1
|
De Battistis F, Djordjevic AB, Saso L, Mantovani A. Constitutive androstane receptor, liver pathophysiology and chemical contaminants: current evidence and perspectives. Front Endocrinol (Lausanne) 2025; 16:1472563. [PMID: 40255499 PMCID: PMC12005993 DOI: 10.3389/fendo.2025.1472563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/11/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction The Constitutive Androstane Receptor (CAR) (NR1I3), a pivotal member of the xenosensor family, plays a key role in the hepatic detoxification of xenobiotic and endobiotic chemicals through the induction of the expression of drug-metabolizing enzymes and transporters. CAR's involvement extends beyond detoxification, influencing gluconeogenesis, lipogenesis, bile acid regulation, and cellular processes such as proliferation, tissue regeneration, and carcinogenesis. This review explores CAR regulation by various factors, highlighting its role in mediating metabolic changes induced by environmental contaminants. Methods A literature search was conducted to identify all articles on the PubMed website in which the CAR-contaminant and CAR-hepatic steatosis relationship is analyzed in both in vitro and in vivo models. Results Numerous contaminants, such as perfluorooctanoic acid (PFOA), Zearalenone mycotoxin, PCB, triazole fungicide propiconazole can activate hepatic nuclear receptors contributing to the development of steatosis through increased de novo lipogenesis, decreased fatty acid oxidation, increased hepatic lipid uptake, and decreased gluconeogenesis. Indirect CAR activation pathways, particularly involving PFOA, are discussed in the context of PPARα-independent mechanisms leading to hepatotoxicity, including hepatocellular hypertrophy and necrosis, and their implications in nonalcoholic steatohepatitis (NASH) and nonalcoholic fatty liver disease (NAFLD). The prevalence of NAFLD, a significant component of metabolic syndrome, underscores the importance of understanding CAR's role in its pathogenesis. Conclusions Experimental and epidemiological data suggest that endocrine disruptors, especially pesticides, play a significant role in NAFLD's development and progression via CAR-regulated pathways. This review advocates for the inclusion of modern toxicological risk assessment tools, such as New Approach Methodologies (NAMs), Adverse Outcome Pathways (AOPs), and Integrated Approaches to Testing and Assessment (IATA), to elucidate CAR-mediated effects and enhance regulatory frameworks.
Collapse
Affiliation(s)
- Francesca De Battistis
- Department of Food Safety, Nutrition, and Veterinary Public Health, Italian National Institute of Health, Rome, Italy
| | - Aleksandra Buha Djordjevic
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, Rome, Italy
| | - Alberto Mantovani
- Italian National Food Safety Committee, Rome, Italy
- Study Centre KOS - Science, Art, Society, Rome, Italy
| |
Collapse
|
2
|
Rogers J, Buerger A, Heintz M, Palermo C, Haws L, Lea I. Evaluation of a hypothesized Sertoli cell-based adverse outcome pathway for effects of diisononyl phthalate on the developing testis. Curr Res Toxicol 2025; 8:100219. [PMID: 40123861 PMCID: PMC11930220 DOI: 10.1016/j.crtox.2025.100219] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 03/25/2025] Open
Abstract
Exposure of pregnant rats to some phthalates during the masculinization programming window (MPW) can lower fetal testis testosterone production and adversely affect development of the fetal male reproductive tract. Some of the effects in rats are androgen-dependent, while others also occur in mice without lower testosterone production. An adverse outcome pathway (AOP) network has been proposed for these developmental effects that includes both androgen-dependent and androgen-independent pathways, the latter of which includes a short list of putative molecular initiating events (MIEs) including peroxisome proliferator activated receptor (PPAR) activation, and effects on Sertoli cells in the developing testes as early key events (KEs) (PMID 34314370). Data from peer-reviewed literature, publicly cited toxicology reports, and EPA's Toxicity Forecaster (ToxCast) were evaluated in the context of this hypothesized Sertoli cell-based AOP and exposure to diisononyl phthalate (DINP). Each of the fifteen identified studies underwent a risk of bias (RoB) assessment, which revealed a high risk of bias for all but one study endpoint. In vitro evidence in kidney, liver, and fibroblast-like cell lines indicates that the DINP metabolites mono-isononyl phthalate (MINP) and mono-hydroxyisononyl phthalate (MHINP) activate PPARα/γ and that mouse PPARα/γ are more sensitive than human PPARα/γ. However, DINP did not activate PPARα-related genes in rat fetal testes at high maternal dosages (PMID 22112501), and it remains unknown whether PPARs are expressed in fetal Sertoli cells. Overall, there is insufficient evidence to evaluate whether PPAR activation in the developing male reproductive tract is causally linked to the KEs in the hypothesized AOP. Regarding the KEs, no in vivo studies were identified that examined the effects of DINP on Sertoli cell proliferation or cytoskeleton; a single in vitro study found no effect of DINP on Sertoli cell proliferation. There was limited and conflicting evidence for the effects of DINP on tubulogenesis, but strong in vivo evidence for increased multinucleated germ (MNG) cells. No evidence was found concerning germ cell apoptosis. For the adverse outcomes (AOs), there was limited in vivo evidence for testicular dysgenesis following altered tubulogenesis, and impaired spermatogenesis following increased MNGs. There was strong evidence against reduced fertility, but this is not a sensitive endpoint in rats given their robust sperm production and excess capacity. In conclusion, following in utero DINP exposure, while PPAR activation (MIE) is plausible, linkage to effects on Sertoli cells and downstream AOPs is lacking. The sparse evidence currently available is insufficient to support the applicability of the hypothesized Sertoli cell-based AOP to DINP.
Collapse
Affiliation(s)
- J.M. Rogers
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, United States
| | - A.N. Buerger
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, United States
| | - M.M. Heintz
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, United States
| | - C.M. Palermo
- ExxonMobil Biomedical Sciences, Inc., Health and Environmental Sciences Division, Annandale, NJ, United States
| | - L.C. Haws
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, United States
| | - I.A. Lea
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, United States
| |
Collapse
|
3
|
Lea IA, Buerger AN, Feifarek D, Mihalchik A, Heintz MM, Haws LC, Nyambego H, Goyak K, Palermo C, Borghoff SJ. Evaluation of the endocrine disrupting potential of Di-isononyl phthalate. Curr Res Toxicol 2025; 8:100220. [PMID: 40092461 PMCID: PMC11910676 DOI: 10.1016/j.crtox.2025.100220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/13/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Low molecular weight ortho-phthalate compounds have been implicated in disruption of androgen pathways when exposure occurs during the masculinization programming window. Di-isononyl phthalate (DINP) is a high molecular weight phthalate and a high production volume chemical. To understand the potential for DINP and its metabolites to disrupt endocrine pathways, a weight of evidence assessment was conducted according to the European Chemicals Agency (ECHA)/ European Food Safety Authority (EFSA) Endocrine Disruptor Guidance (2018). Toxicological data related to estrogen (E), androgen (A), thyroid (T), or steroidogenesis (S) pathways was assessed. Literature searches returned 110 articles from which data were extracted and assessed in conjunction with 105 high-throughput assays. An in-silico assessment of the EATS activity for DINP metabolites also was conducted. Based on the available evidence, DINP did not elicit thyroid- or estrogen-related apical outcomes in vivo. There were no studies evaluating thyroid hormone levels in vivo which, according to the ECHA/EFSA guidance, constitutes a data gap and prevents a conclusion being drawn on the T-pathway. The E, A, and S-pathways were sufficiently assessed to conclude on the endocrine disrupting potential of DINP. Based on the lack of apical outcomes, DINP did not disrupt the E-pathway. For the A and S-pathways, there was limited evidence to support adverse apical outcomes, so a mode of action assessment using a structured adverse outcome pathway (AOP) framework was performed. No biologically plausible link could be established between the key events in the hypothesized AOP that lead to adverse outcomes. Further, no dose or temporal concordance for A- and S-mediated findings were identified. Therefore, DINP does not meet the ECHA/EFSA criteria to be considered an endocrine disruptor.
Collapse
Affiliation(s)
- I A Lea
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| | - A N Buerger
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| | - D Feifarek
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| | - A Mihalchik
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| | - M M Heintz
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| | - L C Haws
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| | - H Nyambego
- ExxonMobil Biomedical Sciences, Inc., Health and Environmental Sciences Division, Annandale, NJ, USA
| | - K Goyak
- ExxonMobil Biomedical Sciences, Inc., Health and Environmental Sciences Division, Annandale, NJ, USA
| | - C Palermo
- ExxonMobil Biomedical Sciences, Inc., Health and Environmental Sciences Division, Annandale, NJ, USA
| | - S J Borghoff
- ToxStrategies LLC, 31 College Place Suite B118, Asheville, NC 28801, USA
| |
Collapse
|
4
|
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
|
5
|
Wang X, Xu M, Shi M, Tian Y, Zhi Y, Han X, Sui H, Wan Y, Jia X, Yang H. Macrophage polarization as a novel endpoint for assessing combined risk of phthalate esters. ENVIRONMENT INTERNATIONAL 2024; 190:108835. [PMID: 38908276 DOI: 10.1016/j.envint.2024.108835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Combined exposure to phthalate esters (PAEs) has garnered increasing attention due to potential synergistic effects on human health. This study aimed to develop an in vitro model using human macrophages to evaluate the combined toxicity of PAEs and explore the underlying mechanisms. A high-throughput screening system was engineered by expressing a PPRE-eGFP reporter in THP-1 monocytes to monitor macrophage polarization upon PAEs exposure. Individual PAEs exhibited varied inhibitory effects on M2 macrophage polarization, with mono(2-ethylhexyl) phthalate (MEHP) being the most potent. Isobologram analysis revealed additive interactions when MEHP was combined with other PAEs, resulting in more pronounced suppression of M2 markers compared to individual compounds. Mechanistic studies suggested PAEs may exert effects by modulating PPARγ activity to inhibit M2 polarization. Notably, an equimolar mixture of six PAEs showed additive inhibition of M2 markers. In vivo experiments corroborated the combined hepatotoxic effects, with mice exposed to a PAEs mixture exhibiting reduced liver weight, dyslipidemia, and decreased hepatic M2 macrophages compared to DEHP alone. Transcriptome analysis highlighted disruptions in PPAR signaling, and distinct pathway alterations on cholesterol metabolism in the mixture group. Collectively, these findings underscore the importance of evaluating mixture effects and provide a novel approach for hazard assessment of combined PAEs exposure with implications for environmental health risk assessment.
Collapse
Affiliation(s)
- Xiaohong Wang
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China; Department of Nutrition and Food Safety, Peking Union Medical College, Research Unit of Food Safety, Chinese Academy of Medical Sciences, Beijing, China
| | - Miao Xu
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Sichuan Chengdu, China
| | - Miaoying Shi
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China; Department of Nutrition and Food Safety, Peking Union Medical College, Research Unit of Food Safety, Chinese Academy of Medical Sciences, Beijing, China
| | - Yaru Tian
- School of Public Health, Southern Medical University, Food Safety and Health Research Center, Guangdong Key Laboratory of Tropical Disease Research, Guangzhou, China; Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yuan Zhi
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Xiaomin Han
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Haixia Sui
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Yi Wan
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Xudong Jia
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Hui Yang
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China; Department of Nutrition and Food Safety, Peking Union Medical College, Research Unit of Food Safety, Chinese Academy of Medical Sciences, Beijing, China; School of Public Health, Southern Medical University, Food Safety and Health Research Center, Guangdong Key Laboratory of Tropical Disease Research, Guangzhou, China.
| |
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
|
Bernal K, Touma C, Erradhouani C, Boronat-Belda T, Gaillard L, Al Kassir S, Le Mentec H, Martin-Chouly C, Podechard N, Lagadic-Gossmann D, Langouet S, Brion F, Knoll-Gellida A, Babin PJ, Sovadinova I, Babica P, Andreau K, Barouki R, Vondracek J, Alonso-Magdalena P, Blanc E, Kim MJ, Coumoul X. Combinatorial pathway disruption is a powerful approach to delineate metabolic impacts of endocrine disruptors. FEBS Lett 2022; 596:3107-3123. [PMID: 35957500 DOI: 10.1002/1873-3468.14465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023]
Abstract
The prevalence of metabolic diseases, such as obesity, diabetes, metabolic syndrome and chronic liver diseases among others, has been rising for several years. Epidemiology and mechanistic (in vivo, in vitro and in silico) toxicology have recently provided compelling evidence implicating the chemical environment in the pathogenesis of these diseases. In this review, we will describe the biological processes that contribute to the development of metabolic diseases targeted by metabolic disruptors, and will propose an integrated pathophysiological vision of their effects on several organs. With regard to these pathomechanisms, we will discuss the needs, and the stakes of evolving the testing and assessment of endocrine disruptors to improve the prevention and management of metabolic diseases that have become a global epidemic since the end of last century.
Collapse
Affiliation(s)
- Kévin Bernal
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Charbel Touma
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Chedi Erradhouani
- Université Paris Cité, France.,Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte, France
| | - Talía Boronat-Belda
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Lucas Gaillard
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Sara Al Kassir
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Hélène Le Mentec
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Corinne Martin-Chouly
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Normand Podechard
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Dominique Lagadic-Gossmann
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Sophie Langouet
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - François Brion
- Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte, France
| | - Anja Knoll-Gellida
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Patrick J Babin
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Iva Sovadinova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karine Andreau
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Robert Barouki
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Jan Vondracek
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Paloma Alonso-Magdalena
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Etienne Blanc
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Min Ji Kim
- INSERM UMR-S 1124, Paris, France.,Université Sorbonne Paris Nord, Bobigny, France
| | - Xavier Coumoul
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| |
Collapse
|
9
|
Associations between maternal mono-(2-ethylhexyl) phthalate levels, nuclear receptor gene polymorphisms, and fatty acid levels in pregnant Japanese women in the Hokkaido study. Reprod Toxicol 2021; 107:22-32. [PMID: 34801790 DOI: 10.1016/j.reprotox.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 11/20/2022]
Abstract
We assessed how the interaction between mono-(2-ethylhexyl) phthalate (MEHP) in maternal sera and the maternal genotypes associated with nuclear receptors affect fatty acid levels in a prospective birth cohort study of pregnant Japanese individuals (n = 437) recruited in Sapporo between 2002 and 2005. We analyzed MEHP and fatty acids using gas chromatography-mass spectrometry. Thirteen single nucleotide polymorphisms of peroxisome proliferator-activated receptor (PPAR) alpha, PPAR gamma (PPARG), PPARG coactivator 1A (PPARGC1A), PPAR delta, constitutive androstane receptor, liver X receptor (LXR) alpha, and LXR beta (LXRB) were analyzed using real-time PCR. Multiple linear regression models were used to confirm the influence of log10-transformed MEHP levels and maternal genotypes on log10-transformed fatty acid levels. When the effects of the interaction between MEHP levels and the maternal PPARGC1A (rs8192678) genotype on oleic acid levels were evaluated, the estimated changes (95 % confidence intervals) in oleic acid levels against MEHP levels, maternal PPARGC1A (rs8192678)-GA/AA genotype, and the interaction between them showed a mean reduction of 0.200 (0.079, 0.322), mean reduction of 0.141 (0.000, 0.283), and mean increase of 0.145 (0.010, 0.281), respectively, after adjusting for the perfluorooctanesulfonate level. The effects of the interaction between MEHP levels and maternal LXRB (rs2303044) genotype on linoleic acid levels was also significant (pint = 0.010). In conclusion, the interaction between MEHP and the maternal genotypes PPARGC1A (rs8192678) and LXRB (rs2303044) decreased fatty acid levels. Further, the interaction between MEHP and PPARGC1A (rs8192678) may have a greater effect on fatty acid levels than the interaction between PFOS and PPARGC1A.
Collapse
|
10
|
Leng Y, Ren L, Niu S, Zhang T, Zhang J. In vitro and in silico investigations of endocrine disruption induced by metabolites of plasticizers through glucocorticoid receptor. Food Chem Toxicol 2021; 155:112413. [PMID: 34273429 DOI: 10.1016/j.fct.2021.112413] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023]
Abstract
The endocrine disruptive capability of plasticizers to activate nuclear receptors has attracted great interest. This study is aimed to assess the potential glucocorticoid effects of metabolites of plasticizers. The effects of metabolites of plasticizers on the transcriptional activity of glucocorticoid receptor (GR) were investigated using reporter gene assays. All of them failed to exhibit agonistic/antagonistic effects on GR. However, a combination of dexamethasone and monobutyl phthalate (MBP) could synergistically activate GR. MBP combined with dexamethasone also enhanced GR nuclear translocation by Western blot, while mifepristone restrained GR cytoplasmic-to-nuclear translocation. MBP co-treated with dexamethasone resulted in synergistic induction of PEPCK and MKP-1 gene expression by real-time PCR and PEPCK protein level by Western blot. Furthermore, the carboxyl and ester groups of MBP have influences on the charge distribution of MBP, leading to change of electrostatic interactions between MBP and GR by calculations on electronic properties. Both hydrophobic and hydrogen bonding interactions play a crucial role in the stabilization between MBP and GR conducted by molecular docking and dynamics simulation. This work confirms that GR could remain stable upon binding to MBP. In conclusion, dexamethasone and MBP could synergistically activate GR, resulting in synergetic enhancement of subsequent GR-mediated endocrine disrupting effect.
Collapse
Affiliation(s)
- Yue Leng
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Shu Niu
- College of New Energy and Environment, Jilin University, Changchun, 130021, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| |
Collapse
|
11
|
Mohammadi H, Ashari S. Mechanistic insight into toxicity of phthalates, the involved receptors, and the role of Nrf2, NF-κB, and PI3K/AKT signaling pathways. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:35488-35527. [PMID: 34024001 DOI: 10.1007/s11356-021-14466-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 05/14/2021] [Indexed: 06/12/2023]
Abstract
The wide use of phthalates, as phthalates are used in the manufacturing of not only plastics but also many others goods, has become a main concern in the current century because of their potency to induce deleterious effects on organism health. The toxic effects of phthalates such as reproductive toxicity, cardiotoxicity, hepatotoxicity, nephrotoxicity, teratogenicity, and tumor development have been widely indicated by previous experimental studies. Some of the important mechanisms of toxicity by phthalates are the induction and promotion of inflammation, oxidative stress, and apoptosis. Awareness of the involved molecular pathways of these mechanisms will permit the detection of exact molecular targets of phthalates to protect or treat their toxicity. Up to now, various transcription factors and signaling pathways have been associated with phthalate-induced toxicity which by influencing on nuclear surface and the expression of different genes can alter cell hemostasis. In different studies, the role of nuclear factor erythroid 2-related factor 2 (Nrf2), nuclear factor-κB (NF-κB), and phosphatidylinositol-3-kinase (PI3K)/AKT signaling pathways in processes of oxidative stress, inflammation, apoptosis, and cancer has been shown following exposure to phthalates. In the present review, we aim to survey experimental studies (in vitro and in vivo) in order to show firstly the most involved receptors and also the importance and the role of the mentioned signaling pathways in phthalate-induced toxicity, and with considering this point, the future studies can focus on these molecular targets as a strategic method to reduce environmental chemicals-induced toxicity especially phthalates toxic effects.
Collapse
Affiliation(s)
- Hamidreza Mohammadi
- Pharmaceutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Toxicology/Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sorour Ashari
- Department of Toxicology/Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
12
|
Oliviero F, Lukowicz C, Boussadia B, Forner-Piquer I, Pascussi JM, Marchi N, Mselli-Lakhal L. Constitutive Androstane Receptor: A Peripheral and a Neurovascular Stress or Environmental Sensor. Cells 2020; 9:E2426. [PMID: 33171992 PMCID: PMC7694609 DOI: 10.3390/cells9112426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
Xenobiotic nuclear receptors (NR) are intracellular players involved in an increasing number of physiological processes. Examined and characterized in peripheral organs where they govern metabolic, transport and detoxification mechanisms, accumulating data suggest a functional expression of specific NR at the neurovascular unit (NVU). Here, we focus on the Constitutive Androstane Receptor (CAR), expressed in detoxifying organs such as the liver, intestines and kidneys. By direct and indirect activation, CAR is implicated in hepatic detoxification of xenobiotics, environmental contaminants, and endogenous molecules (bilirubin, bile acids). Importantly, CAR participates in physiological stress adaptation responses, hormonal and energy homeostasis due to glucose and lipid sensing. We next analyze the emerging evidence supporting a role of CAR in NVU cells including the blood-brain barrier (BBB), a key vascular interface regulating communications between the brain and the periphery. We address the emerging concept of how CAR may regulate specific P450 cytochromes at the NVU and the associated relevance to brain diseases. A clear understanding of how CAR engages during pathological conditions could enable new mechanistic, and perhaps pharmacological, entry-points within a peripheral-brain axis.
Collapse
Affiliation(s)
- Fabiana Oliviero
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| | - Céline Lukowicz
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| | - Badreddine Boussadia
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Isabel Forner-Piquer
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Jean-Marc Pascussi
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Laila Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| |
Collapse
|
13
|
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
|
14
|
Wang JQ, Gao H, Sheng J, Tao XY, Huang K, Zhang YW, Mao LJ, Zhou SS, Jin ZX, Tao FB. Urinary concentrations of phthalate metabolites during gestation and intrahepatic cholestasis of pregnancy: a population-based birth cohort study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11714-11723. [PMID: 31975012 DOI: 10.1007/s11356-020-07675-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/07/2020] [Indexed: 06/10/2023]
Abstract
Phthalates, a class of widely used endocrine-disrupting chemicals (EDCs), are toxic to various organ systems in animals and humans. Intrahepatic cholestasis of pregnancy (ICP) is a reversible liver dysfunction causing cholestasis in late pregnancy. Evidence on the associations between exposure to phthalates and ICP is still lacking. In the present study, we investigated the relationships between urinary concentrations of phthalate metabolites and the risk of ICP in a Chinese population-based birth cohort. Pregnant women participated in the Ma'anshan Birth Cohort (MABC) study in China. Seven phthalate metabolites were detected in a urine sample in early pregnancy. Chemical concentrations were grouped by quartiles, and associations with outcomes were examined using logistic regression with adjustment for urine creatinine, race, education, poverty status, smoking status, alcohol consumption, maternal age, prepregnancy body mass index (BMI), parity, twin pregnancy, and pregnancy-related liver complications. Of 3474 women recruited into the Ma'anshan Birth Cohort, 2760 met the inclusion criteria and contributed to further analysis and biomonitoring data. Elevated odds ratios (ORs) of ICP were observed in the highest quartiles of monomethyl phthalate (MMP) exposure (OR = 1.59, 95% confidence intervals (CI) = 1.01-2.51) and monobutyl phthalate (MBP) exposure (OR = 1.82, 95% CI = 1.16-2.85) in the adjusted analyses. Our findings add to the evidence that supports the role of maternal phthalate exposure in the first trimester of gestation as a risk factor for ICP.
Collapse
Affiliation(s)
- Jian-Qing Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Hui Gao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jie Sheng
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xing-Yong Tao
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yun-Wei Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Lei-Jing Mao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shan-Shan Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhong-Xiu Jin
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
15
|
Toporova L, Balaguer P. Nuclear receptors are the major targets of endocrine disrupting chemicals. Mol Cell Endocrinol 2020; 502:110665. [PMID: 31760044 DOI: 10.1016/j.mce.2019.110665] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022]
Abstract
Endocrine disrupting chemicals (EDCs) are exogenous substances that are suspected to cause adverse effects in the endocrine system mainly by acting through their interaction with nuclear receptors such as the estrogen receptors α and β (ERα and ERβ), the androgen receptor (AR), the pregnan X receptor (PXR), the peroxisome proliferator activated receptors α and γ (PPARα, PPARγ) and the thyroid receptors α and β (TRα and TRβ). More recently, the retinoid X receptors (RXRα, RXRβ and RXRγ), the constitutive androstane receptor (CAR) and the estrogen related receptor γ (ERRγ) have also been identified as targets of EDCs. Finally, nuclear receptors still poorly studied for their interaction with environmental ligands such as the progesterone receptor (PR), the mineralocorticoid receptor (MR), the glucocorticoid receptor (GR), the retinoic acid receptors (RAR α, RARβ and RARγ), the farnesoid X receptor (FXR) and the liver X receptors α and β (LXRα and LXβ) as well are suspected targets of EDCs. Humans are generally exposed to low doses of pollutants, therefore the aim of current research is to identify the targets of EDCs at environmental concentrations. In this review, we analyze recent works referring that nuclear receptors are targets of EDCs and we highlight which EDCs are able to act at low concentrations.
Collapse
Affiliation(s)
- Lucia Toporova
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, ICM, Univ Montpellier, 34090, Montpellier, France.
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, ICM, Univ Montpellier, 34090, Montpellier, France.
| |
Collapse
|
16
|
Zhu Q, Hou J, Yin W, Ye F, Xu T, Cheng J, Yu Z, Wang L, Yuan J. Associations of a mixture of urinary phthalate metabolites with blood lipid traits: A repeated-measures pilot study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 257:113509. [PMID: 31767236 DOI: 10.1016/j.envpol.2019.113509] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/05/2019] [Accepted: 10/27/2019] [Indexed: 06/10/2023]
Abstract
Evidence is available about the associations of phthalates or their metabolites with blood lipids, however, the mixture effects of multiple phthalate metabolites on blood lipid traits remain largely unknown. In this pilot study, 106 individuals at three age groups of <18, 18- and ≥60 years were recruited from the residents (n = 1240) who were randomly selected from two communities in Wuhan city, China. The participants completed the questionnaire survey and physical examination as well as provided urine samples in the winter of 2014 and the summer of 2015. We measured urinary levels of nine phthalate metabolites using a high-performance liquid chromatography-tandem mass spectrometry. We estimated the associations of individual phthalate metabolite with blood lipid traits by linear mixed effect (LME) models, and assessed the overall association of the mixture of nine phthalate metabolites with blood lipid traits using Bayesian kernel machine regression (BKMR) models. LME models revealed the negative association of urinary mono-2-ethylhexyl phthalate (MEHP) with total cholesterol (TC) as well as of urinary mono-benzyl phthalate or urinary MEHP with low density lipoprotein cholesterol (LDL-C). BKMR models revealed the negative overall association of the mixture of nine phthalate metabolites with TC or LDL-C, and DEHP metabolites (especially MEHP) had a greater contribution to TC or LDL-C levels than non-DEHP metabolites. The findings indicated the negative overall association of the mixture of nine phthalate metabolites with TC or LDL-C. Among nine phthalate metabolites, MEHP was the most important component for the changes of TC or LDL-C levels, implying that phthalates exposure may disrupt lipid metabolism in the body.
Collapse
Affiliation(s)
- Qingqing Zhu
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Jian Hou
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Wenjun Yin
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Fang Ye
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Tian Xu
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Juan Cheng
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry, Guangdong Key Laboratory of Environment and Resources, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, PR China
| | - Lin Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China.
| | - Jing Yuan
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China.
| |
Collapse
|
17
|
Estill M, Hauser R, Nassan FL, Moss A, Krawetz SA. The effects of di-butyl phthalate exposure from medications on human sperm RNA among men. Sci Rep 2019; 9:12397. [PMID: 31455814 PMCID: PMC6711971 DOI: 10.1038/s41598-019-48441-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/05/2019] [Indexed: 02/08/2023] Open
Abstract
Endocrine disruptors, such as phthalates, are suspected of affecting reproductive function. The Mesalamine and Reproductive Health Study (MARS) was designed to address the physiological effect of in vivo phthalate exposure on male reproduction in patients with Inflammatory Bowel Disease (IBD). As part of this effort, the effect on sperm RNAs to DBP exposure were longitudinally assessed using a cross-over cross-back binary design of high or background, exposures to DBP. As the DBP level was altered, numerous sperm RNA elements (REs) were differentially expressed, suggesting that exposure to or removal from high DBP produces effects that require longer than one spermatogenic cycle to resolve. In comparison, small RNAs were minimally affected by DBP exposure. While initial study medication (high or background) implicates different biological pathways, initiation on the high-DBP condition activated oxidative stress and DNA damage pathways. The negative correlation of REs with specific genomic repeats suggests a regulatory role. Using ejaculated sperm, this work provides insight into the male germline's response to phthalate exposure.
Collapse
Affiliation(s)
- Molly Estill
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Russ Hauser
- Vincent Memorial Obstetrics and Gynecology Service, Massachusetts General Hospital, Harvard Medical School, and Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Feiby L Nassan
- Departments of Environmental Health and Nutrition, Harvard T. H. Chan School of Public Health, MA, 02115, USA
| | - Alan Moss
- Department of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | - Stephen A Krawetz
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
18
|
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
|
19
|
Lu X, Xu X, Lin Y, Zhang Y, Huo X. Phthalate exposure as a risk factor for hypertension. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:20550-20561. [PMID: 29862479 DOI: 10.1007/s11356-018-2367-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/22/2018] [Indexed: 02/05/2023]
Abstract
Phthalates are ubiquitous in environment. Hypertension is a major risk factor for cardiovascular diseases. Phthalate exposure is associated with hypertension in multiple studies. This review aims to summarize the scientific literature on associations between phthalate exposure and hypertension and discuss the mechanisms in the relationship. We identified and reviewed original articles published to March 2018, using PubMed and Web of Science to search the terms "phthalate(s)," "phthalic acid," "blood pressure," "high blood pressure," "hypertension," "prehypertension," and "cardiovascular disease." Findings were summarized based on the relevance to the themes, including presentation of main phthalates and their major metabolites as well as associations of phthalate exposure with blood pressure in epidemiological and experimental studies. We identified ten population-based investigations and five toxicological experiments. Epidemiological data underscored a possible correlation between phthalate exposure and hypertension in adults, whereas individual study in children stands on the opposite. Experimental studies mainly targeted the increasing effect of phthalates on blood pressure. This review suggested some underlying mechanisms of phthalate-associated hypertension. Considering the current evidence, phthalate might be risk factors of hypertension. However, the effect of phthalate exposure in early life on blood pressure in later life or adulthood is still unclear. Well-designed longitudinal and molecular mechanism studies are indispensable.
Collapse
Affiliation(s)
- Xueling Lu
- Laboratory of Environmental Medicine and Developmental Toxicology and Guangdong Provincial Key Laboratory of Infectious Diseases, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology and Guangdong Provincial Key Laboratory of Infectious Diseases, Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yucong Lin
- Tabor Academy, Marion, MA, USA
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511486, Guangdong, China
| | - Yu Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology and Guangdong Provincial Key Laboratory of Infectious Diseases, Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology and Guangdong Provincial Key Laboratory of Infectious Diseases, Shantou University Medical College, Shantou, 515041, Guangdong, China.
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511486, Guangdong, China.
| |
Collapse
|
20
|
Bogen KT. Biphasic hCAR Inhibition-Activation by Two Aminoazo Liver Carcinogens. NUCLEAR RECEPTOR RESEARCH 2018. [DOI: 10.11131/2018/101321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|