1
|
Sun L, He S, Chen J, Su A, Mao Q, Zhang W, Pan Y, Hu J, Feng D, Ouyang Y. Hepatic injury and metabolic perturbations in mice exposed to perfluorodecanoic acid revealed by metabolomics and lipidomics. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117475. [PMID: 39662454 DOI: 10.1016/j.ecoenv.2024.117475] [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: 07/05/2024] [Revised: 10/09/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Perfluorodecanoic acid (PFDA) is a typical perfluoroalkyl substances frequently encountered in populations, posing significant risks to human health. However, research on the effects of PFDA exposure on organism metabolism and related pathogenic mechanisms is severely lacking. In this study, serum and liver samples of C57BL/6 J mice exposed to different doses of PFDA were analyzed by UPLC-HRMS-based metabolomics and lipidomics techniques. Both 1 mg/kg and 10 mg/kg PFDA exposure induced liver damage, while only 10 mg/kg PFDA exposure caused weight loss. Metabolomics analysis revealed that 330 and 515 metabolites were significantly altered in the serum and liver of mice after PFDA exposure, respectively. Most amino acids and peptides increased in the serum but decreased in the liver. Lipidomics analysis indicated that 281 and 408 lipids experienced significant alterations in the serum and liver after PFDA exposure, respectively. Most lipids, particularly multiple triacylglycerols, were downregulated in a dose-dependent manner in both serum and liver. Taken together, PFDA can induce changes in the amino acid metabolism pathway, disrupt fatty acid β-oxidation, and down-regulate glycolipid pathways in mice, resulting in disturbances in energy metabolism. These findings suggested that the liver is a critical target organ for PFDA exposure, and will also help inform future risk assessment.
Collapse
Affiliation(s)
- Lvyun Sun
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Shuling He
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jiali Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Amei Su
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Qiuyao Mao
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Wenyuan Zhang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Ying Pan
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jiaqian Hu
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Disheng Feng
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Yang Ouyang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
2
|
Vujic E, Ferguson SS, Brouwer KLR. Effects of PFAS on human liver transporters: implications for health outcomes. Toxicol Sci 2024; 200:213-227. [PMID: 38724241 DOI: 10.1093/toxsci/kfae061] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) have become internationally recognized over the past three decades as persistent organic pollutants used in the production of various consumer and industrial goods. Research efforts continue to gauge the risk that historically used, and newly produced, PFAS may cause to human health. Numerous studies report toxic effects of PFAS on the human liver as well as increased serum cholesterol levels in adults. A major concern with PFAS, also dubbed "forever chemicals," is that they accumulate in the liver and kidney and persist in serum. The mechanisms responsible for their disposition and excretion in humans are poorly understood. A better understanding of the interaction of PFAS with liver transporters, as it pertains to the disposition of PFAS and other xenobiotics, could provide mechanistic insight into human health effects and guide efforts toward risk assessment of compounds in development. This review summarizes the current state of the literature on the emerging relationships (eg, substrates, inhibitors, modulators of gene expression) between PFAS and specific hepatic transporters. The adaptive and toxicological responses of hepatocytes to PFAS that reveal linkages to pathologies and epidemiological findings are highlighted. The evidence suggests that our understanding of the molecular landscape of PFAS must improve to determine their impact on the expression and function of hepatocyte transporters that play a key role in PFAS or other xenobiotic disposition. From here, we can assess what role these changes may have in documented human health outcomes.
Collapse
Affiliation(s)
- Ena Vujic
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Li S, Gao Z, Zhong M, Bi H, Li J. Identification of the mechanisms underlying per- and polyfluoroalkyl substance-induced hippocampal neurotoxicity as determined by network pharmacology and molecular docking analyses. Toxicol Res (Camb) 2023; 12:1126-1134. [PMID: 38145100 PMCID: PMC10734622 DOI: 10.1093/toxres/tfad104] [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: 06/06/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 12/26/2023] Open
Abstract
Background Per- and polyfluoroalkyl substances (PFASs) are a class of environmental contaminants that pose significant health risks to both animals and humans. Although the hippocampal neurotoxic effects of numerous PFASs have been reported, the underlying mechanisms of combined exposure to PFASs-induced hippocampal neurotoxicity remain unclear. Methods In this study, network pharmacology analysis was performed to identify the intersectional targets of PFASs for possible associations with hippocampal neurotoxicity. The evaluation of the influence of PFASs on intersectional targets was assessed using a weighted method. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the screened targets were performed, the intersected hub targets calculated by various algorithms were screened in the network and molecular docking was also used to analyze binding activities. Results Our results indicated that eight PFASs, which acted on key targets (MYC, ESR1, STAT3, RELA, MAPK3) impacted the NF-κB signaling pathway, STAT3 signaling pathway, and MAPK signaling pathways to exert neurotoxicity in the hippocampus. The molecular docking results revealed that PFASs have strong binding potential to the hub targets. Conclusions Our findings provided a basis for future studies to investigate the detailed mechanisms of PFASs-induced hippocampal neurotoxicity and to develop preventative and control strategies.
Collapse
Affiliation(s)
- Shirui Li
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Zhihui Gao
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Meihan Zhong
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Haoran Bi
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Jianan Li
- Department of Occupational and Environmental Health, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| |
Collapse
|
4
|
Xu H, Zhong X, Wang T, Wu S, Guan H, Wang D. (-)-Epigallocatechin-3-Gallate Reduces Perfluorodecanoic Acid-Exacerbated Adiposity and Hepatic Lipid Accumulation in High-Fat Diet-Fed Male C57BL/6J Mice. Molecules 2023; 28:7832. [PMID: 38067561 PMCID: PMC10708200 DOI: 10.3390/molecules28237832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Perfluorodecanoic acid (PFDA), an enduring and harmful organic pollutant, is widely employed in diverse food-related sectors. Our previous studies have provided evidence that PFDA has the potential to facilitate obesity and hepatic fat accumulation induced by high-fat diet (HFD) intake. Epigallocatechin-3-gallate (EGCG), a polyphenol found in green tea, has been suggested to possess potential preventive effects against metabolic abnormalities and fatty liver. The purpose of this research was to investigate the effects of EGCG on PFDA-exacerbated adiposity and hepatic lipid accumulation in HFD-fed mice. The results showed that EGCG reduced body weight gain; tissue and organ weights; blood glucose, serum insulin, HOMA-IR, leptin, and lipid parameters; serum inflammatory cytokines (IL-1β, IL-18, IL-6, and TNF-α); and hepatic lipid accumulation in PFDA-exposed mice fed an HFD. Further work showed that EGCG improved liver function and glucose homeostasis in mice fed an HFD and co-exposed to PFDA. The elevated hepatic mRNA levels of SREBP-1 and associated lipogenic genes, NLRP3, and caspase-1 in PFDA-exposed mice fed an HFD were significantly decreased by EGCG. Our work provides evidence for the potential anti-obesity effect of EGCG on co-exposure to HFD and PFDA and may call for further research on the bioactivity of EGCG to attenuate the endocrine disruption effects of long-term exposure to pollutants.
Collapse
Affiliation(s)
- Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| | - Xu Zhong
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| | - Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, China;
| | - Shanshan Wu
- College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China;
| | - Huanan Guan
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| |
Collapse
|
5
|
Yang L, Chen Y, Ji H, Zhang X, Zhou Y, Li J, Wang Y, Xie Z, Yuan W, Liang H, Miao M. Per- and Poly-fluoroalkyl Substances and Bile Acid Profiles in Pregnant Women. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:15869-15881. [PMID: 37821457 DOI: 10.1021/acs.est.3c05106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Alterations in bile acid (BA) profiles are closely associated with adverse outcomes in pregnant women and their offspring and may be one potential pathway underlying the related metabolic effects of per- and poly-fluoroalkyl substances (PFAS) exposure. However, evidence of associations between PFAS exposure and BA profiles in pregnant women is scarce. This study examined the associations of individual PFAS and PFAS mixture with BA profiles of pregnant women. We obtained quantitative data on the plasma concentrations of 13 PFAS and 15 BAs in 645 pregnant women from the Jiashan birth cohort. In Bayesian kernel machine regression models, the PFAS mixture was associated with increased plasma CA, TCA, TCDCA, and GLCA levels but with decreased GCA and LCA concentrations. Furthermore, the PFAS mixture was associated with increased concentrations of total BAs and the secondary/primary BA ratio but with decreased conjugated/unconjugated and glycine/taurine-conjugated BA ratios. PFHxS, PFUdA, PFOS, PFNA, and PFDA were the dominant contributors. The results of the linear regression analysis of individual PFAS were generally similar. Our findings provide the first epidemiological evidence for the associations of a PFAS mixture with BA profiles in pregnant women and may provide explanatory insights into the biological pathways underlying the related metabolic effects of PFAS exposure.
Collapse
Affiliation(s)
- Lan Yang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Yao Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Honglei Ji
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Xi Zhang
- Clinical Research Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200092, China
| | - Yan Zhou
- Hubei Provincial Key Laboratory of Applied Toxicology, National Reference Laboratory of Dioxin, Hubei Provincial Center for Disease Control and Prevention, Wuhan 430079, China
| | - Jianhui Li
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Hospital of SIPPR, Shanghai 200032, China
| | - Yan Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhenzhen Xie
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Wei Yuan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Hong Liang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| | - Maohua Miao
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai 200237, China
| |
Collapse
|
6
|
Ogunsuyi OM, Fasakin PT, Ajibiye OP, Ogunsuyi OI, Adekoya KO. Perfluoroundecanoic acid induces DNA damage, reproductive and pathophysiological dysfunctions via oxidative stress in male Swiss mice. CHEMOSPHERE 2023; 338:139491. [PMID: 37453524 DOI: 10.1016/j.chemosphere.2023.139491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Perfluoroundecanoic acid (PFUnA) is an eleven carbon-chain compound that belongs to the perfluoroalkyl carboxylic acid family. It has been detected in the human blood, effluents, and surface/ground waters, but its toxic effects to the DNA and reproductive system remain unclear. This study was aimed at exploring the toxicity of PFUnA on the hepatic DNA, organ-system and reproductive system in orally treated male Swiss mice. In this present study, administration of PFUnA for 28 days with five doses (0.1, 0.3, 05, 0.7 and 1.0 mg kg-1 b.w./d) in male Swiss mice induced significant hepatic DNA damage which was observed using the alkaline comet assay and equally altered hematological and clinical biochemical parameters. In addition to testicular atrophy, sperm count and sperm motility significantly decreased while sperm abnormalities increased after 35 days exposure. Serum LH and FSH levels were remarkably increased while serum testosterone levels were strikingly reduced. Histopathology revealed the liver, kidney, and testis as potential targets of PFUnA toxicity. Increased activities of superoxide dismutase (SOD) and catalase (CAT), as well as levels of glutathione-s-transferase (GST) and reduced glutathione (GSH), with consistent reduction of glutathione peroxidase (GPx) and reduced glutathione (GSH) in the liver and testis induced oxidative stress. In conclusion, PFUnA exhibited both genotoxicity and reproductive toxicity via oxidative stress induction.
Collapse
Affiliation(s)
- Opeoluwa M Ogunsuyi
- Department of Cell Biology and Genetics, Faculty of Science, University of Lagos, Akoka-Yaba, Lagos, Nigeria.
| | - Peter T Fasakin
- Department of Cell Biology and Genetics, Faculty of Science, University of Lagos, Akoka-Yaba, Lagos, Nigeria
| | - Oluwatobi P Ajibiye
- Department of Cell Biology and Genetics, Faculty of Science, University of Lagos, Akoka-Yaba, Lagos, Nigeria
| | - Olusegun I Ogunsuyi
- Department of Biological Science, College of Basic and Applied Sciences, Mountain Top University, Ibafo, Ogun State, Nigeria
| | - Khalid O Adekoya
- Department of Cell Biology and Genetics, Faculty of Science, University of Lagos, Akoka-Yaba, Lagos, Nigeria
| |
Collapse
|
7
|
Wang T, Xu H, Guo Y, Guo Y, Guan H, Wang D. Perfluorodecanoic acid promotes high-fat diet-triggered adiposity and hepatic lipid accumulation by modulating the NLRP3/caspase-1 pathway in male C57BL/6J mice. Food Chem Toxicol 2023; 178:113943. [PMID: 37451596 DOI: 10.1016/j.fct.2023.113943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/07/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Perfluorodecanoic acid (PFDA), a chemical contaminant, may casue became obesity, which makes it a public health concern. In this study, we investigated the effects of PFDA on adiposity development and hepatic lipid accumulation in mice fed with a high-fat diet (HFD). Animals were assigned to two diet treatments (low-fat and high-fat); and PFDA was administered through drinking water for 12 weeks. The contaminant promoted body weight gain and adiposity in HFD-fed mice. Moreover, HFD-fed mice exposed to PFDA had impaired glucose metabolism, inflammation and hepatic lipid accumulation compared to mice fed HFD alone. PFDA activated the expression of hepatic NLRP3 and caspase-1, and induced that of SREBP-1c expression in the liver of HFD-fed mice. PFDA exposure in HFD-fed mice significantly inhibited hepatic AMPK expression than animals fed HFD without PFDA exposure. Furthermore, MCC950, an NLRP3 inhibitor, suppressed the upregulation of NLRP3 and caspase-1 expression, and inhibited the expression of SREBP-1c and the accumulation of hepatic lipid in mice exposed to PFDA. Thus, PFDA may enhance HFD-induced adiposity and hepatic lipid accumulation through the NLRP3/caspase-1 pathway. This contaminant may be a key risk factor for obesity development in individuals consuming high-fat foods, particularly Western diet.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Yu Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Huanan Guan
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| |
Collapse
|
8
|
Heintz MM, Haws LC, Klaunig JE, Cullen JM, Thompson CM. Assessment of the mode of action underlying development of liver lesions in mice following oral exposure to HFPO-DA and relevance to humans. Toxicol Sci 2023; 192:15-29. [PMID: 36629480 PMCID: PMC10025879 DOI: 10.1093/toxsci/kfad004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)propanoate) is a short-chain polyfluorinated alkyl substance (PFAS) used in the manufacture of some types of fluorinated polymers. Like many PFAS, toxicity studies with HFPO-DA indicate the liver is the primary target of toxicity in rodents following oral exposure. Due to the structural diversity of PFAS, the mode of action (MOA) can differ between PFAS for the same target tissue. There is significant evidence for involvement of peroxisome proliferator-activated receptor alpha (PPARα) activation based on molecular and histopathological responses in the liver following HFPO-DA exposure, but other MOAs have also been hypothesized based on limited evidence. The MOA underlying the liver effects in mice exposed to HFPO-DA was assessed in the context of the Key Events (KEs) outlined in the MOA framework for PPARα activator-induced rodent hepatocarcinogenesis. The first 3 KEs (ie, PPARα activation, alteration of cell growth pathways, and perturbation of cell growth/survival) are supported by several lines of evidence from both in vitro and in vivo data available for HFPO-DA. In contrast, alternate MOAs, including cytotoxicity, PPARγ and mitochondrial dysfunction are generally not supported by the scientific literature. HFPO-DA-mediated liver effects in mice are not expected in humans as only KE 1, PPARα activation, is shared across species. PPARα-mediated gene expression in humans produces only a subset (ie, lipid modulating effects) of the responses observed in rodents. As such, the adverse effects observed in rodent livers should not be used as the basis of toxicity values for HFPO-DA for purposes of human health risk assessment.
Collapse
Affiliation(s)
| | | | - James E Klaunig
- School of Public Health, Indiana University, Bloomington, Indiana 47405, USA
| | - John M Cullen
- North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina 27606, USA
| | | |
Collapse
|
9
|
Luo Y, Kang J, Luo J, Yan Z, Li S, Lu Z, Song Y, Zhang X, Yang J, Liu A. Hepatocytic AP-1 and STAT3 contribute to chemotaxis in alphanaphthylisothiocyanate-induced cholestatic liver injury. Toxicol Lett 2023; 373:184-193. [PMID: 36460194 DOI: 10.1016/j.toxlet.2022.11.020] [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: 08/07/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
The development of cholestatic liver injury (CLI) involves inflammation, but the dominant pathway mediating the chemotaxis is not yet established. This work explored key signaling pathway mediating chemotaxis in CLI and the role of Kupffer cells in the inflammatory liver injury. Probe inhibitors T-5224 (100 mg/kg) for AP-1 and C188-9 (100 mg/kg) for STAT3 were used to validate key inflammatory pathways in alpha-naphthylisothiocyanate (ANIT, 100 mg/kg)-induced CLI. Two doses of GdCl3 (10 mg/kg and 40 mg/kg) were used to delete Kupffer cells and explore their role in CLI. Serum and liver samples were collected for biochemical and mechanism analysis. The liver injury in ANIT-treated mice were significantly increased supported by biochemical and histopathological changes, and neutrophils gathering around the necrotic loci. Inhibitor treatments down-regulated liver injury biomarkers except the level of total bile acid. The chemokine Ccl2 increased by 170-fold and to a less degree Cxcl2 by 45-fold after the ANIT treatment. p-c-Jun and p-STAT3 were activated in the group A but inhibited by the inhibitors in western blot analysis. The immunofluorescence results showed AP-1 not STAT3 responded to inhibitors in ANIT-induced CLI. With or without GdCl3, there was no significant difference in liver injury among the CLI groups. In necrotic loci in CLI, CXCL2 colocalized with hepatocyte biomarker Albumin, not with the F4/80 in Kupffer cells. Conclusively, AP-1 played a more critical role in the inflammation cascade than STAT3 in ANIT-induced CLI. Hepatocytes, not the Kupffer cells released chemotactic factors mediating the chemotaxis in CLI.
Collapse
Affiliation(s)
- Yishuang Luo
- School of Medicine, Ningbo University, 315211 Ningbo, China; Ningbo Haishu District Center for Disease Control and Prevention, 315000 Ningbo, China
| | - Jinyu Kang
- School of Medicine, Ningbo University, 315211 Ningbo, China; The Affiliated Lihuili Hospital, Ningbo University, 315000 Ningbo, China
| | - Jia Luo
- School of Medicine, Ningbo University, 315211 Ningbo, China
| | - Zheng Yan
- School of Medicine, Ningbo University, 315211 Ningbo, China
| | - Shengtao Li
- School of Medicine, Ningbo University, 315211 Ningbo, China
| | - Zhuoheng Lu
- School of Medicine, Ningbo University, 315211 Ningbo, China
| | - Yufei Song
- The Affiliated Lihuili Hospital, Ningbo University, 315000 Ningbo, China
| | - Xie Zhang
- The Affiliated Lihuili Hospital, Ningbo University, 315000 Ningbo, China
| | - Julin Yang
- Ningbo College of Health Sciences, 315100 Ningbo, China
| | - Aiming Liu
- School of Medicine, Ningbo University, 315211 Ningbo, China.
| |
Collapse
|
10
|
Wang T, Xu H, Guo Y, Li Z, Ye H, Wu L, Guo Y, Wang D. Perfluorodecanoic acid promotes adipogenesis via NLRP3 inflammasome-mediated pathway in HepG2 and 3T3-L1 cells. Food Chem Toxicol 2022; 171:113520. [PMID: 36423729 DOI: 10.1016/j.fct.2022.113520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/17/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022]
Abstract
Perfluorodecanoic acid (PFDA) is a toxic persistent pollutant that is extensively used in food applications, such as food packaging and cookware. Emerging evidence indicates that PFDA exposure were associated with higher plasma triglyceride concentration in human. In contrast, it is unknown how PFDA might affect adipogenesis. To explore the effects and underlying mechanisms of PFDA on lipid metabolism in this study, both HepG2 cells and 3T3-L1 differentiation model were used. The results showed that PFDA promoted the cellular triglyceride accumulation and triglyceride content in concentration-dependent manners. Furthermore, PFDA activated the NLRP3 inflammasome, which is crucial for the induction of lipogenic genes expression including fatty acid synthase (FAS), hydroxymethylglutaryl coenzyme A synthase (HMGCS), and stearoyl-CoA desaturase 1 (SCD1). Additionally, PFDA-induced adipogenesis was abolished by caspase-1 inhibitor and siNLRP3 in HepG2 cells. Moreover, after PFDA treatment, the expression of SREBP1, an important regulator of lipid metabolism, was increased, as well as its target genes, and PFDA-induced SREBP1 enhanced expression can be abolished by caspase-1 inhibitor and siNLRP3 as well. Together, these results provide to understanding of the potential health implications of exposure to PFDA on lipid accumulation, and suggest that PFDA can promote adipogenesis via an NLRP3 inflammasome-mediated SREBP1 pathway.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Yu Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Zhanming Li
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Hua Ye
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Liang Wu
- School of Medicine, Jiangsu University, 212013, Zhenjiang, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| |
Collapse
|
11
|
Ye X, Zhang T, Han H. PPARα: A potential therapeutic target of cholestasis. Front Pharmacol 2022; 13:916866. [PMID: 35924060 PMCID: PMC9342652 DOI: 10.3389/fphar.2022.916866] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The accumulation of bile acids in the liver leads to the development of cholestasis and hepatocyte injury. Nuclear receptors control the synthesis and transport of bile acids in the liver. Among them, the farnesoid X receptor (FXR) is the most common receptor studied in treating cholestasis. The activation of this receptor can reduce the amount of bile acid synthesis and decrease the bile acid content in the liver, alleviating cholestasis. Ursodeoxycholic acid (UDCA) and obeticholic acid (OCA) have a FXR excitatory effect, but the unresponsiveness of some patients and the side effect of pruritus seriously affect the results of UDCA or OCA treatment. The activator of peroxisome proliferator-activated receptor alpha (PPARα) has emerged as a new target for controlling the synthesis and transport of bile acids during cholestasis. Moreover, the anti-inflammatory effect of PPARα can effectively reduce cholestatic liver injury, thereby improving patients’ physiological status. Here, we will focus on the function of PPARα and its involvement in the regulation of bile acid transport and metabolism. In addition, the anti-inflammatory effects of PPARα will be discussed in some detail. Finally, we will discuss the application of PPARα agonists for cholestatic liver disorders.
Collapse
Affiliation(s)
- Xiaoyin Ye
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Tong Zhang, ; Han Han,
| | - Han Han
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Tong Zhang, ; Han Han,
| |
Collapse
|
12
|
Costello E, Rock S, Stratakis N, Eckel SP, Walker DI, Valvi D, Cserbik D, Jenkins T, Xanthakos SA, Kohli R, Sisley S, Vasiliou V, La Merrill MA, Rosen H, Conti DV, McConnell R, Chatzi L. Exposure to per- and Polyfluoroalkyl Substances and Markers of Liver Injury: A Systematic Review and Meta-Analysis. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:46001. [PMID: 35475652 PMCID: PMC9044977 DOI: 10.1289/ehp10092] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Experimental evidence indicates that exposure to certain pollutants is associated with liver damage. Per- and polyfluoroalkyl substances (PFAS) are persistent synthetic chemicals widely used in industry and consumer products and bioaccumulate in food webs and human tissues, such as the liver. OBJECTIVE The objective of this study was to conduct a systematic review of the literature and meta-analysis evaluating PFAS exposure and evidence of liver injury from rodent and epidemiological studies. METHODS PubMed and Embase were searched for all studies from earliest available indexing year through 1 December 2021 using keywords corresponding to PFAS exposure and liver injury. For data synthesis, results were limited to studies in humans and rodents assessing the following indicators of liver injury: serum alanine aminotransferase (ALT), nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, or steatosis. For human studies, at least three observational studies per PFAS were used to conduct a weighted z-score meta-analysis to determine the direction and significance of associations. For rodent studies, data were synthesized to qualitatively summarize the direction and significance of effect. RESULTS Our search yielded 85 rodent studies and 24 epidemiological studies, primarily of people from the United States. Studies focused primarily on legacy PFAS: perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS), perfluorononanoic acid (PFNA), and perfluorohexanesulfonic acid. Meta-analyses of human studies revealed that higher ALT levels were associated with exposure to PFOA (z-score= 6.20, p<0.001), PFOS (z-score= 3.55, p<0.001), and PFNA (z-score= 2.27, p=0.023). PFOA exposure was also associated with higher aspartate aminotransferase and gamma-glutamyl transferase levels in humans. In rodents, PFAS exposures consistently resulted in higher ALT levels and steatosis. CONCLUSION There is consistent evidence for PFAS hepatotoxicity from rodent studies, supported by associations of PFAS and markers of liver function in observational human studies. This review identifies a need for additional research evaluating next-generation PFAS, mixtures, and early life exposures. https://doi.org/10.1289/EHP10092.
Collapse
Affiliation(s)
- Elizabeth Costello
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sarah Rock
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nikos Stratakis
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sandrah P. Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Douglas I. Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dora Cserbik
- Barcelona Institute for Global Health, Barcelona, Spain
| | - Todd Jenkins
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stavra A. Xanthakos
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Stephanie Sisley
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, Davis, California, USA
| | - Hugo Rosen
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - David V. Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Leda Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
13
|
EFSA Panel on Contaminants in the Food Chain (EFSA CONTAM Panel), Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Vleminckx C, Wallace H, Barregård L, Ceccatelli S, Cravedi J, Halldorsson TI, Haug LS, Johansson N, Knutsen HK, Rose M, Roudot A, Van Loveren H, Vollmer G, Mackay K, Riolo F, Schwerdtle T. Risk to human health related to the presence of perfluoroalkyl substances in food. EFSA J 2020; 18:e06223. [PMID: 32994824 PMCID: PMC7507523 DOI: 10.2903/j.efsa.2020.6223] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluoroalkyl substances (PFASs) in food. Based on several similar effects in animals, toxicokinetics and observed concentrations in human blood, the CONTAM Panel decided to perform the assessment for the sum of four PFASs: PFOA, PFNA, PFHxS and PFOS. These made up half of the lower bound (LB) exposure to those PFASs with available occurrence data, the remaining contribution being primarily from PFASs with short half-lives. Equal potencies were assumed for the four PFASs included in the assessment. The mean LB exposure in adolescents and adult age groups ranged from 3 to 22, the 95th percentile from 9 to 70 ng/kg body weight (bw) per week. Toddlers and 'other children' showed a twofold higher exposure. Upper bound exposure was 4- to 49-fold higher than LB levels, but the latter were considered more reliable. 'Fish meat', 'Fruit and fruit products' and 'Eggs and egg products' contributed most to the exposure. Based on available studies in animals and humans, effects on the immune system were considered the most critical for the risk assessment. From a human study, a lowest BMDL 10 of 17.5 ng/mL for the sum of the four PFASs in serum was identified for 1-year-old children. Using PBPK modelling, this serum level of 17.5 ng/mL in children was estimated to correspond to long-term maternal exposure of 0.63 ng/kg bw per day. Since accumulation over time is important, a tolerable weekly intake (TWI) of 4.4 ng/kg bw per week was established. This TWI also protects against other potential adverse effects observed in humans. Based on the estimated LB exposure, but also reported serum levels, the CONTAM Panel concluded that parts of the European population exceed this TWI, which is of concern.
Collapse
|
14
|
Therapeutic action against chronic cholestatic liver injury by low-dose fenofibrate involves anti-chemotaxis via JNK–AP1–CCL2/CXCL2 signaling. Pharmacol Rep 2020; 72:935-944. [DOI: 10.1007/s43440-019-00043-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/16/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022]
|
15
|
Xu M, Zhang T, Lv C, Niu Q, Zong W, Tang J, Liu R. Perfluorodecanoic acid-induced oxidative stress and DNA damage investigated at the cellular and molecular levels. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 185:109699. [PMID: 31561076 DOI: 10.1016/j.ecoenv.2019.109699] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 05/15/2023]
Abstract
Perfluorodecanoic acid (PFDA) has been widely used in production of many daily necessities because of its special nature. Althoughtoxic effects of PFDA to organisms have been reported, there is little research on the genotoxicity induced by oxidative stress of PFDA on the cellular and molecular levels simultaneously. Thus, we investigated the DNA oxidative damage caused by PFDA in mouse hepatocytes. On the cellular level, an increase in ROS content indicated that PFDA caused oxidative stress in mouse hepatocytes. In addition, after PFDA exposure, the comet assay confirmed DNA strand breaks and an increased 8-OHdG content demonstrated DNA oxidative damage. On the molecular level, the microenvironment of aromatic amino acids, skeleton and secondary structure of catalase (CAT) were varied after PFDA exposure and the enzyme activity was reduced because PFDA bound near the heme groups of CAT. Moreover, PFDA was shown to interact with DNA molecule by groove binding. This study suggests that PFDA can cause genotoxicity by inducing oxidative stress both on the cellular and molecular levels.
Collapse
Affiliation(s)
- Mengchen Xu
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong, 266237, PR China
| | - Tong Zhang
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong, 266237, PR China
| | - Chao Lv
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong, 266237, PR China
| | - Qigui Niu
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong, 266237, PR China
| | - Wansong Zong
- College of Population, Resources and Environment, Shandong Normal University, 88# East Wenhua Road, Jinan, 250014, PR China
| | - Jingchun Tang
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Engineering Center of Environmental Diagnosis and Contamination Remediation, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, PR China
| | - Rutao Liu
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong, 266237, PR China.
| |
Collapse
|
16
|
Liu YZ, Zhang ZP, Fu ZW, Yang K, Ding N, Hu LG, Fang ZZ, Zhuo X. Per- and polyfluoroalkyl substances display structure-dependent inhibition towards UDP-glucuronosyltransferases. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:113093. [PMID: 31472452 DOI: 10.1016/j.envpol.2019.113093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/31/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a large group of chemicals and can be detected in environmental and human samples all over the world. Toxicity of existing and emerging PFASs will be a long-term source of concern. This study aimed to investigate structure-dependent inhibitory effects of 14 PFASs towards the activity of 11 UDP-glucuronosyltransferase (UGT) isoforms. In vitro UGTs-catalyzed glucuronidation of 4-methylumbelliferone (4-MU) was employed to determine the inhibition of PFASs towards different UGT isoforms. All the PFASs showed <75% of inhibition or stimulation effects on UGT1A3, UGT1A7, UGT1A9, UGT2B4, UGT2B7 and UGT2B17. However, PFASs showed broad inhibition on the activity of UGT1A1 and UGT1A8. The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 μmoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. UGT1A8 was inhibited by 97.6%, 94.8%, 86.3%, 83.4% and 77.1% by PFDA, PFTA, perfluorooctadecanoic acid (PFOcDA), PFDoA and PFOS, respectively. Additionally, PFDA significantly inhibited UGT1A6 and UGT1A10 by 96.8% and 91.6%, respectively. PFDoA inhibited the activity of UGT2B15 by 88.2%. PFDA and PFOS exhibited competitive inhibition towards UGT1A1, and PFDA and PFTA showed competitive inhibition towards UGT1A8. The inhibition kinetic parameter (Ki) were 3.15, 1.73, 13.15 and 20.21 μmoL/L for PFDA-1A1, PFOS-1A1, PFDA-1A8 and PFTA-1A8, respectively. The values were calculated to be 0.3 μmoL/L and 1.3 μmoL/L for the in vivo inhibition of PFDA towards UGT1A1-and UGT1A8-catalyzed metabolism of substances, and 0.2 μmoL/L and 2.0 μmoL/L for the inhibition of PFOS towards UGT1A1 and the inhibition of PFTA towards UGT1A8, respectively. Molecular docking indicated that hydrogen bonds and hydrophobic interactions contributed to the interaction between PFASs and UGT isoforms. In conclusion, exposure to PFASs might inhibit the activity of UGTs to disturb metabolism of endogenous compounds and xenobiotics. The structure-related effects of PFASs on UGTs would be very important for risk assessment of PFASs.
Collapse
Affiliation(s)
- Yong-Zhe Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China
| | - Zhi-Peng Zhang
- Department of Surgery, Peking University Third Hospital, Beijing, China
| | - Zhi-Wei Fu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China
| | - Kun Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China
| | - Ning Ding
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Shaanxi, Xi'an, 710061, China
| | - Li-Gang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin 300070, China.
| | - Xiaozhen Zhuo
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Shaanxi, Xi'an, 710061, China.
| |
Collapse
|
17
|
Wang D, Gao Q, Wang T, Kan Z, Li X, Hu L, Peng CY, Qian F, Wang Y, Granato D. Green tea polyphenols and epigallocatechin-3-gallate protect against perfluorodecanoic acid induced liver damage and inflammation in mice by inhibiting NLRP3 inflammasome activation. Food Res Int 2019; 127:108628. [PMID: 31882076 DOI: 10.1016/j.foodres.2019.108628] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/02/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022]
Abstract
Perfluorodecanoic acid (PFDA) is a highly toxic food contaminant that is extensively used in food applications as surface antifouling agent. In this present study, we aimed to assess whether green tea polyphenols (GTPs) and epigallocatechin-3-gallate (EGCG) exert protective effects against PFDA-induced liver damage and inflammation in mice. A mouse model to evaluate liver toxicity was established by giving mice drinking water containing different concentrations of PFDA. GTPs or EGCG (0.32%, w/v) were co-administered to mice exposed to PFDA in drinking water. Overall, GTPs and EGCG extended the survival time and inhibited weight loss among mice who received a lower dose of PFDA. Moreover, GTPs and EGCG ameliorated hepatic oxidative stress, cell apoptosis, necrosis, steatosis, edema, and degeneration, reduced hepatic inflammation and NLRP3 inflammasome activation caused by a moderate dose of PFDA. Taken together, these results show that GTPs or EGCG (or green tea intake) supplements can be beneficial for people exposed to PFDA.
Collapse
Affiliation(s)
- Dongxu Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Qiang Gao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, PR China
| | - Zhipeng Kan
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Xin Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Lizhen Hu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Chuan-Yi Peng
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Frank Qian
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Yijun Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China.
| | - Daniel Granato
- Food Processing and Quality, Production Systems Unit, Natural Resources Institute Finland, Luke, Maarintie 6, Open Innovation House, FI-02150 Espoo, Finland.
| |
Collapse
|
18
|
Wiener RC, Waters C. Perfluoroalkyls/polyfluoroalkyl substances and dental caries experience in children, ages 3-11 years, National Health and Nutrition Examination Survey, 2013-2014. J Public Health Dent 2019; 79:307-319. [PMID: 31286520 DOI: 10.1111/jphd.12329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 05/17/2019] [Accepted: 06/11/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The objective of this research is to determine the association of seven perfluoroalkyl and polyfluoroalkyl substances versus dental caries experience in US children, ages 3-11 years. METHODS A cross-sectional study design was used in the analysis of National Health and Nutrition Examination Survey 2013-2014 serological data of perfluoroalkyl and polyfluoroalkyl substances. The seven perfluoroalkyl and polyfluoroalkyl substances were: 2-(N-methyl-perfluorooctane sulfonamide) acetic acid; perfluorodecanoic acid; perfluorononanoic acid; perfluorohexane sulfonic acid; linear isomers of perfluorooctanoate; linear perfluorooctane sulfonate; and monomethyl branched isomers of perfluorooctane sulfonate. Two summative variables were created: monomethyl branch isomers of perfluorooctane sulfonic acid with linear isomer of perfluorooctane and branch isomers of perfluorooctanoate with linear isomer perfluorooctonate. RESULTS In unadjusted logistic regression, in which the comparison was between the less than 75th percentile reference group and the 75th and above percentile group, higher perfluorodecanoic acid was associated with dental caries experience [unadjusted odds ratio: 1.79 (95% CI: 1.19, 2.46; P = 0.0069); adjusted odds ratio: 1.54 (95% CI: 1.03, 2.30; P = 0.0385)]. CONCLUSIONS Of the seven examined perfluoroalkyl and polyfluoroalkyl substances, only perfluorodecanoic acid had an association with dental caries experience in an unadjusted model and adjusted logistic regression model.
Collapse
Affiliation(s)
- R Constance Wiener
- Dental Practice and Rural Health, West Virginia University, Morgantown, WV, USA
| | - Christopher Waters
- Department of Dental Research, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
19
|
Hua H, Dai M, Luo Y, Lin H, Xu G, Hu X, Xu L, Zhang H, Tang Z, Chang L, Liu A, Yang J. Basal PPARα inhibits bile acid metabolism adaptation in chronic cholestatic model induced by α-naphthylisothiocyanate. Toxicol Lett 2018; 300:31-39. [PMID: 30352267 DOI: 10.1016/j.toxlet.2018.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/26/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022]
Abstract
Cholestasis is one of the most challenging diseases to be treated in current hepatology. However little is known about the adaptation difference and the underlying mechanism between acute and chronic cholestasis. In this study, wild-type and Pparα-null mice were orally administered diet containing 0.05% ANIT to induce chronic cholestasis. Biochemistry, histopathology and serum metabolome analysis exhibited the similar toxic phenotype between wild-type and Pparα-null mice. Bile acid metabolism was strongly adapted in Pparα-null mice but not in wild-type mice. The Shp and Fxr mRNA was found to be doubled in cholestatic Pparα-null mice compared with the control group. Western blot confirmed the up-regulated expression of FXR in Pparα-null mice treated with ANIT. Inflammation was found to be stronger in Pparα-null mice than those in wild-type mice in chronic cholestasis. These data chain indicated that bile acid metabolism and inflammation signaling were different between wild-type and Pparα-null mice developing chronic cholestasis, although their toxic phenotypes could not be discriminated. So basal PPARα cross-talked with FXR and inhibited bile acid metabolism adaptation in chronic cholestasis.
Collapse
Affiliation(s)
- Huiying Hua
- Medical School of Ningbo University, Ningbo 315211, China
| | - Manyun Dai
- Medical School of Ningbo University, Ningbo 315211, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yishuang Luo
- Medical School of Ningbo University, Ningbo 315211, China
| | - Hante Lin
- Medical School of Ningbo University, Ningbo 315211, China
| | - Gangming Xu
- Medical School of Ningbo University, Ningbo 315211, China
| | - Xiaowei Hu
- Medical School of Ningbo University, Ningbo 315211, China
| | - Liping Xu
- Medical School of Ningbo University, Ningbo 315211, China
| | - Haoyue Zhang
- Medical School of Ningbo University, Ningbo 315211, China
| | - Zhiyuan Tang
- Medical School of Ningbo University, Ningbo 315211, China
| | - Liming Chang
- Medical School of Ningbo University, Ningbo 315211, China
| | - Aiming Liu
- Medical School of Ningbo University, Ningbo 315211, China.
| | - Julin Yang
- Ningbo College of Health Sciences, Ningbo 315100, China.
| |
Collapse
|
20
|
Dai M, Hua H, Lin H, Xu G, Hu X, Li F, Gonzalez FJ, Liu A, Yang J. Targeted Metabolomics Reveals a Protective Role for Basal PPARα in Cholestasis Induced by α-Naphthylisothiocyanate. J Proteome Res 2018; 17:1500-1508. [PMID: 29498526 DOI: 10.1021/acs.jproteome.7b00838] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
α-Naphthylisothiocyanate (ANIT) is an experimental agent used to induce intrahepatic cholestasis. The Ppara-null mouse line is widely employed to explore the physiological and pathological roles of PPARα. However, little is known about how PPARα influences the hepatotoxicity of ANIT. In the present study, wild-type and Ppara-null mice were orally treated with ANIT to induce cholestasis. The serum metabolome of wild-type mice segregated from that of the Ppara-null mice, driven by changes of bile acid (BA) metabolites. Alkaline phosphatase and total BAs were elevated preferentially in Ppara-null mice, which correlated with changes in Cyp7a1, Cyp8b1, Mrp3, Cyp3a11, Cyp2b10, Ugt1a2, and Ugt1a5 genes and showed cross-talk between basal PPARα and potentially adaptive pathways. Il6, Tnfa, and target genes in the STAT3 pathway ( Socs3, Fga, Fgb, and Fgg) were up-regulated in Ppara-null mice but not in wild-type mice. The JNK pathway was activated in both mouse lines, while NF-κB and STAT3 were activated only in Ppara-null mice. These data suggest protection against cholestasis by basal PPARα involves regulation of BA metabolism and inhibition of NF-κB/STAT3 signaling. Considering studies on the protective effects of both basal and activated PPARα, caution should be exercised when one attempts to draw conclusions in which the PPARα is modified by genetic manipulation, fasting, or activation in pharmacological and toxicological studies.
Collapse
Affiliation(s)
- Manyun Dai
- Zhejiang Key Laboratory of Pathophysiology , Medical School of Ningbo University , Ningbo 315211 , China
| | - Huiying Hua
- Zhejiang Key Laboratory of Pathophysiology , Medical School of Ningbo University , Ningbo 315211 , China
| | - Hante Lin
- Zhejiang Key Laboratory of Pathophysiology , Medical School of Ningbo University , Ningbo 315211 , China
| | - Gangming Xu
- Zhejiang Key Laboratory of Pathophysiology , Medical School of Ningbo University , Ningbo 315211 , China
| | - Xiaowei Hu
- Zhejiang Key Laboratory of Pathophysiology , Medical School of Ningbo University , Ningbo 315211 , China
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming 650201 , China
| | - Frank J Gonzalez
- Laboratory of Metabolism , National Cancer Institute, NIH , Bethesda , Maryland 20892 , United States
| | - Aiming Liu
- Zhejiang Key Laboratory of Pathophysiology , Medical School of Ningbo University , Ningbo 315211 , China
| | - Julin Yang
- Ningbo College of Health Sciences , Ningbo 315100 , China
| |
Collapse
|
21
|
Shi C, Min L, Yang J, Dai M, Song D, Hua H, Xu G, Gonzalez FJ, Liu A. Peroxisome Proliferator-Activated Receptor α Activation Suppresses Cytochrome P450 Induction Potential in Mice Treated with Gemfibrozil. Basic Clin Pharmacol Toxicol 2017; 121:169-174. [PMID: 28374976 DOI: 10.1111/bcpt.12794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/29/2017] [Indexed: 12/19/2022]
Abstract
Gemfibrozil, a peroxisome proliferator-activated receptor α (PPARα) agonist, is widely used for hypertriglyceridaemia and mixed hyperlipidaemia. Drug-drug interaction of gemfibrozil and other PPARα agonists has been reported. However, the role of PPARα in cytochrome P450 (CYP) induction by fibrates is not well known. In this study, wild-type mice were first fed gemfibrozil-containing diets (0.375%, 0.75% and 1.5%) for 14 days to establish a dose-response relationship for CYP induction. Then, wild-type mice and Pparα-null mice were treated with a 0.75% gemfibrozil-containing diet for 7 days. CYP3a, CYP2b and CYP2c were induced in a dose-dependent manner by gemfibrozil. In Pparα-null mice, their mRNA level, protein level and activity were induced more than those in wild-type mice. So, gemfibrozil induced CYP, and this action was inhibited by activated PPARα. These data suggested that the induction potential of CYPs was suppressed by activated PPARα, showing a potential role of this receptor in drug-drug interactions and metabolic diseases treated with fibrates.
Collapse
Affiliation(s)
- Cunzhong Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luo Min
- Medical School of Ningbo University, Ningbo, China
| | - Julin Yang
- Ningbo College of Health Sciences, Ningbo, China
| | - Manyun Dai
- Medical School of Ningbo University, Ningbo, China
| | - Danjun Song
- Medical School of Ningbo University, Ningbo, China
| | - Huiying Hua
- Medical School of Ningbo University, Ningbo, China
| | - Gangming Xu
- Medical School of Ningbo University, Ningbo, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, NIH, Bethesda, USA
| | - Aiming Liu
- Medical School of Ningbo University, Ningbo, China
| |
Collapse
|