1
|
Zhu Q, Xie X, Fang L, Huang C, Li J. Chronic alcohol intake disrupts cytochrome P450 enzyme activity in alcoholic fatty liver disease: insights into metabolic alterations and therapeutic targets. Front Chem 2025; 13:1509785. [PMID: 40433307 PMCID: PMC12106329 DOI: 10.3389/fchem.2025.1509785] [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: 10/11/2024] [Accepted: 01/08/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Alcoholic fatty liver disease (AFLD) is a common consequence of chronic alcohol consumption, characterized by lipid accumulation and oxidative stress in the liver. Cytochrome P450 (CYP450) enzymes play essential roles in metabolizing alcohol and other compounds. However, the specific long-term effects of alcohol on these enzymes remain unclear. Methods This study the examines influence of prolonged ethanol exposure on CYP450 activity and expression in AFLD using a rat model. Key enzymes such as CYP2E1, CYP2D6, and CYP3A1 were assessed in relation to lipid accumulation and oxidative stress. Results Significant alterations were identified in the expression and activity of CYP2E1, CYP2D6, and CYP3A1, which were associated with increased lipid accumulation and oxidative stress in the liver. Additionally, the expression of P-glycoprotein (P-gp) was elevated, suggesting that chronic alcohol intake may impact drug transport and excretion. Discussion These findings provide new insights into the molecular mechanisms of AFLD and highlight the potential of CYP450 modulation as a therapeutic target. By elucidating how long-term ethanol exposure disrupts hepatic CYP450 enzyme profiles, this research lays the groundwork for developing personalized therapeutic strategies to improve outcomes for patients with AFLD.
Collapse
Affiliation(s)
- Qian Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xuefeng Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ling Fang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Nielsen G, Gondim DD, Cave MC, Heiger-Bernays WJ, Webster TF, Schlezinger JJ. Perfluorooctanoic acid increases serum cholesterol in a PPARα-dependent manner in female mice. Arch Toxicol 2025; 99:2087-2105. [PMID: 40021516 DOI: 10.1007/s00204-025-03984-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a large group of persistent chemicals that are pervasive in the environment leading to widespread exposure for humans. Perfluorooctanoic acid (PFOA), one of the most commonly measured PFAS in people, disrupts liver and serum lipid homeostasis as shown in animal toxicity and human epidemiological studies. We tested the hypothesis that the effects of PFOA exposure in mice expressing mouse PPARα (mPPARα) are driven largely through PPARα-dependent mechanisms while non-PPARα dependent mechanisms will be more apparent in mice expressing human PPARα (hPPARα). Female and male mPPARα, hPPARα, and PPARα null mice were exposed to PFOA (0.5, 1.4 or 6.2 mg PFOA/L) via drinking water for 14 weeks. Concurrently, mice consumed an American diet containing human diet-relevant amounts of fat and cholesterol. Here, we focused on the effects in female mice, given the dearth of data reported on PFAS-induced effects in females. Increasing the duration of PFOA exposure reduced weight gain in all genotypes of female mice while end-of-study body fat was lower in PFOA exposed hPPARα and PPARα null mice. Serum cholesterol, but not triacylglyceride, concentrations were increased by PFOA exposure in a PPARα-dependent manner. Hepatic triacylglycerides were higher in vehicle-exposed mPPARα and PPARα null mice than hPPARα mice, and PFOA significantly reduced concentrations in mPPARα and PPARα null mice only. In contrast, PFOA increased hepatic cholesterol content in a PPARα-dependent manner. Changes in liver and serum cholesterol may be explained by a strong, PPARα-dependent downregulation of Cyp7a1 expression. PFOA significantly increased PPARα target gene expression in mPPARα mice. Other nuclear receptors were examined: CAR target gene expression was only induced by PFOA in hPPARα and PPARα null mice. PXR target gene expression was induced by PFOA in all genotypes. Results were similar in male mice with two exceptions: (1) vehicle-exposed male mice of all genotypes were equally susceptible to diet-induced hepatic steatosis; (2) male mice drank less water, resulting in lower serum PFOA levels, which may explain the less significant changes in lipid endpoints. Overall, our results show that PFOA modifies triacylglyceride and cholesterol homeostasis independently and that PPARα plays an important role in PFOA-induced increases in liver and serum cholesterol.
Collapse
Affiliation(s)
- G Nielsen
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA
| | - D D Gondim
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY, USA
| | - M C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY, USA
| | - W J Heiger-Bernays
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA
| | - T F Webster
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA
| | - J J Schlezinger
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA.
| |
Collapse
|
3
|
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
|
4
|
Zhou X, Hu F, Chen Y, Xie K, Hong WJ, Li M, Guo LH. Insights into toxicological mechanisms of per-/polyfluoroalkyl substances by using omics-centered approaches. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125634. [PMID: 39755359 DOI: 10.1016/j.envpol.2025.125634] [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: 11/14/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
The extensive presence of per-/polyfluoroalkyl substances (PFASs) in the environment and their adverse effects on organisms have garnered increasing concern. With the shift of industrial development from legacy to emerging PFASs, expanding the understanding of molecular responses to legacy and emerging PFASs is essential to accurately assess their risks to organisms. Compared with traditional toxicological approaches, omics technologies including transcriptomics, proteomics, metabolomics/lipidomics, and microbiomics allow comprehensive analysis of the molecular changes that occur in organisms after PFAS exposure. This paper comprehensively reviews the insights of omics approaches, especially the multi-omics approach, on the toxic mechanisms of both legacy and emerging PFASs in recent five years, focusing on hepatotoxicity, developmental toxicity, immunotoxicity, reproductive toxicity, neurotoxicity, and the endocrine-disrupting effect. PFASs exert various toxic effects via lipid and amino acid metabolism disruption, perturbations in several cell signal pathways, and binding to nuclear receptors. Notably, integrating multi-omics offers a thorough insight into the mechanisms of toxicity associated with PFASs. The gut microbiota plays an essential regulatory role in the toxic mechanisms of PFAS-induced hepatotoxicity. Finally, further research directions for PFAS toxicology based on omics technologies are prospected.
Collapse
Affiliation(s)
- Xinyi Zhou
- College of Energy Environment and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang, 310016, China
| | - Fanglin Hu
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, 310016, China
| | - Yafang Chen
- College of Energy Environment and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang, 310016, China
| | - Kun Xie
- College of Energy Environment and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang, 310016, China
| | - Wen-Jun Hong
- College of Energy Environment and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang, 310016, China
| | - Minjie Li
- College of Energy Environment and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang, 310016, China.
| | - Liang-Hong Guo
- College of Energy Environment and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang, 310016, China; School of Environment, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China.
| |
Collapse
|
5
|
Murase W, Kubota A, Hakota R, Yasuda A, Ikeda A, Nakagawa K, Shizu R, Yoshinari K, Kojima H. Comparative study on gene expression profiles in the liver of male neonatal mice prenatally exposed to PFOA and its alternative HFPO-DA. Toxicology 2025; 511:154048. [PMID: 39778857 DOI: 10.1016/j.tox.2025.154048] [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: 10/19/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Hexafluoropropylene oxide dimer acid (HFPO-DA), which belongs to the class of perfluoroalkyl ether carboxylic acid (PFECA), is a new alternative to perfluorooctanoic acid (PFOA). However, whether HFPO-DA is a safer alternative to PFOA in neonates remains unclear. In this study, we evaluated neonatal hepatic toxicity on postnatal days 9-10 by orally exposing pregnant CD-1 mice to 0.3 or 3.0 mg/kg/day (low or high doses) of HFPO-DA or PFOA from gestation days 15-17. The results showed that exposure of pregnant mice to HFPO-DA and PFOA induced similar phenotypic effects, including significant decreases in neonatal body weight (BW) and significant increases in liver weight relative to BW in the high-dose. Notably, HFPO-DA exposure significantly decreased in neonatal BW in the low-dose group, whereas PFOA did not. Comprehensive gene expression analysis revealed significant alterations in 408 and 1402 differentially expressed genes (DEGs) in the liver of neonates from the low- and high-dose HFPO-DA groups, respectively, while PFOA significantly altered 0 and 292 DEGs in the corresponding groups. Gene set enrichment analysis indicated that the DEGs induced by HFPO-DA and PFOA were enriched in pathway related to "PPAR signaling", "fatty acid metabolism", and "biological oxidations". In addition, transactivation assays revealed that mouse (m)PPARα and mPPARγ activity of HFPO-DA exceeds that of PFOA and molecular docking simulations analysis predicted that the binding conformation differ between PFOA and HFPO-DA. Overall, our findings demonstrate that HFPO-DA consistently affected neonatal phenotypes, liver gene expression and the molecular initiating events involving PPARα/γ, at lower concentrations than PFOA.
Collapse
Affiliation(s)
- 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
| | - Ryo Hakota
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Ayaka Yasuda
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Atsuko Ikeda
- Faculty of Health Sciences, Hokkaido University, 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
| | - Koji Nakagawa
- 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
| | - Ryota Shizu
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kouichi Yoshinari
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, 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
|
6
|
Gao J, Li T, Guo W, Yan M, Liu J, Yao X, Lv M, Ding Y, Qin H, Wang M, Liu R, Liu J, Shi C, Shi J, Qu G, Jiang G. Arginine Metabolism Reprogramming in Perfluorooctanoic Acid (PFOA)-Induced Liver Injury. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:1506-1518. [PMID: 39792631 DOI: 10.1021/acs.est.4c07971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Perfluorooctanoic acid (PFOA) is a persistent pollutant that has gained worldwide attention, owing to its widespread presence in the environment. Previous studies have reported that PFOA upregulates lipid metabolism and is associated with liver injury in humans. However, when the fatty acid degradation pathway is activated, lipid accumulation still occurs, suggesting the presence of unknown pathways and mechanisms that remain to be elucidated. In this study, adult C57BL/6N mice were exposed to PFOA at 0.1, 1, and 10 mg/kg/day. Using integrated metabolomics and transcriptomics, it was uncovered that arginine metabolism was differentially downregulated in all three groups. In vitro studies confirmed the downregulation of arginine metabolism in MIHA cell lines treated with PFOA. Supplementation of arginine could effectively rescue liver injury and downregulate the chemokine levels caused by PFOA. This finding highlights the contribution of arginine metabolism in maintaining liver health following PFOA exposure and suggests potential mechanisms of metabolic and immune modulation.
Collapse
Affiliation(s)
- Jie Gao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Tiantian Li
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
- State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing 100048, China
| | - Wei Guo
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
- State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing 100048, China
| | - Meilin Yan
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
- State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing 100048, China
| | - Junran Liu
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
- State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing 100048, China
| | - Xiaolong Yao
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
- State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing 100048, China
| | - Meilin Lv
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Yun Ding
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong Province 266237, China
| | - Hua Qin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Minghao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Sino-Danish College, Sino-Danish Center for Education and Research, UCAS, Beijing 100190, P. R. China
| | - Runzeng Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong Province 266237, China
| | - Jun Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Chunzhen Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
- State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing 100048, China
| | - Jianbo Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| |
Collapse
|
7
|
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
|
8
|
Wang X, Lv Y, Qiang X, Liang S, Li R, Zhan J, Liu J. Perfluorooctanoic acid (PFOA) and its alternative perfluorobutanoic acid (PFBA) alter hepatic bile acid profiles via different pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175312. [PMID: 39122034 DOI: 10.1016/j.scitotenv.2024.175312] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/28/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
The disruption of per- and polyfluoroalkyl substances (PFASs) on bile acid (BA) homeostasis has raised public concerns, making the evaluation of their effects and underlying mechanisms a high priority. Although the use of perfluorooctanoic acid (PFOA) has been restricted, it remains a widespread legacy PFAS in the environment. Concurrently, the use of its prevalent short-chain alternative, perfluorobutanoic acid (PFBA), is increasing, yet the toxicity assessment of PFBA remains inadequate. In this study, C57BL/6N mice were exposed to PFOA and PFBA (0.4 or 10 mg/kg body weight) by gavage for 28 days. The results showed that both PFOA and PFBA significantly increased hepatic weight, although PFBA exhibited lower bioaccumulation than PFOA in the liver. Targeted metabolomics revealed that PFOA significantly decreased total BA levels and altered their composition. Conversely, PFBA, without significantly altering total BA levels, notably changed their composition, such as increasing the proportion of cholic acid. Further investigations using in vivo and in vitro assays suggested that PFOA inhibited the expression of Cyp7A1, a key BA synthetase, potentially via PPARα activation, thereby reducing BA levels. In contrast, PFBA enhanced Cyp7A1 expression, associated with the inhibition of intestinal Farnesoid X receptor-fibroblast growth factor 15 (FXR-FGF15) pathway. This study evaluated the differences in the BA-interfering effects of PFOA and PFBA and shed light on the potential mechanisms, which will provide new insights into the health risks of legacy PFASs and their alternatives.
Collapse
Affiliation(s)
- Xiaoyuan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yinchuan Lv
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Xin Qiang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Shumin Liang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Ruosi Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Jing Zhan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Jiaying Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
9
|
Xu X, Chen R, Lu L, Cheng J, He X, Pan H, Zhang M, Yi H, Tang S. Roles of NR1I3 and NR1H4 polymorphisms in the susceptibility to antituberculosis drug-induced liver injury in China: a case‒control study. Front Genet 2024; 15:1428319. [PMID: 39512799 PMCID: PMC11541836 DOI: 10.3389/fgene.2024.1428319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Objective The pathogenesis of antituberculosis drug-induced liver injury (AT-DILI) remains largely unknown. The current investigation aimed to determine the genetic contribution of the nuclear receptor subfamily 1 Group I member 3 (NR1I3) and nuclear receptor subfamily 1 Group H member 4 (NR1H4) genes to the risk of AT-DILI in the Chinese population. Methods A 1:4 matched case‒control study was conducted, and five single nucleotide polymorphisms (SNPs) in the NR1I3 and NR1H4 genes were detected and assessed. Utilizing a multivariate conditional logistic regression model, the effects of haplotype and genotype on the risk of AT-DILI were examined. Extended subgroup analysis was carried out based on sex. The distribution of the peak value of serum liver enzymes also compared among different genotypes. Results 224 AT-DILI cases and 896 controls were included in this study. No significant difference was observed in genotypes or haplotypes frequencies between AT-DILI cases and controls. However, comparisons of liver function indicators revealed significant differences in the peak values of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and total bilirubin (TBil) among patients with different genotypes of NR1H4 rs56163822 (GG vs. GT vs. TT, 27.1 U/L vs. 26.0 U/L vs. 23.0 U/L, p = 0.020; 34.0 U/L vs. 31.0 U/L vs. 30.6 U/L, p = 0.008; 15.5 μmol/L vs. 15.0 μmol/L vs. 13.7 μmol/L, p = 0.029, respectively), as well as in the peak values of ALT and AST among male patients with different genotypes of NR1H4 rs56163822 (29.0 U/L vs. 26.9 U/L vs. 22.6 U/L, p = 0.002; 34.0 U/L vs. 32.0 U/L vs. 30.5 U/L, p = 0.019, respectively). Conclusion Based on this 1:4 individual-matched case‒control study, the SNP rs56163822 in the NR1H4 gene may be linked to the susceptibility to AT-DILI in Chinese patients receiving anti-TB treatment. Further studies in larger varied populations are needed to validate our findings.
Collapse
Affiliation(s)
- Xiaoyan Xu
- Department of Infectious Disease Control and Prevention, Changshu Center for Disease Control and Prevention, Suzhou, China
| | - Ruina Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lihuan Lu
- Department of Tuberculosis, The Second People’s Hospital of Changshu, Changshu, China
| | - Jingru Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaomin He
- Department of Infectious Disease, The People’s Hospital of Taixing, Taixing, China
| | - Hongqiu Pan
- Department of Tuberculosis, The Third People’s Hospital of Zhenjiang Affiliated to Jiangsu University, Zhenjiang, China
| | - Meiling Zhang
- Department of Infectious Disease, The Jurong Hospital Affiliated to Jiangsu University, Jurong, China
| | - Honggang Yi
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shaowen Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Liu X, Chen R, Peng Y, Zhou Y, Xia M, Wu X, Wang Y, Yin W, Han Y, Yu M. Perfluorooctanoic acid (PFOA) induces cardiotoxicity by activating the Keap1/Nrf2 pathway in zebrafish (Danio rerio) embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117098. [PMID: 39366304 DOI: 10.1016/j.ecoenv.2024.117098] [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: 05/23/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 10/06/2024]
Abstract
Perfluorooctanoic acid (PFOA), a perfluoroalkyl compound, is linked to congenital heart diseases, though its underlying mechanisms remain unclear. We hypothesized that PFOA induces cardiac defects through the inhibition of the Keap1/Nrf2 pathway, leading to oxidative damage in cardiomyocytes. In this study, zebrafish embryos exposed to PFOA showed significant cardiac malformations and dysfunction, characterized by excessive reactive oxygen species (ROS), malondialdehyde (MDA) production, decreased superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) activities. Additionally, we observed dysregulation in the expression of key cardiac development genes (vmhc, gata4, nkx2.5, and sox9b). PFOA also reduced the expression of keap1, nrf2, and ho-1. After overexpression of Nrf2, levels of ROS and MDA decreased, while levels of SOD, CAT, and GSH-Px increased. Additionally, cardiomyocyte apoptosis and cardiac malformations were alleviated. These findings have suggested that PFOA induces oxidative stress through Keap1/Nrf2 pathway inhibition, ultimately leading to cardiac defects.
Collapse
Affiliation(s)
- Xing Liu
- School of public health, Yangzhou University, Yangzhou 225009, China.
| | - Ruobing Chen
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Yuting Peng
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Yueyue Zhou
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Mingzhu Xia
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Xinyi Wu
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Yuchi Wang
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Wenjiao Yin
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Yuyang Han
- School of public health, Yangzhou University, Yangzhou 225009, China
| | - Meng Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Dauwe Y, Mary L, Oliviero F, Dubois L, Rousseau-Bacquie E, Gomez J, Gayrard V, Mselli-Lakhal L. Synergistic Steatosis Induction in Mice: Exploring the Interactions and Underlying Mechanisms between PFOA and Tributyltin. Cells 2024; 13:940. [PMID: 38891072 PMCID: PMC11171786 DOI: 10.3390/cells13110940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially characterized by steatosis and potentially progresses to steatohepatitis and fibrosis. Epidemiological studies have linked NAFLD occurrence to the exposure to environmental contaminants like PFAS. This study aims to assess the simultaneous activation of nuclear receptors via perfluorooctanoic acid (PFOA) and RXR coactivation via Tributyltin (TBT), examining their combined effects on steatogenic mechanisms. Mice were exposed to PFOA (10 mg/kg/day), TBT (5 mg/kg/day) or a combination of them for three days. Mechanisms underlying hepatic steatosis were explored by measuring nuclear receptor target gene and lipid metabolism key gene expressions, by quantifying plasma lipids and hepatic damage markers. This study elucidated the involvement of the Liver X Receptor (LXR) in the combined effect on steatosis and highlighted the permissive nature of the LXR/RXR heterodimer. Antagonistic effects of TBT on the PFOA-induced activation of the Pregnane X Receptor (PXR) and Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) were also observed. Overall, this study revealed complex interactions between PFOA and TBT, shedding light on their combined impact on liver health.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Laïla Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), Ecole Nationale Veterinaire de Toulouse (ENVT), INP-Purpan, Université Paul Sabatier (UPS), 31027 Toulouse, France; (Y.D.); (L.M.); (F.O.); (L.D.); (E.R.-B.); (J.G.); (V.G.)
| |
Collapse
|
12
|
Li R, Zhang Z, Xuan Y, Wang Y, Zhong Y, Zhang L, Zhang J, Chen Q, Yu S, Yuan J. HNF4A as a potential target of PFOA and PFOS leading to hepatic steatosis: Integrated molecular docking, molecular dynamic and transcriptomic analyses. Chem Biol Interact 2024; 390:110867. [PMID: 38199259 DOI: 10.1016/j.cbi.2024.110867] [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: 11/20/2023] [Revised: 12/30/2023] [Accepted: 01/08/2024] [Indexed: 01/12/2024]
Abstract
Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) are indeed among the most well known and extensively studied Per- and polyfluoroalkyl substances (PFASs), and increasing evidence confirm their effects on human health, especially liver steatosis. Nonetheless, the molecular mechanisms of their initiation of hepatic steatosis is still elusive. Therefore, potential targets of PFOA/PFOS must be explored to ameliorate its adverse consequences. This research aims to investigate the molecular mechanisms of PFOA and PFOS-induced liver steatosis, with emphasis on identifying a potential target that links these PFASs to liver steatosis. The potential target that causes PFOA and PFOS-induced liver steatosis have been explored and determined based on molecular docking, molecular dynamics (MD) simulation, and transcriptomics analysis. In silico results show that PFOA/PFOS can form a stable binding conformation with HNF4A, and PFOA/PFOS may interact with HNF4A to affect the downstream conduction mechanism. Transcriptome data from PFOA/PFOS-induced human stem cell spheres showed that HNF4A was inhibited, suggesting that PFOA/PFOS may constrain its function. PFOS mainly down-regulated genes related to cholesterol synthesis while PFOA mainly up-regulated genes related to fatty acid β-oxidation. This study explored the toxicological mechanism of liver steatosis caused by PFOA/PFOS. These compounds might inhibit and down-regulate HNF4A, which is the molecular initiation events (MIE) that induces liver steatosis.
Collapse
Affiliation(s)
- Rui Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Zijing Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yuxin Xuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yulu Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yuyan Zhong
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Lingyin Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Jinrui Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Qian Chen
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shuling Yu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, Henan, 475004, PR China
| | - Jintao Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China.
| |
Collapse
|