Power University, Beijing, China
- College of Environmental Science and Engineering, North China Electric Power University, Beijing, China
| | - Yu Li
- MOE Key Laboratory of Resources and Environmental Systems Optimization, North China Electric Power University, Beijing, China
- College of Environmental Science and Engineering, North China Electric Power University, Beijing, China
| |
Collapse
|
12
|
Liu S, Wang K, Svoboda LK, Rygiel CA, Neier K, Jones TR, Cavalcante RG, Colacino JA, Dolinoy DC, Sartor MA. Perinatal DEHP exposure induces sex- and tissue-specific DNA methylation changes in both juvenile and adult mice. ENVIRONMENTAL EPIGENETICS 2021; 7:dvab004. [PMID: 33986952 PMCID: PMC8107644 DOI: 10.1093/eep/dvab004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/12/2021] [Accepted: 03/17/2021] [Indexed: 05/04/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a type of phthalate plasticizer found in a variety of consumer products and poses a public health concern due to its metabolic and endocrine disruption activities. Dysregulation of epigenetic modifications, including DNA methylation, has been shown to be an important mechanism for the pathogenic effects of prenatal exposures, including phthalates. In this study, we used an established mouse model to study the effect of perinatal DEHP exposure on the DNA methylation profile in liver (a primary target tissue of DEHP) and blood (a common surrogate tissue) of both juvenile and adult mice. Despite exposure ceasing at 3 weeks of age (PND21), we identified thousands of sex-specific differential DNA methylation events in 5-month old mice, more than identified at PND21, both in blood and liver. Only a small number of these differentially methylated cytosines (DMCs) overlapped between the time points, or between tissues (i.e. liver and blood), indicating blood may not be an appropriate surrogate tissue to estimate the effects of DEHP exposure on liver DNA methylation. We detected sex-specific DMCs common between 3-week and 5-month samples, pointing to specific DNA methylation alterations that are consistent between weanling and adult mice. In summary, this is the first study to assess the genome-wide DNA methylation profiles in liver and blood at two different aged cohorts in response to perinatal DEHP exposure. Our findings cast light on the implications of using surrogate tissue instead of target tissue in human population-based studies and identify epigenetic biomarkers for DEHP exposure.
Collapse
Affiliation(s)
- Siyu Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Kai Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Laurie K Svoboda
- Environmental Health Sciences, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Christine A Rygiel
- Environmental Health Sciences, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Kari Neier
- Environmental Health Sciences, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Tamara R Jones
- Environmental Health Sciences, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Raymond G Cavalcante
- Epigenomics Core, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Justin A Colacino
- Environmental Health Sciences, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
- Nutritional Sciences, University of Michigan, 500 S State St., Ann Arbor, MI 48109, USA
| | - Dana C Dolinoy
- Correspondence address. Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, USA. Tel: +734-647-3155; Fax: +734-936-7283; E-mail: (D.C.D.); Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave., Ann Arbor, MI 48109-2218, USA . Tel: +734-763-8013; Fax: +734-615-6553; E-mail: (M.A.S.)
| | - Maureen A Sartor
- Correspondence address. Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, USA. Tel: +734-647-3155; Fax: +734-936-7283; E-mail: (D.C.D.); Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave., Ann Arbor, MI 48109-2218, USA . Tel: +734-763-8013; Fax: +734-615-6553; E-mail: (M.A.S.)
| |
Collapse
|
13
|
Neier K, Montrose L, Chen K, Malloy MA, Jones TR, Svoboda LK, Harris C, Song PXK, Pennathur S, Sartor MA, Dolinoy DC. Short- and long-term effects of perinatal phthalate exposures on metabolic pathways in the mouse liver. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa017. [PMID: 33391822 PMCID: PMC7757125 DOI: 10.1093/eep/dvaa017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 06/12/2023]
Abstract
Phthalates have been demonstrated to interfere with metabolism, presumably by interacting with peroxisome proliferator-activated receptors (PPARs). However, mechanisms linking developmental phthalate exposures to long-term metabolic effects have not yet been elucidated. We investigated the hypothesis that developmental phthalate exposure has long-lasting impacts on PPAR target gene expression and DNA methylation to influence hepatic metabolic profiles across the life course. We utilized an established longitudinal mouse model of perinatal exposures to diethylhexyl phthalate and diisononyl phthalate, and a mixture of diethylhexyl phthalate+diisononyl phthalate. Exposure was through the diet and spanned from 2 weeks before mating until weaning at postnatal day 21 (PND21). Liver tissue was analyzed from the offspring of exposed and control mice at PND21 and in another cohort of exposed and control mice at 10 months of age. RNA-seq and pathway enrichment analyses indicated that acetyl-CoA metabolic processes were altered in diisononyl phthalate-exposed female livers at both PND21 and 10 months (FDR = 0.0018). Within the pathway, all 13 significant genes were potential PPAR target genes. Promoter DNA methylation was altered at three candidate genes, but persistent effects were only observed for Fasn. Targeted metabolomics indicated that phthalate-exposed females had decreased acetyl-CoA at PND21 and increased acetyl-CoA and acylcarnitines at 10 months. Together, our data suggested that perinatal phthalate exposures were associated with short- and long-term activation of PPAR target genes, which manifested as increased fatty acid production in early postnatal life and increased fatty acid oxidation in adulthood. This presents a novel molecular pathway linking developmental phthalate exposures and metabolic health outcomes.
Collapse
Affiliation(s)
- Kari Neier
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Luke Montrose
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Kathleen Chen
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Maureen A Malloy
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Tamara R Jones
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Laurie K Svoboda
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Craig Harris
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Peter X K Song
- Biostatistics, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, 1500 East Medical Center Drive 48109 MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, 1137 E. Catherine St. 48109 MI, USA
| | - Maureen A Sartor
- Biostatistics, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
- Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 100 Washtenaw Avenue 48109 MI, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
- Nutritional Sciences, University of Michigan, Ann Arbor, 1415 Washington Heights 48109 MI, USA
| |
Collapse
|
14
|
Preidis GA, Soni KG, Suh JH, Halder T, Kim KH, Choi JM, Li F, Devaraj S, Conner ME, Coarfa C, Jung SY, Moore DD. Coagulopathy in Malnourished Mice Is Sexually Dimorphic and Regulated by Nutrient-Sensing Nuclear Receptors. Hepatol Commun 2020; 4:1835-1850. [PMID: 33305154 PMCID: PMC7706303 DOI: 10.1002/hep4.1622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 11/23/2022] Open
Abstract
Liver dysfunction, including coagulopathy, is a prominent feature of protein-energy malnutrition. To identify mechanisms underlying malnutrition-associated coagulopathy, we administered a low-protein low-fat diet to lactating dams and examined hepatic transcription and plasma coagulation parameters in young adult weanlings. Malnutrition impacted body composition to a greater extent in male versus female mice. Transcriptional profiles suggested opposing effects of nutrient-sensing nuclear receptors, namely induction of peroxisome proliferator-activated receptor α (PPARα) targets and repression of farnesoid-X-receptor (FXR) targets. Coagulopathy with decreased synthesis of fibrinogen-α (FGA) and factor 11 (F11) was observed in malnourished male animals but not female animals. In primary mouse hepatocytes, FXR agonist increased and PPARα agonist decreased Fga and F11 messenger RNA expression. Nuclear receptor DNA response elements were identified in the Fga and F11 gene regulatory regions, and opposing effects of FXR and PPARα were confirmed with luciferase assays. Unexpectedly, hepatic PPARα protein was markedly depleted in malnourished male liver and was not enriched on Fga or F11 response elements. Rather, there was loss of FXR binding at these response elements. Reduced PPARα protein was associated with loss of hepatocyte peroxisomes, which are necessary for bile acid biosynthesis, and with decreased concentrations of bile acids that function as FXR ligands, most notably the FXR agonist chenodeoxycholic acid. Conclusion: Malnutrition impairs growth and liver synthetic function more severely in male mice than in female mice. Malnourished male mice are coagulopathic and exhibit decreased hepatocyte peroxisomes, FXR agonist bile acids, FXR binding on Fga and F11 gene regulatory elements, and coagulation factor synthesis. These effects are absent in female mice, which have low baseline levels of PPARα, suggesting that nutrient-sensing nuclear receptors regulate coagulation factor synthesis in response to host nutritional status in a sex-specific manner.
Collapse
Affiliation(s)
- Geoffrey A. Preidis
- Section of Gastroenterology, Hepatology & NutritionDepartment of PediatricsBaylor College of Medicine and Texas Children’s HospitalHoustonTXUSA
| | - Krishnakant G. Soni
- Section of Gastroenterology, Hepatology & NutritionDepartment of PediatricsBaylor College of Medicine and Texas Children’s HospitalHoustonTXUSA
| | - Ji Ho Suh
- Section of Gastroenterology, Hepatology & NutritionDepartment of PediatricsBaylor College of Medicine and Texas Children’s HospitalHoustonTXUSA
| | - Tripti Halder
- Section of Gastroenterology, Hepatology & NutritionDepartment of PediatricsBaylor College of Medicine and Texas Children’s HospitalHoustonTXUSA
| | - Kang Ho Kim
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Jong Min Choi
- Advanced Technology CoreMass Spectrometry Proteomics CoreBaylor College of MedicineHoustonTXUSA
| | - Feng Li
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Department of Pathology and ImmunologyBaylor College of MedicineHoustonTXUSA
| | - Sridevi Devaraj
- Department of Pathology and ImmunologyBaylor College of MedicineHoustonTXUSA
| | - Margaret E. Conner
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTXUSA
| | - Cristian Coarfa
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Sung Yun Jung
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Advanced Technology CoreMass Spectrometry Proteomics CoreBaylor College of MedicineHoustonTXUSA
- Verna and Marrs McLean Department of Biochemistry and Molecular BiologyBaylor College of MedicineHoustonTXUSA
| | - David D. Moore
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
15
|
Küblbeck J, Niskanen J, Honkakoski P. Metabolism-Disrupting Chemicals and the Constitutive Androstane Receptor CAR. Cells 2020; 9:E2306. [PMID: 33076503 PMCID: PMC7602645 DOI: 10.3390/cells9102306] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
During the last two decades, the constitutive androstane receptor (CAR; NR1I3) has emerged as a master activator of drug- and xenobiotic-metabolizing enzymes and transporters that govern the clearance of both exogenous and endogenous small molecules. Recent studies indicate that CAR participates, together with other nuclear receptors (NRs) and transcription factors, in regulation of hepatic glucose and lipid metabolism, hepatocyte communication, proliferation and toxicity, and liver tumor development in rodents. Endocrine-disrupting chemicals (EDCs) constitute a wide range of persistent organic compounds that have been associated with aberrations of hormone-dependent physiological processes. Their adverse health effects include metabolic alterations such as diabetes, obesity, and fatty liver disease in animal models and humans exposed to EDCs. As numerous xenobiotics can activate CAR, its role in EDC-elicited adverse metabolic effects has gained much interest. Here, we review the key features and mechanisms of CAR as a xenobiotic-sensing receptor, species differences and selectivity of CAR ligands, contribution of CAR to regulation hepatic metabolism, and evidence for CAR-dependent EDC action therein.
Collapse
Affiliation(s)
- Jenni Küblbeck
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
| | - Jonna Niskanen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210 Kuopio, Finland;
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Campus Box 7569, Chapel Hill, NC 27599-7569, USA
| |
Collapse
|
16
|
Early microRNA indicators of PPARα pathway activation in the liver. Toxicol Rep 2020; 7:805-815. [PMID: 32642447 PMCID: PMC7334544 DOI: 10.1016/j.toxrep.2020.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/01/2020] [Accepted: 06/19/2020] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNA species that play key roles in post-transcriptional regulation of gene expression. MiRNAs also serve as a promising source of early biomarkers for different environmental exposures and health effects, although there is limited information linking miRNA changes to specific target pathways. In this study, we measured liver miRNAs in male B6C3F1 mice exposed to a known chemical activator of the peroxisome proliferator-activated receptor alpha (PPARα) pathway, di(2-ethylhexyl) phthalate (DEHP), for 7 and 28 days at concentrations of 0, 750, 1500, 3000, or 6000 ppm in feed. At the highest dose tested, DEHP altered 61 miRNAs after 7 days and 171 miRNAs after 28 days of exposure, with 48 overlapping miRNAs between timepoints. Analysis of these 48 common miRNAs indicated enrichment in PPARα–related targets and other pathways related to liver injury and cancer. Four of the 10 miRNAs exhibiting a clear dose trend were linked to the PPARα pathway: mmu-miRs-125a-5p, -182−5p, -20a−5p, and -378a−3p. mmu-miRs-182−5p and -378a−3p were subsequently measured using digital drop PCR across a dose range for DEHP and two related phthalates with weaker PPARα activity, di-n-octyl phthalate and n-butyl benzyl phthalate, following 7-day exposures. Analysis of mmu-miRs-182−5p and -378a−3p by transcriptional benchmark dose analysis correctly identified DEHP as having the greatest potency. However, benchmark dose estimates for DEHP based on these miRNAs (average 163; range 126−202 mg/kg-day) were higher on average than values for PPARα target genes (average 74; range 29−183 mg/kg-day). These findings identify putative miRNA biomarkers of PPARα pathway activity and suggest that early miRNA changes may be used to stratify chemical potency.
Collapse
Key Words
- AIC, Akaike Information Criterion
- ALT, alanine aminotransferase
- AOP, adverse outcome pathway
- AST, aspartate aminotransferase
- Acox1, acyl-Coenzyme A oxidase 1
- Adverse outcome pathway (AOP)
- AhR, aryl hydrocarbon receptor
- BBP, n-butyl benzyl phthalate
- BMD, benchmark dose
- BMDA, apical-based benchmark dose
- BMDL, BMD lower confidence interval
- BMDT, transcriptional-based benchmark dose
- BMR, benchmark response
- BROD, benzyloxyresorufin O-debenzylation
- Benchmark dose (BMD)
- Biomarkers
- CAR, constitutive androstane receptor
- DEGs, differentially expressed genes
- DEHP, di (2-thylhexyl) phthalate
- DEmiRs, differentially expressed miRNAs
- DNOP, di-n-octyl phthalate
- EPA, U.S. Environmental Protection Agency
- EROD, ethoxyresorufin O-dealkylation
- GEO, Gene Expression Omnibus
- HCA, hepatocellular adenoma
- HCC, hepatocellular carcinoma
- Hepatocellular carcinoma
- IPA, Ingenuity Pathway Analysis
- Liver toxicity
- MOA, mode of action
- MicroRNAs
- Mode of action (MOA)
- Nrf2, nuclear receptor erythroid 2-like 2
- POD, point-of-departure
- PPARα, peroxisome proliferator-activated receptor alpha
- PROD, pentoxyresorufin O-depentylation
- PXR, pregnane X receptor
- Peroxisome proliferator-activated receptor alpha (PPARα)
- Phthalate
- SDH, sorbitol dehydrogenase
- TMM, trimmed mean of M-values
- ddPCR, droplet digital polymerase chain reaction
- mRNA, messenger RNA
- miRNAs, microRNAs
- mtDNA, mitochondrial
- rRNA, ribosomal RNA
- smallRNA-seq, small RNA sequencing
- tRNA, transfer RNA
Collapse
|
17
|
Yamaguchi T, Maeda M, Ogata K, Abe J, Utsumi T, Kimura K. The effects on the endocrine system under hepatotoxicity induction by phenobarbital and di(2-ethylhexyl)phthalate in intact juvenile male rats. J Toxicol Sci 2019; 44:459-469. [PMID: 31270302 DOI: 10.2131/jts.44.459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Phenobarbital (PB) and Di (2-ethylhexyl) phthalate (DEHP), an anti-epileptic drug and a plasticizer used in flexible polyvinylchloride formulations, respectively, are well-known typical hepatotoxicants. This study investigated the effects of PB (100 mg/kg/day) or DEHP (500 mg/kg/day) on the endocrine system in intact juvenile/peripubertal male rats exposed for 31 days beginning on postnatal day 23. Slight hormone level changes, histopathological changes in thyroid gland or induction of UDP-glucuronosyltransferase in liver were observed in both the PB and DEHP groups. One of the assumed mechanisms inducing thyroid effects is predictable to be secondary changes based on the enhancement in thyroid hormone metabolism via the induction of hepatic microsomal enzymes. No reproductive system-related changes in organ weights, histopathology, and sexual maturation were observed in both groups. Lower testosterone level was observed in the PB group. CYP2B and CYP3A, which are involved in testosterone metabolism, were induced in liver of the PB group. There was no change of 17β-hydroxysteroid dehydrogenase activity in testis of both groups. Lower testosterone level in the PB-treated male rats was attributed to an indirect, hepatotoxicity-associated effect on the reproductive system and not to direct effects on testis such as the antiandrogenic activity and the inhibition of steroidogenesis. These results did not indicate that PB or DEHP exposure affects the endocrine system directly.
Collapse
Affiliation(s)
- Takafumi Yamaguchi
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd.,Laboratory of Animal Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| | - Minoru Maeda
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd
| | - Keiko Ogata
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd
| | - Jun Abe
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd
| | - Toru Utsumi
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd
| | - Koji Kimura
- Laboratory of Animal Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University
| |
Collapse
|
18
|
Ito Y, Kamijima M, Nakajima T. Di(2-ethylhexyl) phthalate-induced toxicity and peroxisome proliferator-activated receptor alpha: a review. Environ Health Prev Med 2019; 24:47. [PMID: 31279339 PMCID: PMC6612219 DOI: 10.1186/s12199-019-0802-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/28/2019] [Indexed: 11/10/2022] Open
Abstract
The plasticizer di(2-ethylhexyl) phthalate (DEHP) has been widely used in the manufacture of polyvinyl chloride-containing products such as medical and consumer goods. Humans can easily be exposed to it because DEHP is ubiquitous in the environment. Recent research on the adverse effects of DEHP has focused on reproductive and developmental toxicity in rodents and/or humans. DEHP is a representative of the peroxisome proliferators. Therefore, peroxisome proliferator-activated receptor alpha (PPARα)-dependent pathways are the expected mode of action of several kinds of DEHP-induced toxicities. In this review, we summarize DEHP kinetics and its mechanisms of carcinogenicity and reproductive and developmental toxicity in relation to PPARα. Additionally, we give an overview of the impacts of science policy on exposure sources.
Collapse
Affiliation(s)
- Yuki Ito
- Department of Occupational and Environmental Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601 Japan
| | - Michihiro Kamijima
- Department of Occupational and Environmental Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601 Japan
| | - Tamie Nakajima
- College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501 Japan
| |
Collapse
|
19
|
Rampersaud A, Lodato NJ, Shin A, Waxman DJ. Widespread epigenetic changes to the enhancer landscape of mouse liver induced by a specific xenobiotic agonist ligand of the nuclear receptor CAR. Toxicol Sci 2019; 171:315-338. [PMID: 31236583 PMCID: PMC6760311 DOI: 10.1093/toxsci/kfz148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022] Open
Abstract
CAR (Nr1i3), a liver nuclear receptor and xenobiotic sensor, induces drug, steroid and lipid metabolism and dysregulates genes linked to hepatocellular carcinogenesis, but its impact on the liver epigenome is poorly understood. TCPOBOP, a halogenated xenochemical and highly specific CAR agonist ligand, induces localized chromatin opening or closing at several thousand mouse liver genomic regions, discovered as differential DNase-hypersensitive sites (ΔDHS). Active enhancer and promoter histone marks induced by TCPOBOP were enriched at opening DHS and TCPOBOP-inducible genes. Enrichment of CAR binding and CAR motifs was seen at opening DHS and their inducible drug/lipid metabolism gene targets, and at many constitutively open DHS located nearby. TCPOBOP-responsive cell cycle and DNA replication genes co-dependent on MET/EGFR signaling for induction were also enriched for CAR binding. A subset of opening DHS and many closing DHS mapping to TCPOBOP-responsive target genes did not bind CAR, indicating an indirect mechanism for their changes in chromatin accessibility. TCPOBOP-responsive DHS were also enriched for induced binding of RXRA, CEBPA and CEBPB, and for motifs for liver-enriched factors that may contribute to liver-specific transcriptional responses to TCPOBOP exposure. These studies elucidate the enhancer landscape of TCPOBOP-exposed liver and the widespread epigenetic changes that are induced by both direct and indirect mechanisms linked to CAR activation. The global maps of thousands of environmental chemical-induced epigenetic changes described here constitute a rich resource for further research on xenochemical effects on liver chromatin states and the epigenome.
Collapse
Affiliation(s)
- Andy Rampersaud
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA USA
| | - Nicholas J Lodato
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA USA
| | - Aram Shin
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA USA
| |
Collapse
|
20
|
Hsiao WT, Su HM, Su KP, Chen SH, Wu HP, You YL, Fu RH, Chao PM. Deficiency or activation of peroxisome proliferator-activated receptor α reduces the tissue concentrations of endogenously synthesized docosahexaenoic acid in C57BL/6J mice. Nutr Res Pract 2019; 13:286-294. [PMID: 31388404 PMCID: PMC6669072 DOI: 10.4162/nrp.2019.13.4.286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/12/2019] [Accepted: 04/02/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND/OBJECTIVES Docosahexaenoic acid (DHA), an n-3 long chain polyunsaturated fatty acid (LCPUFA), is acquired by dietary intake or the in vivo conversion of α-linolenic acid. Many enzymes participating in LCPUFA synthesis are regulated by peroxisome proliferator-activated receptor alpha (PPARα). Therefore, it was hypothesized that the tissue accretion of endogenously synthesized DHA could be modified by PPARα. MATERIALS/METHODS The tissue DHA concentrations and mRNA levels of genes participating in DHA biosynthesis were compared among PPARα homozygous (KO), heterozygous (HZ), and wild type (WT) mice (Exp I), and between WT mice treated with clofibrate (PPARα agonist) or those not treated (Exp II). In ExpII, the expression levels of the proteins associated with DHA function in the brain cortex and retina were also measured. An n3-PUFA depleted/replenished regimen was applied to mitigate the confounding effects of maternal DHA. RESULTS PPARα ablation reduced the hepatic Acox, Fads1, and Fads2 mRNA levels, as well as the DHA concentration in the liver, but not in the brain cortex. In contrast, PPARα activation increased hepatic Acox, Fads1, Fads2 and Elovl5 mRNA levels, but reduced the DHA concentrations in the liver, retina, and phospholipid of brain cortex, and decreased mRNA and protein levels of the brain-derived neurotrophic factor in brain cortex. CONCLUSIONS LCPUFA enzyme expression was altered by PPARα. Either PPARα deficiency or activation-decreased tissue DHA concentration is a stimulus for further studies to determine the functional significance.
Collapse
Affiliation(s)
- Wen-Ting Hsiao
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan
| | - Hui-Min Su
- Graduate Institute of Physiology, National Taiwan University, Taipei 100, Taiwan
| | - Kuan-Pin Su
- Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung 404, Taiwan
| | - Szu-Han Chen
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan
| | - Hai-Ping Wu
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan
| | - Yi-Ling You
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan
| | - Ru-Huei Fu
- Graduate Institute of Immunology, China Medical University, Taichung 404, Taiwan
| | - Pei-Min Chao
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan
| |
Collapse
|
21
|
Lodato NJ, Rampersaud A, Waxman DJ. Impact of CAR Agonist Ligand TCPOBOP on Mouse Liver Chromatin Accessibility. Toxicol Sci 2019; 164:115-128. [PMID: 29617930 DOI: 10.1093/toxsci/kfy070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Activation of the nuclear receptor and transcription factor CAR (Nr1i3) by its specific agonist ligand TCPOBOP (1, 4-bis[2-(3, 5-dichloropyridyloxy)]benzene) dysregulates hundreds of genes in mouse liver and is linked to male-biased hepatocarcinogenesis. To elucidate the genomic organization of CAR-induced gene responses, we investigated the distribution of TCPOBOP-responsive RefSeq coding and long noncoding RNA (lncRNA) genes across the megabase-scale topologically associating domains (TADs) that segment the genome, and which provide a structural framework that functionally constrains enhancer-promoter interactions. We show that a subset of TCPOBOP-responsive genes cluster within TADs, and that TCPOBOP-induced genes and TCPOBOP-repressed genes are often found in different TADs. Further, using DNase-seq and DNase hypersensitivity site (DHS) analysis, we identified several thousand genomic regions (ΔDHS) where short-term exposure to TCPOBOP induces localized changes (increases or decreases) in mouse liver chromatin accessibility, many of which cluster in TADs together with TCPOBOP-responsive genes. Sites of chromatin opening were highly enriched nearby genes induced by TCPOBOP and chromatin closing was highly enriched nearby genes repressed by TCPOBOP, consistent with TCPOBOP-responsive ΔDHS serving as enhancers and promoters that positively regulate CAR-responsive genes. Gene expression changes lagged behind chromatin opening or closing for a subset of TCPOBOP-responsive ΔDHS. ΔDHS that were specifically responsive to TCPOBOP in male liver were significantly enriched for genomic regions with a basal male bias in chromatin accessibility; however, the male-biased response of hepatocellular carcinoma-related genes to TCPOBOP was not associated with a correspondingly male-biased ΔDHS response. These studies elucidate the genome-wide organization of CAR-responsive genes and of the thousands of associated genomic sites where TCPOBOP exposure induces both rapid and persistent changes in chromatin accessibility.
Collapse
Affiliation(s)
- Nicholas J Lodato
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - Andy Rampersaud
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
22
|
Huang Y, Wu C, Ye Y, Zeng J, Zhu J, Li Y, Wang W, Zhang W, Chen Y, Xie H, Zhang H, Liu J. The Increase of ROS Caused by the Interference of DEHP with JNK/p38/p53 Pathway as the Reason for Hepatotoxicity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E356. [PMID: 30691201 PMCID: PMC6388128 DOI: 10.3390/ijerph16030356] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 01/06/2023]
Abstract
As the most commonly used plasticizer, Di-(2-ethylhexyl)-phthalate (DEHP) exists everywhere in the environment due to the widespread use of polyvinyl chloride (PVC) in human life, and it is also a recognized environmental pollutant. Studies have proved the hepatotoxicity of DEHP, however the mechanism has not been adequately explored, especially the role of the reactive oxygen species (ROS) in it. In the present study, 21 day-old ICR mice were administered DEHP with dose of 0, 125, 250, and 375 mg/kg/day for 28 days by intragastrical gavage. After contamination, histopathology displayed that liver tissue were damaged mildly with the effect of DEHP; a significant increase of the serum liver function index (including aspartate transaminase (AST) and alanine transaminase (ALT)) were observed. Additionally, the level of lipid peroxidation markedly rise, especially ROS and malondialdehyde (MDA), but the activation of superoxide dismutase (SOD) was obviously decreased in mice liver. In addition, DEHP promoted the phosphorylation of JNK and p38MAPK proteins in mice liver, as well as increased the expression of p53 protein and decreased the level of DNA methylation in the p53 gene promoter region. These results indicated that the hepatotoxicity of mice caused by DEHP may be through activating the JNK/p38MAPK/p53 signaling pathway and further promoting the generation of ROS to induce lipid peroxidation in liver, and the role of DNA methylation may be inevitable.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Chuancheng Wu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Youbin Ye
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Jingwen Zeng
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Jianlin Zhu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Yuchen Li
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Wenxiang Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Xueyan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Wenchang Zhang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Yiqin Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Xueyan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Hongyuan Xie
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Hongmei Zhang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| | - Jin Liu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyuan Road No.1, Minhou County, Fuzhou 350108, China.
| |
Collapse
|
23
|
Felter SP, Foreman JE, Boobis A, Corton JC, Doi AM, Flowers L, Goodman J, Haber LT, Jacobs A, Klaunig JE, Lynch AM, Moggs J, Pandiri A. Human relevance of rodent liver tumors: Key insights from a Toxicology Forum workshop on nongenotoxic modes of action. Regul Toxicol Pharmacol 2018; 92:1-7. [PMID: 29113941 PMCID: PMC11350555 DOI: 10.1016/j.yrtph.2017.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/21/2022]
Abstract
The Toxicology Forum sponsored a workshop in October 2016, on the human relevance of rodent liver tumors occurring via nongenotoxic modes of action (MOAs). The workshop focused on two nuclear receptor-mediated MOAs (Constitutive Androstane Receptor (CAR) and Peroxisome Proliferator Activated Receptor-alpha (PPARα), and on cytotoxicity. The goal of the meeting was to review the state of the science to (1) identify areas of consensus and differences, data gaps and research needs; (2) identify reasons for inconsistencies in current regulatory positions; and (3) consider what data are needed to demonstrate a specific MOA, and when additional research is needed to rule out alternative possibilities. Implications for quantitative risk assessment approaches were discussed, as were implications of not considering MOA and dose in hazard characterization and labeling schemes. Most, but not all, participants considered the CAR and PPARα MOAs as not relevant to humans based on quantitative and qualitative differences. In contrast, cytotoxicity is clearly relevant to humans, but a threshold applies. Questions remain for all three MOAs concerning what data are necessary to determine the MOA and to what extent it is necessary to exclude other MOAs.
Collapse
Affiliation(s)
- Susan P Felter
- Procter and Gamble, Central Product Safety, Mason, OH, United States.
| | | | - Alan Boobis
- Department of Medicine, Imperial College London, London, UK
| | - J Christopher Corton
- National Health and Environmental Effects Research Lab, US EPA, Durham, NC, United States
| | - Adriana M Doi
- BASF Corporation, Research Triangle Park, NC, United States
| | - Lynn Flowers
- Office of Science Policy, US EPA, Washington DC, United States
| | - Jay Goodman
- Michigan State University, Dept. Pharmacology and Toxicology, East Lansing, MI, United States
| | - Lynne T Haber
- Risk Science Center, Dept. of Environmental Health, University of Cincinnati, Cincinnati, OH, United States
| | - Abigail Jacobs
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | | | | | - Jonathan Moggs
- Novartis Institutes for BioMedical Research, Preclinical Safety, Translational Medicine, Basel, Switzerland
| | - Arun Pandiri
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
24
|
Corton JC, Peters JM, Klaunig JE. The PPARα-dependent rodent liver tumor response is not relevant to humans: addressing misconceptions. Arch Toxicol 2017; 92:83-119. [PMID: 29197930 DOI: 10.1007/s00204-017-2094-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
A number of industrial chemicals and therapeutic agents cause liver tumors in rats and mice by activating the nuclear receptor peroxisome proliferator-activated receptor α (PPARα). The molecular and cellular events by which PPARα activators induce rodent hepatocarcinogenesis have been extensively studied elucidating a number of consistent mechanistic changes linked to the increased incidence of liver neoplasms. The weight of evidence relevant to the hypothesized mode of action (MOA) for PPARα activator-induced rodent hepatocarcinogenesis is summarized here. Chemical-specific and mechanistic data support concordance of temporal and dose-response relationships for the key events associated with many PPARα activators. The key events (KE) identified in the MOA are PPARα activation (KE1), alteration in cell growth pathways (KE2), perturbation of hepatocyte growth and survival (KE3), and selective clonal expansion of preneoplastic foci cells (KE4), which leads to the apical event-increases in hepatocellular adenomas and carcinomas (KE5). In addition, a number of concurrent molecular and cellular events have been classified as modulating factors, because they potentially alter the ability of PPARα activators to increase rodent liver cancer while not being key events themselves. These modulating factors include increases in oxidative stress and activation of NF-kB. PPARα activators are unlikely to induce liver tumors in humans due to biological differences in the response of KEs downstream of PPARα activation. This conclusion is based on minimal or no effects observed on cell growth pathways and hepatocellular proliferation in human primary hepatocytes and absence of alteration in growth pathways, hepatocyte proliferation, and tumors in the livers of species (hamsters, guinea pigs and cynomolgus monkeys) that are more appropriate human surrogates than mice and rats at overlapping dose levels. Despite this overwhelming body of evidence and almost universal acceptance of the PPARα MOA and lack of human relevance, several reviews have selectively focused on specific studies that, as discussed, contradict the consensus opinion and suggest uncertainty. In the present review, we systematically address these most germane suggested weaknesses of the PPARα MOA.
Collapse
Affiliation(s)
- J Christopher Corton
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr, MD-B105-03, Research Triangle Park, NC, 27711, USA.
| | - Jeffrey M Peters
- The Department of Veterinary and Biomedical Sciences and Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, 16803, USA
| | - James E Klaunig
- Department of Environmental Health, Indiana University, Bloomington, IN, 47402, USA
| |
Collapse
|
25
|
PPARα-independent transcriptional targets of perfluoroalkyl acids revealed by transcript profiling. Toxicology 2017; 387:95-107. [PMID: 28558994 DOI: 10.1016/j.tox.2017.05.013] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 01/09/2023]
Abstract
Perfluoroalkyl acids (PFAAs) are ubiquitous and persistent environmental contaminants. Compounds such as perfluoroocanoic acid (PFOA), perfluorooctane sulfonate (PFOS), perfluorononanoic acid (PFNA), and perfluorohexane sulfonate (PFHxS) are readily found in the tissues of humans and wildlife. While PFOA and PFOS have been the subject of numerous studies since they were first described over a decade ago, less is known about the biological activity of PFHxS and PFNA. Most PFAAs are activators of peroxisome proliferator-activated receptor α (PPARα), although the biological effects of these compounds are likely mediated by other factors in addition to PPARα. To evaluate the effects of PFHxS and PFNA, male wild-type and Pparα-null mice were dosed by oral gavage with PFHxS (3 or 10mg/kg/day), PFNA (1 or 3mg/kg/day), or vehicle for 7days, and liver gene expression was evaluated by full-genome microarrays. Gene expression patterns were then compared to historical in-house data for PFOA and PFOS in addition to the experimental hypolipidemic agent, WY-14,643. While WY-14,643 altered most genes in a PPARα-dependent manner, approximately 11-24% of regulated genes in PFAA-treated mice were independent of PPARα. The possibility that PFAAs regulate gene expression through other molecular pathways was evaluated. Using data available through a microarray database, PFAA gene expression profiles were found to exhibit significant similarity to profiles from mouse tissues exposed to agonists of the constitutive activated receptor (CAR), estrogen receptor α (ERα), and PPARγ. Human PPARγ and ERα were activated by all four PFAAs in trans-activation assays from the ToxCast screening program. Predictive gene expression biomarkers showed that PFAAs activate CAR in both genotypes and cause feminization of the liver transcriptome through suppression of signal transducer and activator of transcription 5B (STAT5B). These results indicate that, in addition to activating PPARα as a primary target, PFAAs also have the potential to activate CAR, PPARγ, and ERα as well as suppress STAT5B.
Collapse
|
26
|
Kakehashi A, Stefanov VE, Ishii N, Okuno T, Fujii H, Kawai K, Kawada N, Wanibuchi H. Proteome Characteristics of Non-Alcoholic Steatohepatitis Liver Tissue and Associated Hepatocellular Carcinomas. Int J Mol Sci 2017; 18:ijms18020434. [PMID: 28218651 PMCID: PMC5343968 DOI: 10.3390/ijms18020434] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/24/2017] [Accepted: 02/09/2017] [Indexed: 12/18/2022] Open
Abstract
To uncover mechanisms of nonalcoholic steatohepatitis (NASH) associated hepatocarcinogenesis, we compared the proteomes of human NASH-associated liver biopsies, resected hepatocellular carcinomas (HCCs) and HCCs of HCV⁺ patients with normal liver tissue of patients with gastrointestinal tumor metastasis, in formalin-fixed paraffin-embedded samples obtained after surgery in our hospital during the period from 2006 to 2011. In addition, proteome analysis of liver tumors in male STAM NASH-model mice was performed. Similar changes in the proteome spectrum such as overexpression of enzymes involved in lipid, cholesterol and bile acid biosynthesis and examples associated with suppression of fatty acid oxidation and catabolism, alcohol metabolism, mitochondrial function as well as low expression levels of cytokeratins 8 and 18 were observed in both human NASH biopsies and NASH HCCs, but not HCV⁺ HCCs. Alterations in downstream protein expression pointed to significant activation of transforming growth factor β, SMAD family member 3, β-catenin, Nrf2, SREBP-LXRα and nuclear receptor-interacting protein 1 (NRIP1), and inhibition of PPARs and p53 in human NASH biopsies and/or HCCs, suggesting their involvement in accumulation of lipids, development of fibrosis, oxidative stress, cell proliferation and suppression of apoptosis in NASH hepatocarcinogenesis. In STAM mice, PPARs inhibition was not obvious, while expression of cytokeratins 8 and 18 was elevated, indicative of essential differences between human and mouse NASH pathogenesis.
Collapse
Affiliation(s)
- Anna Kakehashi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka 545-8585, Japan.
| | - Vasily E Stefanov
- Department of Biochemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
| | - Naomi Ishii
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka 545-8585, Japan.
| | - Takahiro Okuno
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka 545-8585, Japan.
| | - Hideki Fujii
- Department of Hepatology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan.
| | - Kazuaki Kawai
- Department of Environmental Oncology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan.
| | - Norifumi Kawada
- Department of Hepatology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan.
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka 545-8585, Japan.
| |
Collapse
|
27
|
Ling J, Lopez-Dee ZP, Cottell C, Wolfe L, Nye D. Regulation of mRNA Translation Is a Novel Mechanism for Phthalate Toxicity. PLoS One 2016; 11:e0167914. [PMID: 27992464 PMCID: PMC5167351 DOI: 10.1371/journal.pone.0167914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/22/2016] [Indexed: 12/29/2022] Open
Abstract
Phthalates are a group of plasticizers that are widely used in many consumer products and medical devices, thus generating a huge burden to human health. Phthalates have been known to cause a number of developmental and reproductive disorders functioning as endocrine modulators. They are also involved in carcinogenesis with mechanisms less understood. To further understand the molecular mechanisms of phthalate toxicity, in this study we reported a new effect of phthalates on mRNA translation/protein synthesis, a key regulatory step of gene expression. Butyl benzyl phthalate (BBP) was found to directly inhibit mRNA translation in vitro but showed a complicated pattern of affecting mRNA translation in cells. In human kidney embryonic cell (HEK-293T), BBP increased cap-dependent mRNA translation at lower concentrations but showed inhibitory effect at higher concentrations. Cap-independent translation was not affected. On the other hand, mono (2-ethylhexyl) phthalate (MEHP) as a major metabolite of another important phthalate di (2-ethylhexyl) phthalate (DEHP) inhibited both can-dependent and -independent mRNA translation in vivo. In contrast, BBP and MEHP exhibited an overall promoting effect on mRNA translation in cancer cells. Mechanistic studies identified that the level and phosphorylation of eIF4E-BP (eIF4E binding protein) and the amount of eIF4GI in eIF4F complex were altered in accordance with the effect of BBP on translation. BBP was also identified to directly bind to eIF4E, providing a further mechanism underlying the regulation of mRNA by phthalate. At the cellular level BBP inhibited normal cell growth but slightly promoted cancer cells (HT29) growth. Overall, this study provides the first evidence that phthalates can directly regulate mRNA translation as a novel mechanism to mediate their biological toxicities.
Collapse
Affiliation(s)
- Jun Ling
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- * E-mail:
| | - Zenaida P. Lopez-Dee
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
| | - Colby Cottell
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- Department of Sciences, Marywood University, Scranton, Pennsylvania, United States of America
| | - Laura Wolfe
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- Department of Sciences, Marywood University, Scranton, Pennsylvania, United States of America
| | - Derek Nye
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| |
Collapse
|
28
|
Laurenzana EM, Coslo DM, Vigilar MV, Roman AM, Omiecinski CJ. Activation of the Constitutive Androstane Receptor by Monophthalates. Chem Res Toxicol 2016; 29:1651-1661. [PMID: 27551952 DOI: 10.1021/acs.chemrestox.6b00186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Humans in industrialized areas are continuously exposed to phthalate plasticizers, prompting concerns of their potential toxicities. Previous studies from our laboratory and others have shown that various phthalates activate several mammalian nuclear receptors, in particular the constitutive androstane receptor (CAR), the pregnane X receptor (PXR), and the peroxisomal proliferator-activated receptors (PPARs), although often at concentration levels of questionable relevance to human exposure. We discovered that di(2-ethylhexyl) phthalate (DEHP) and di-isononyl phthalate (DiNP), two of the highest volume production agents, were potent activators of human CAR2 (hCAR2), a unique human CAR splice variant and, to a lesser degree, human PXR (hPXR). These diphthalates undergo rapid metabolism in mammalian systems, initially to their major monophthalate derivatives MEHP and MiNP. Although MEHP and MiNP are reported activators of the rodent PPARs, with lower affinities for the corresponding human PPARs, it remains unclear whether these monophthalate metabolites activate hCAR2 or hPXR. In this investigation, we assessed the relative activation potential of selected monophthalates and other low molecular weight phthalates against hCAR, the most prominent hCAR splice variants, as well as hPXR and human PPAR. Using transactivation and mammalian two-hybrid protein interaction assays, we demonstrate that these substances indeed activate hCARs and hPXR but to varying degrees. MEHP and MiNP exhibit potent activation of hCAR2 and hPXR with higher affinities for these receptors than for the hPPARs. The rank order potency for MEHP and MiNP was hCAR2 > hPXR > hPPARs. Results from primary hepatocyte experiments also reflect the MEHP and MiNP upregulation of the respective human target genes. We conclude that both di- and monophthalates are potently selective hCAR2 activators and effective hPXR activators. These results implicate these targets as important mediators of selective phthalate effects in humans. The striking differential affinities for these compounds between human and rodent nuclear receptors further implies that biological results obtained from rodent models may be of only limited relevance for interpolating phthalate-mediated effects in humans.
Collapse
Affiliation(s)
- Elizabeth M Laurenzana
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University , 101 Life Sciences Building, University Park, Pennsylvania 16802, United States
| | - Denise M Coslo
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University , 101 Life Sciences Building, University Park, Pennsylvania 16802, United States
| | - M Veronica Vigilar
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University , 101 Life Sciences Building, University Park, Pennsylvania 16802, United States
| | - Anthony M Roman
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University , 101 Life Sciences Building, University Park, Pennsylvania 16802, United States
| | - Curtis J Omiecinski
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University , 101 Life Sciences Building, University Park, Pennsylvania 16802, United States
| |
Collapse
|
29
|
Martinez-Arguelles DB, Papadopoulos V. Prenatal phthalate exposure: epigenetic changes leading to lifelong impact on steroid formation. Andrology 2016; 4:573-84. [DOI: 10.1111/andr.12175] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/11/2016] [Accepted: 01/28/2016] [Indexed: 01/02/2023]
Affiliation(s)
- D. B. Martinez-Arguelles
- The Research Institute of the McGill University Health Centre; McGill University; Montreal QC Canada
- Department of Medicine; McGill University; Montreal QC Canada
| | - V. Papadopoulos
- The Research Institute of the McGill University Health Centre; McGill University; Montreal QC Canada
- Department of Medicine; McGill University; Montreal QC Canada
- Department of Biochemistry; McGill University; Montreal QC Canada
- Department of Pharmacology & Therapeutics; McGill University; Montreal Quebec Canada
| |
Collapse
|
30
|
Lv Z, Cheng J, Huang S, Zhang Y, Wu S, Qiu Y, Geng Y, Zhang Q, Huang G, Ma Q, Xie X, Zhou S, Wu T, Ke Y. DEHP induces obesity and hypothyroidism through both central and peripheral pathways in C3H/He mice. Obesity (Silver Spring) 2016; 24:368-78. [PMID: 26663559 DOI: 10.1002/oby.21359] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/01/2015] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Di(2-ethylhexyl) phthalate (DEHP) is reported to cause obesity and hypothyroidism in both humans and rodents, but the underlying mechanisms were largely unknown. This study was designed to clarify the effects and the mechanisms of DEHP on the pathogenesis of obesity and hypothyroidism and to discover the relationship between them. METHODS Male C3H/He mice were treated with DEHP for 5 weeks, and the body weight, food intake, and body temperature were recorded during the exposure. After exposure, key organs and serum were analyzed by Q-PCR, Western blot, and ELISA. RESULTS DEHP induced significant body weight gain and adipogenesis in all exposure groups except for 0.05 mg/kg. Marked hyperphagia and daytime hypothermia were also observed, which were accompanied by disturbed hypothalamic neuropeptide expression and reduced BAT UCP1 expression. In addition, WAT lipid metabolism was significantly deceased at low dose (0.5 mg/kg) and increased at high dose (50 and 200 mg/kg). DEHP also induced hypothyroidism, which was probably attributed to the combined effects of hepatic CAR activation and hypothalamic TRH inhibition induced by hypothalamic leptin resistance. CONCLUSIONS Chronic DEHP exposure could induce obesity by interrupting energy homeostasis, which is probably due to the synergistic effects of hypothyroidism and hypothalamic leptin resistance.
Collapse
Affiliation(s)
- Ziquan Lv
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinquan Cheng
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Suli Huang
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanwei Zhang
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Shuang Wu
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yangshen Qiu
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yijie Geng
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Qian Zhang
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Guanqin Huang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Quan Ma
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xing Xie
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Shiquan Zhou
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Tangchun Wu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuebin Ke
- Key Laboratory of Genetics & Molecular Medicine of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
31
|
A Systems Biology Approach for Identifying Hepatotoxicant Groups Based on Similarity in Mechanisms of Action and Chemical Structure. Methods Mol Biol 2016; 1425:339-59. [PMID: 27311473 DOI: 10.1007/978-1-4939-3609-0_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
When evaluating compound similarity, addressing multiple sources of information to reach conclusions about common pharmaceutical and/or toxicological mechanisms of action is a crucial strategy. In this chapter, we describe a systems biology approach that incorporates analyses of hepatotoxicant data for 33 compounds from three different sources: a chemical structure similarity analysis based on the 3D Tanimoto coefficient, a chemical structure-based protein target prediction analysis, and a cross-study/cross-platform meta-analysis of in vitro and in vivo human and rat transcriptomics data derived from public resources (i.e., the diXa data warehouse). Hierarchical clustering of the outcome scores of the separate analyses did not result in a satisfactory grouping of compounds considering their known toxic mechanism as described in literature. However, a combined analysis of multiple data types may hypothetically compensate for missing or unreliable information in any of the single data types. We therefore performed an integrated clustering analysis of all three data sets using the R-based tool iClusterPlus. This indeed improved the grouping results. The compound clusters that were formed by means of iClusterPlus represent groups that show similar gene expression while simultaneously integrating a similarity in structure and protein targets, which corresponds much better with the known mechanism of action of these toxicants. Using an integrative systems biology approach may thus overcome the limitations of the separate analyses when grouping liver toxicants sharing a similar mechanism of toxicity.
Collapse
|
32
|
Lake AD, Wood CE, Bhat VS, Chorley BN, Carswell GK, Sey YM, Kenyon EM, Padnos B, Moore TM, Tennant AH, Schmid JE, George BJ, Ross DG, Hughes MF, Corton JC, Simmons JE, McQueen CA, Hester SD. Dose and Effect Thresholds for Early Key Events in a PPARα-Mediated Mode of Action. Toxicol Sci 2015; 149:312-25. [PMID: 26519955 DOI: 10.1093/toxsci/kfv236] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Current strategies for predicting adverse health outcomes of environmental chemicals are centered on early key events in toxicity pathways. However, quantitative relationships between early molecular changes in a given pathway and later health effects are often poorly defined. The goal of this study was to evaluate short-term key event indicators using qualitative and quantitative methods in an established pathway of mouse liver tumorigenesis mediated by peroxisome proliferator-activated receptor alpha (PPARα). Male B6C3F1 mice were exposed for 7 days to di (2-ethylhexyl) phthalate (DEHP), di-n-octyl phthalate (DNOP), and n-butyl benzyl phthalate (BBP), which vary in PPARα activity and liver tumorigenicity. Each phthalate increased expression of select PPARα target genes at 7 days, while only DEHP significantly increased liver cell proliferation labeling index (LI). Transcriptional benchmark dose (BMDT) estimates for dose-related genomic markers stratified phthalates according to hypothetical tumorigenic potencies, unlike BMDs for non-genomic endpoints (relative liver weights or proliferation). The 7-day BMDT values for Acot1 as a surrogate measure for PPARα activation were 29, 370, and 676 mg/kg/day for DEHP, DNOP, and BBP, respectively, distinguishing DEHP (liver tumor BMD of 35 mg/kg/day) from non-tumorigenic DNOP and BBP. Effect thresholds were generated using linear regression of DEHP effects at 7 days and 2-year tumor incidence values to anchor early response molecular indicators and a later phenotypic outcome. Thresholds varied widely by marker, from 2-fold (Pdk4 and proliferation LI) to 30-fold (Acot1) induction to reach hypothetical tumorigenic expression levels. These findings highlight key issues in defining thresholds for biological adversity based on molecular changes.
Collapse
Affiliation(s)
- April D Lake
- *Curriculum in Toxicology, University of North Carolina, Chapel Hill, North Carolina 27599; Oak Ridge Institute for Science and Education (ORISE) participant at the National Health and Environmental Effects Research Laboratory (NHEERL), Office of Research and Development (ORD), U.S. Environmental Protection Agency (U.S. EPA), Research Triangle Park, North Carolina 27711; Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Charles E Wood
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | | | - Brian N Chorley
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Gleta K Carswell
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Yusupha M Sey
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Elaina M Kenyon
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Beth Padnos
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Tanya M Moore
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Alan H Tennant
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | | | - Barbara Jane George
- Office of the Associate Director for Health, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - David G Ross
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Michael F Hughes
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - J Christopher Corton
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Jane Ellen Simmons
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Charlene A McQueen
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711
| | - Susan D Hester
- Integrated Systems Toxicology Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina 27711;
| |
Collapse
|
33
|
Mathieu-Denoncourt J, Wallace SJ, de Solla SR, Langlois VS. Plasticizer endocrine disruption: Highlighting developmental and reproductive effects in mammals and non-mammalian aquatic species. Gen Comp Endocrinol 2015; 219:74-88. [PMID: 25448254 DOI: 10.1016/j.ygcen.2014.11.003] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 10/24/2022]
Abstract
Due to their versatility, robustness, and low production costs, plastics are used in a wide variety of applications. Plasticizers are mixed with polymers to increase flexibility of plastics. However, plasticizers are not covalently bound to plastics, and thus leach from products into the environment. Several studies have reported that two common plasticizers, bisphenol A (BPA) and phthalates, induce adverse health effects in vertebrates; however few studies have addressed their toxicity to non-mammalian species. The aim of this review is to compare the effects of plasticizers in animals, with a focus on aquatic species. In summary, we identified three main chains of events that occur in animals exposed to BPA and phthalates. Firstly, plasticizers affect development by altering both the thyroid hormone and growth hormone axes. Secondly, these chemicals interfere with reproduction by decreasing cholesterol transport through the mitochondrial membrane, leading to reduced steroidogenesis. Lastly, exposure to plasticizers leads to the activation of peroxisome proliferator-activated receptors, the increase of fatty acid oxidation, and the reduction in the ability to cope with the augmented oxidative stress leading to reproductive organ malformations, reproductive defects, and decreased fertility.
Collapse
Affiliation(s)
- Justine Mathieu-Denoncourt
- Chemistry and Chemical Engineering Department, Royal Military College of Canada, Kingston, ON K7K 7B4, Canada
| | - Sarah J Wallace
- Chemistry and Chemical Engineering Department, Royal Military College of Canada, Kingston, ON K7K 7B4, Canada
| | - Shane R de Solla
- Wildlife and Landscape Science Directorate, Environment Canada, Burlington, ON L7R 4A6, Canada
| | - Valerie S Langlois
- Chemistry and Chemical Engineering Department, Royal Military College of Canada, Kingston, ON K7K 7B4, Canada.
| |
Collapse
|
34
|
Yoo HS, Cichocki JA, Kim S, Venkatratnam A, Iwata Y, Kosyk O, Bodnar W, Sweet S, Knap A, Wade T, Campbell J, Clewell HJ, Melnyk SB, Chiu WA, Rusyn I. The Contribution of Peroxisome Proliferator-Activated Receptor Alpha to the Relationship Between Toxicokinetics and Toxicodynamics of Trichloroethylene. Toxicol Sci 2015; 147:339-49. [PMID: 26136231 DOI: 10.1093/toxsci/kfv134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Exposure to the ubiquitous environmental contaminant trichloroethylene (TCE) is associated with cancer and non-cancer toxicity in both humans and rodents. Peroxisome proliferator-activated receptor-alpha (PPARα) is thought to be playing a role in liver toxicity in rodents through activation of the receptor by the TCE metabolite trichloroacetic acid (TCA). However, most studies using genetically altered mice have not assessed the potential for PPARα to alter TCE toxicokinetics, which may lead to differences in TCA internal doses and hence confound inferences as to the role of PPARα in TCE toxicity. To address this gap, male and female wild type (129S1/SvImJ), Pparα-null, and humanized PPARα (hPPARα) mice were exposed intragastrically to 400 mg/kg TCE in single-dose (2, 5 and 12 h) and repeat-dose (5 days/week, 4 weeks) studies. Interestingly, following either a single- or repeat-dose exposure to TCE, levels of TCA in liver and kidney were lower in Pparα-null and hPPARα mice as compared with those in wild type mice. Levels of trichloroethanol (TCOH) were similar in all strains. TCE-exposed male mice consistently had higher levels of TCA and TCOH in all tissues compared with females. Additionally, in both single- and repeat-dose studies, a similar degree of induction of PPARα-responsive genes was observed in liver and kidney of hPPARα and wild type mice, despite the difference in hepatic and renal TCA levels. Additional sex- and strain-dependent effects were observed in the liver, including hepatocyte proliferation and oxidative stress, which were not dependent on TCA or TCOH levels. These data demonstrate that PPARα status affects the levels of the putative PPARα agonist TCA following TCE exposure. Therefore, interpretations of studies using Pparα-null and hPPARα mice need to consider the potential contribution of genotype-dependent toxicokinetics to observed differences in toxicity, rather than attributing such differences only to receptor-mediated toxicodynamic effects.
Collapse
Affiliation(s)
- Hong Sik Yoo
- *Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina
| | - Joseph A Cichocki
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Sungkyoon Kim
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Abhishek Venkatratnam
- *Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Yasuhiro Iwata
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Oksana Kosyk
- *Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina
| | - Wanda Bodnar
- *Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina
| | - Stephen Sweet
- Geochemical and Environmental Research Group, Texas A&M University, College Station, Texas
| | - Anthony Knap
- Geochemical and Environmental Research Group, Texas A&M University, College Station, Texas
| | - Terry Wade
- Geochemical and Environmental Research Group, Texas A&M University, College Station, Texas
| | - Jerry Campbell
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina; and
| | - Harvey J Clewell
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina; and
| | - Stepan B Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA;
| |
Collapse
|
35
|
Zhang H, Zhang Z, Nakanishi T, Wan Y, Hiromori Y, Nagase H, Hu J. Structure-dependent activity of phthalate esters and phthalate monoesters binding to human constitutive androstane receptor. Chem Res Toxicol 2015; 28:1196-204. [PMID: 25938866 DOI: 10.1021/acs.chemrestox.5b00028] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The present study investigated the human constitutive androstane receptor (CAR) binding activities of 23 phthalate esters and 10 phthalate monoesters using a fast and sensitive human CAR yeast two-hybrid assay. Of 23 phthalate esters, 16 were evaluated as positive, and the 10% relative effective concentrations (REC10) ranged from 0.28 (BBP) to 29.51 μM (DEHP), whereas no obvious binding activities were found for the phthalate esters having alkyl chains more than six carbons in length. Of 10 phthalate monoesters, only monoethyl phthalate (MEP), monoisobutyl phthalate (MIBP), and mono-(2-ethyhexyl) tetrabromophthalate (TBMEHP) elicited human CAR binding activities. The REC10 values of MEP and MIBP were 4.27 and 14.13 μM, respectively, higher than those of their corresponding phthalate esters (1.45 μM for DEP and 0.83 μM for DIBP), whereas TBMEHP (0.66 μM) was much lower than TBHP (>10(2) μM). A molecular docking method was performed to simulate the interaction modes between phthalates and human CAR, and active phthalates were found to lie at almost the same site in the human CAR pocket. The docking results suggest that the strong binding of phthalates to human CAR arises primarily from hydrophobic interactions, π-π interactions, and steric effects and that weak hydrogen bonds and weak halogen bonds greatly contribute to the high binding activity of TBMEHP. In conclusion, the current study clarified that an extensive array of phthalates are activators of human CAR.
Collapse
Affiliation(s)
- Hong Zhang
- †MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Zhaobin Zhang
- †MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Tsuyoshi Nakanishi
- ‡Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu 501-1196, Japan
| | - Yi Wan
- †MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Youhei Hiromori
- ‡Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu 501-1196, Japan
| | - Hisamistu Nagase
- ‡Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu 501-1196, Japan
| | - Jianying Hu
- †MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
36
|
Oshida K, Vasani N, Jones C, Moore T, Hester S, Nesnow S, Auerbach S, Geter DR, Aleksunes LM, Thomas RS, Applegate D, Klaassen CD, Corton JC. Identification of chemical modulators of the constitutive activated receptor (CAR) in a gene expression compendium. NUCLEAR RECEPTOR SIGNALING 2015; 13:e002. [PMID: 25949234 PMCID: PMC4422105 DOI: 10.1621/nrs.13002] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/27/2015] [Indexed: 01/31/2023]
Abstract
The nuclear receptor family member constitutive activated receptor (CAR) is
activated by structurally diverse drugs and environmentally-relevant chemicals
leading to transcriptional regulation of genes involved in xenobiotic metabolism
and transport. Chronic activation of CAR increases liver cancer incidence in
rodents, whereas suppression of CAR can lead to steatosis and insulin
insensitivity. Here, analytical methods were developed to screen for chemical
treatments in a gene expression compendium that lead to alteration of CAR
activity. A gene expression biomarker signature of 83 CAR-dependent genes was
identified using microarray profiles from the livers of wild-type and CAR-null
mice after exposure to three structurally-diverse CAR activators (CITCO,
phenobarbital, TCPOBOP). A rank-based algorithm (Running Fisher’s
algorithm (p-value ≤ 10-4)) was used to evaluate the
similarity between the CAR biomarker signature and a test set of 28 and 32
comparisons positive or negative, respectively, for CAR activation; the test
resulted in a balanced accuracy of 97%. The biomarker signature was used to
identify chemicals that activate or suppress CAR in an annotated mouse
liver/primary hepatocyte gene expression database of ~1850 comparisons. CAR was
activated by 1) activators of the aryl hydrocarbon receptor (AhR) in wild-type
but not AhR-null mice, 2) pregnane X receptor (PXR) activators in wild-type and
to lesser extents in PXR-null mice, and 3) activators of PPARα in
wild-type and PPARα-null mice. CAR was consistently activated by five
conazole fungicides and four perfluorinated compounds. Comparison of effects in
wild-type and CAR-null mice showed that the fungicide propiconazole increased
liver weight and hepatocyte proliferation in a CAR-dependent manner, whereas the
perfluorinated compound perfluorooctanoic acid (PFOA) increased these endpoints
in a CAR-independent manner. A number of compounds suppressed CAR coincident
with increases in markers of inflammation including acetaminophen, concanavalin
A, lipopolysaccharide, and 300 nm silica particles. In conclusion, we have shown
that a CAR biomarker signature coupled with a rank-based similarity method
accurately predicts CAR activation. This analytical approach, when applied to a
gene expression compendium, increased the universe of known chemicals that
directly or indirectly activate CAR, highlighting the promiscuous nature of CAR
activation and signaling through activation of other xenobiotic-activated
receptors.
Collapse
Affiliation(s)
- Keiyu Oshida
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Naresh Vasani
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Carlton Jones
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Tanya Moore
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Susan Hester
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Stephen Nesnow
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Scott Auerbach
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - David R Geter
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Lauren M Aleksunes
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Russell S Thomas
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Dawn Applegate
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - Curtis D Klaassen
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| | - J Christopher Corton
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, (KO, NV, CJ, TM, SH, SN), NIEHS (SA) and Bayer CropScience (DRG), Research Triangle Park, NC 27711; Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ (LMA), The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709 (RST), RegeneMed, San Diego, CA (DA), Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA (CDK) and the Integrated Systems Toxicology Division, National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, NC 27711 (JCC)
| |
Collapse
|
37
|
Cocci P, Mosconi G, Arukwe A, Mozzicafreddo M, Angeletti M, Aretusi G, Palermo FA. Effects of Diisodecyl Phthalate on PPAR:RXR-Dependent Gene Expression Pathways in Sea Bream Hepatocytes. Chem Res Toxicol 2015; 28:935-47. [PMID: 25825955 DOI: 10.1021/tx500529x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Evidence that endocrine-disrupting chemicals (EDCs) may target metabolic disturbances, beyond interference with the functions of the endocrine systems has recently accumulated. Among EDCs, phthalate plasticizers like the diisodecyl phthalate (DiDP) are commonly found contaminants of aquatic environments and have been suggested to function as obesogens by activating peroxisome proliferator activated receptors (PPARs), a subset of nuclear receptors (NRs) that act as metabolic sensors, playing pivotal roles in lipid homeostasis. However, little is known about the modulation of PPAR signaling pathways by DiDP in fish. In this study, we have first investigated the ligand binding efficiency of DiDP to the ligand binding domains of PPARs and retinoid-X-receptor-α (RXRα) proteins in fish using a molecular docking approach. Furthermore, in silico predictions were integrated by in vitro experiments to show possible dose-relationship effects of DiDP on PPAR:RXR-dependent gene expression pathways using sea bream hepatocytes. We observed that DiDP shows high binding efficiency with piscine PPARs demonstrating a greater preference for RXRα. Our studies also demonstrated the coordinate increased expression of PPARs and RXRα, as well as their downstream target genes in vitro. Principal component analysis (PCA) showed the strength of relationship between transcription of most genes involved in fatty acid metabolism and PPAR mRNA levels. In particular, fatty acid binding protein (FABP) was highly correlated to all PPARs. The results of this study suggest that DiDP can be considered an environmental stressor that activates PPAR:RXR signaling to promote long-term changes in lipid homeostasis leading to potential deleterious physiological consequences in teleost fish.
Collapse
Affiliation(s)
- Paolo Cocci
- †School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino (MC), Italy
| | - Gilberto Mosconi
- †School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino (MC), Italy
| | - Augustine Arukwe
- ‡Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, 7491 Trondheim, Norway
| | - Matteo Mozzicafreddo
- †School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino (MC), Italy
| | - Mauro Angeletti
- †School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino (MC), Italy
| | - Graziano Aretusi
- §Controllo Statistico, Pescara, Italy.,⊥Marine Protected Area Torre del Cerrano, 64025 Pineto (TE), Italy
| | - Francesco Alessandro Palermo
- †School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino (MC), Italy
| |
Collapse
|
38
|
Lin Y, Min L, Huang Q, Chen Y, Fang C, Sun X, Dong S. The combined effects of DEHP and PCBs on phospholipase in the livers of mice. ENVIRONMENTAL TOXICOLOGY 2015; 30:197-204. [PMID: 23804495 DOI: 10.1002/tox.21885] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/24/2013] [Accepted: 05/27/2013] [Indexed: 06/02/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) and polychlorinated biphenyls (PCBs) are two widely distributed pollutants that are of great concern due to their adverse health effects. However, few studies have investigated the combined effects of DEHP and PCBs. In this study, adult mice were continuously exposed to mixtures of DEHP (15 mg/kg bodyweight/day) and Aroclor 1254 (7.5 mg/kg bodyweight/day) for 12 days to investigate the combined effects of these compounds. The results showed that the ratio of the liver weight to the body weight was higher in the treated group than that in the control group. The effects of combined exposure on three important receptors, the proliferator-activated receptor (PPAR), estrogen receptor (ER), and aryl hydrocarbon receptor (AHR), were investigated. The mRNA level of PPARγ was significantly up-regulated after exposure. The expression level of ERα was decreased in the male treated group. In contrast, the expression levels of AHR and related genes (cyp1a1 and cyp1b1) were not markedly affected. The expression level of phospholipase A (PLA) was significantly down-regulated at both the mRNA and protein levels in male mice after combined treatment. In all, our study demonstrated the combined effects of DEHP and PCBs on the expression levels of key receptors in mice. The combined exposure led to a decrease in phospholipase in male mice.
Collapse
Affiliation(s)
- Yi Lin
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
39
|
Derivation of an oral reference dose (RfD) for the plasticizer, di-(2-propylheptyl)phthalate (Palatinol® 10-P). Regul Toxicol Pharmacol 2014; 70:65-74. [DOI: 10.1016/j.yrtph.2014.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/30/2014] [Accepted: 06/01/2014] [Indexed: 11/19/2022]
|
40
|
LeBaron MJ, Rasoulpour RJ, Gollapudi BB, Sura R, Kan HL, Schisler MR, Pottenger LH, Papineni S, Eisenbrandt DL. Characterization of Nuclear Receptor-Mediated Murine Hepatocarcinogenesis of the Herbicide Pronamide and Its Human Relevance. Toxicol Sci 2014; 142:74-92. [DOI: 10.1093/toxsci/kfu155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
41
|
Ibrahim ZS, Ahmed MM, El-Shazly SA, Ishizuka M, Fujita S. Clofibric acid induces hepatic CYP 2B1/2 via constitutive androstane receptor not via peroxisome proliferator activated receptor alpha in rat. Biosci Biotechnol Biochem 2014; 78:1550-9. [PMID: 25052003 DOI: 10.1080/09168451.2014.923302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Peroxisome proliferator activated receptor α (PPARα) ligands, fibrates used to control hyperlipidemia. We demonstrated CYP2B induction by clofibric acid (CFA) however, the mechanism was not clear. In this study, HepG2 cells transfected with expression plasmid of mouse constitutive androstane receptor (CAR) or PPARα were treated with CFA, phenobarbital (PB) or TCPOBOP. Luciferase assays showed that CFA increased CYP2B1 transcription to the same level as PB, or TCPOBOP in HepG2 transfected with mouse CAR But failed to induce it in PPARα transfected cells. CYP2B expressions were increased with PB or CFA in Wistar female rats (having normal levels of CAR) but not in Wistar Kyoto female rats (having low levels of CAR). The induction of CYP2B by PB or CFA was comparable to nuclear CAR levels. CAR nuclear translocation was induced by CFA in both rat strains. This indicates that fibrates can activate CAR and that fibrates-insulin sensitization effect may occur through CAR, while hypolipidemic effect may operate through PPARα.
Collapse
Affiliation(s)
- Zein Shaban Ibrahim
- a Faculty of Veterinary Medicine, Department of Physiology , Kafrelsheikh University , Kafrelsheikh , Egypt
| | | | | | | | | |
Collapse
|
42
|
Wood CE, Jokinen MP, Johnson CL, Olson GR, Hester S, George M, Chorley BN, Carswell G, Carter JH, Wood CR, Bhat VS, Corton JC, DeAngelo AB. Comparative time course profiles of phthalate stereoisomers in mice. Toxicol Sci 2014; 139:21-34. [PMID: 24496636 DOI: 10.1093/toxsci/kfu025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
More efficient models are needed to assess potential carcinogenicity hazard of environmental chemicals based on early events in tumorigenesis. Here, we investigated time course profiles for key events in an established cancer mode of action. Using a case study approach, we evaluated two reference phthalates, di(2-ethylhexyl) phthalate (DEHP) and its stereoisomer di-n-octyl phthalate (DNOP), across the span of a two-year carcinogenicity bioassay. Male B6C3F1 mice received diets with no phthalate added (control), DEHP at 0.12, 0.60, or 1.20%, or DNOP at 0.10, 0.50, or 1.00% (n = 80-83/group) for up to 104 weeks with six interim evaluations starting at week 4. Mean phthalate doses were 139, 845, and 3147 mg/kg/day for DEHP and 113, 755, and 1281 mg/kg/day for DNOP groups, respectively. Incidence and number of hepatocellular tumors (adenoma and/or carcinoma) were greater at ≥ 60 weeks for all DEHP groups with time and dose trends, whereas DNOP had no significant effects. Key events supported a peroxisome proliferator-activated receptor alpha (PPARα) mode of action for DEHP, with secondary cytotoxicity at the high dose, whereas DNOP induced modest increases in PPARα activity without proliferative or cytotoxic effects. Threshold estimates for later tumorigenic effects were identified at week 4 for relative liver weight (+24%) and PPARα activity (+79%) relative to the control group. Benchmark doses (BMDs) for these measures at week 4 clearly distinguished DEHP and DNOP and showed strong concordance with values at later time points and tumorigenic BMDs. Other target sites included testis and kidney, which showed degenerative changes at higher doses of DEHP but not DNOP. Our results highlight marked differences in the chronic toxicity profiles of structurally similar phthalates and demonstrate quantitative relationships between early bioindicators and later tumor outcomes.
Collapse
Affiliation(s)
- Charles E Wood
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27709
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Corton JC, Cunningham ML, Hummer BT, Lau C, Meek B, Peters JM, Popp JA, Rhomberg L, Seed J, Klaunig JE. Mode of action framework analysis for receptor-mediated toxicity: The peroxisome proliferator-activated receptor alpha (PPARα) as a case study. Crit Rev Toxicol 2013; 44:1-49. [PMID: 24180432 DOI: 10.3109/10408444.2013.835784] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Several therapeutic agents and industrial chemicals induce liver tumors in rodents through the activation of the peroxisome proliferator-activated receptor alpha (PPARα). The cellular and molecular events by which PPARα activators induce rodent hepatocarcinogenesis has been extensively studied and elucidated. This review summarizes the weight of evidence relevant to the hypothesized mode of action (MOA) for PPARα activator-induced rodent hepatocarcinogenesis and identifies gaps in our knowledge of this MOA. Chemical-specific and mechanistic data support concordance of temporal and dose-response relationships for the key events associated with many PPARα activators including a phthalate ester plasticizer di(2-ethylhexyl) phthalate (DEHP) and the drug gemfibrozil. While biologically plausible in humans, the hypothesized key events in the rodent MOA, for PPARα activators, are unlikely to induce liver tumors in humans because of toxicodynamic and biological differences in responses. This conclusion is based on minimal or no effects observed on growth pathways, hepatocellular proliferation and liver tumors in humans and/or species (including hamsters, guinea pigs and cynomolgous monkeys) that are more appropriate human surrogates than mice and rats at overlapping dose levels. Overall, the panel concluded that significant quantitative differences in PPARα activator-induced effects related to liver cancer formation exist between rodents and humans. On the basis of these quantitative differences, most of the workgroup felt that the rodent MOA is "not relevant to humans" with the remaining members concluding that the MOA is "unlikely to be relevant to humans". The two groups differed in their level of confidence based on perceived limitations of the quantitative and mechanistic knowledge of the species differences, which for some panel members strongly supports but cannot preclude the absence of effects under unlikely exposure scenarios.
Collapse
|
44
|
Martinez-Arguelles DB, Campioli E, Culty M, Zirkin BR, Papadopoulos V. Fetal origin of endocrine dysfunction in the adult: the phthalate model. J Steroid Biochem Mol Biol 2013; 137:5-17. [PMID: 23333934 DOI: 10.1016/j.jsbmb.2013.01.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/27/2012] [Accepted: 01/07/2013] [Indexed: 11/16/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a plasticizer with endocrine disrupting properties that is found ubiquitously in the environment as well as in human amniotic fluid, umbilical cord blood, human milk, semen, and saliva. It is used in the industry to add flexibility to polyvinyl chloride-derived plastics and its wide spread use and presence has resulted in constant human exposure through fetal development and postnatal life. Epidemiological studies have suggested an association between phthalate exposures and human reproductive effects in infant and adult populations. The effects of fetal exposure to phthalates on the male reproductive system were unequivocally shown on animal models, principally rodents, in which short term deleterious reproductive effects are well established. By contrast, information on the long term effects of DEHP in utero exposure on gonadal function are scarce, while its potential effects on other organs are just starting to emerge. The present review focuses on these novel findings, which suggest that DEHP exerts more complex and broader disruptive effects on the endocrine system and metabolism than previously thought. This article is part of a Special Issue entitled "CSR 2013".
Collapse
Affiliation(s)
- D B Martinez-Arguelles
- The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
45
|
Paul KB, Thompson JT, Simmons SO, Vanden Heuvel JP, Crofton KM. Evidence for triclosan-induced activation of human and rodent xenobiotic nuclear receptors. Toxicol In Vitro 2013; 27:2049-60. [PMID: 23899473 DOI: 10.1016/j.tiv.2013.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/05/2013] [Accepted: 07/19/2013] [Indexed: 10/26/2022]
Abstract
The bacteriostat triclosan (2,4,4'-trichloro-2'-hydroxydiphenylether) (TCS) decreases rat serum thyroxine via putative nuclear receptor (NR) interaction(s) and subsequent transcriptional up-regulation of hepatic catabolism and clearance. However, due to the evolutionary divergence of the constitutive androstane and pregnane-X receptors (CAR, PXR), TCS-mediated downstream effects may be species-dependent. To test the hypothesis that TCS activates xenobiotic NRs across species, cell-based NR reporter assays were employed to assess potential activation of rat, mouse, and human PXR, and rat, mouse, and three splice variants of human CAR. TCS activated hPXR, acted as an inverse agonist of hCAR1, and as a weak agonist of hCAR3. TCS failed to activate rPXR in full-length receptor reporter assays, and instead acted as a modest inverse agonist of rCAR. Consistent with the rat data, TCS also failed to activate mPXR and was a modest inverse agonist of mCAR. These data suggest that TCS may interact with multiple NRs, including hPXR, hCAR1, hCAR3, and rCAR in order to potentially affect hepatic catabolism. Overall these data support the conclusion that TCS may interact with NRs to regulate hepatic catabolism and downstream thyroid hormone homeostasis in both rat and human models, though perhaps by divergent mechanisms.
Collapse
Affiliation(s)
- Katie B Paul
- University of North Carolina at Chapel Hill, Curriculum in Toxicology, CB 7270, Chapel Hill, NC 27599, United States; Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, United States
| | | | | | | | | |
Collapse
|
46
|
Carbone V, Velkov T. Interaction of Phthalates and Phenoxy Acid Herbicide Environmental Pollutants with Intestinal Intracellular Lipid Binding Proteins. Chem Res Toxicol 2013; 26:1240-50. [DOI: 10.1021/tx400170t] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Vincenzo Carbone
- Animal Nutrition and Health, AgResearch Limited, Grasslands Research Centre, Tennent
Drive, Private Bag 11008, Palmerston North 4442, New Zealand
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics,
Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville 3052, Victoria, Australia
| |
Collapse
|
47
|
Obesity and metabolic comorbidities: environmental diseases? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:640673. [PMID: 23577225 PMCID: PMC3613100 DOI: 10.1155/2013/640673] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/22/2013] [Accepted: 02/05/2013] [Indexed: 12/20/2022]
Abstract
Obesity and metabolic comorbidities represent increasing health problems. Endocrine disrupting compounds (EDCs) are exogenous agents that change endocrine function and cause adverse health effects. Most EDCs are synthetic chemicals; some are natural food components as phytoestrogens. People are exposed to complex mixtures of chemicals throughout their lives. EDCs impact hormone-dependent metabolic systems and brain function. Laboratory and human studies provide compelling evidence that human chemical contamination can play a role in obesity epidemic. Chemical exposures may increase the risk of obesity by altering the differentiation of adipocytes. EDCs can alter methylation patterns and normal epigenetic programming in cells. Oxidative stress may be induced by many of these chemicals, and accumulating evidence indicates that it plays important roles in the etiology of chronic diseases. The individual sensitivity to chemicals is variable, depending on environment and ability to metabolize hazardous chemicals. A number of genes, especially those representing antioxidant and detoxification pathways, have potential application as biomarkers of risk assessment. The potential health effects of combined exposures make the risk assessment process more complex compared to the assessment of single chemicals. Techniques and methods need to be further developed to fill data gaps and increase the knowledge on harmful exposure combinations.
Collapse
|
48
|
Caldwell JC. DEHP: Genotoxicity and potential carcinogenic mechanisms—A review. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2012; 751:82-157. [DOI: 10.1016/j.mrrev.2012.03.001] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 03/22/2012] [Accepted: 03/22/2012] [Indexed: 10/28/2022]
|
49
|
Posnack NG, Swift LM, Kay MW, Lee NH, Sarvazyan N. Phthalate exposure changes the metabolic profile of cardiac muscle cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:1243-51. [PMID: 22672789 PMCID: PMC3440133 DOI: 10.1289/ehp.1205056] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 06/06/2012] [Indexed: 05/18/2023]
Abstract
BACKGROUND Phthalates are common plasticizers present in medical-grade plastics and other everyday products. They can also act as endocrine-disrupting chemicals and have been linked to the rise in metabolic disorders. However, the effect of phthalates on cardiac metabolism remains largely unknown. OBJECTIVES We examined the effect of di(2-ethylhexyl)phthalate (DEHP) on the metabolic profile of cardiomyocytes because alterations in metabolic processes can lead to cell dysfunction. METHODS Neonatal rat cardiomyocytes were treated with DEHP at a concentration and duration comparable to clinical exposure (50-100 μg/mL, 72 hr). We assessed the effect of DEHP on gene expression using microarray analysis. Physiological responses were examined via fatty acid utilization, oxygen consumption, mitochondrial mass, and Western blot analysis. RESULTS Exposure to DEHP led to up-regulation of genes associated with fatty acid transport, esterification, mitochondrial import, and β-oxidation. The functional outcome was an increase in myocyte fatty acid-substrate utilization, oxygen consumption, mitochondrial mass, PPARα (peroxisome proliferator-activated receptor α) protein expression, and extracellular acidosis. Treatment with a PPARα agonist (Wy-14643) only partially mimicked the effects observed in DEHP-treated cells. CONCLUSIONS Data suggest that DEHP exposure results in metabolic remodeling of cardiomyocytes, whereby cardiac cells increase their dependence on fatty acids for energy production. This fuel switch may be regulated at both the gene expression and posttranscription levels. Our findings have important clinical implications because chronic dependence on fatty acids is associated with an accumulation in lipid intermediates, lactate, protons, and reactive oxygen species. This dependence can sensitize the heart to ischemic injury and ventricular dysfunction.
Collapse
Affiliation(s)
- Nikki Gillum Posnack
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA.
| | | | | | | | | |
Collapse
|
50
|
Plasticizers May Activate Human Hepatic Peroxisome Proliferator-Activated Receptor α Less Than That of a Mouse but May Activate Constitutive Androstane Receptor in Liver. PPAR Res 2012; 2012:201284. [PMID: 22792086 PMCID: PMC3388330 DOI: 10.1155/2012/201284] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 03/19/2012] [Accepted: 03/21/2012] [Indexed: 12/05/2022] Open
Abstract
Dibutylphthalate (DBP), di(2-ethylhexyl)phthalate (DEHP), and di(2-ethylhexyl)adipate (DEHA) are used as plasticizers. Their metabolites activate peroxisome proliferator-activated receptor (PPAR) α, which may be related to their toxicities. However, species differences in the receptor functions between rodents and human make it difficult to precisely extrapolate their toxicity from animal studies to human. In this paper, we compared the species differences in the activation of mouse and human hepatic PPARα by these plasticizers using wild-type (mPPARα) and humanized PPARα (hPPARα) mice. At 12 weeks old, each genotyped male mouse was classified into three groups, and fed daily for 2 weeks per os with corn oil (vehicle control), 2.5 or 5.0 mmol/kg DBP (696, 1392 mg/kg), DEHP (977, 1953 mg/kg), and DEHA (926, 1853 mg/kg), respectively. Generally, hepatic PPARα of mPPARα mice was more strongly activated than that of hPPARα mice when several target genes involving β-oxidation of fatty acids were evaluated. Interestingly, all plasticizers also activated hepatic constitutive androstane receptor (CAR) more in hPPARα mice than in mPPARα mice. Taken together, these plasticizers activated mouse and human hepatic PPARα as well as CAR. The activation of PPARα was stronger in mPPARα mice than in hPPARα mice, while the opposite was true of CAR.
Collapse
|