1
|
Ren J, Zuo J, Yin B, Huang D, Wen R, Pei H, Liu J, Zhang Y, Zhu S, Zhen S, Ma Y. Flaxseed Oil Alleviates PFOS-Induced Liver Injury by Regulating Hepatic Cholesterol Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23465-23477. [PMID: 39392608 DOI: 10.1021/acs.jafc.4c04438] [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: 10/12/2024]
Abstract
Perfluorooctanesulfonate (PFOS) is a widespread, persistent environmental pollutant that exerts apparent liver toxicity. Flaxseed oil (FO), a dietary oil rich in α-linolenic acid, has been demonstrated to possess a diverse array of health benefits. However, whether FO protects against PFOS-induced liver injury and its underlying mechanisms remain unclear. C57/BL6 mice were orally treated with different concentrations of FO alone or in combination with 10 mg/kg of PFOS for 28 consecutive days. Blood and liver tissues were collected for proteomic, histopathological, biochemical, immunohistochemical, and molecular examinations. Results demonstrated that FO supplementation reduced PFOS-induced liver injury, as evidenced by a decrease in histopathological changes, serum transaminase (ALT and AST) levels, levels of oxidative stress, and inflammatory cytokine (TNF-α, IL-1β, and IL-6) levels. Proteomic analyses showed that differentially expressed proteins were enriched in cholesterol metabolic pathways when comparing the PFOS group to the FO supplementation groups. The expression of cholesterol metabolism-related proteins was also subsequently measured, revealing that FO supplementation decreased the protein expressions of SREBP2, HMGCR, and LDLR while increasing the expression of CYP7A1. This study demonstrates that FO can alleviate PFOS-induced hepatotoxicity by regulating hepatic cholesterol metabolism, indicating that FO may serve as an effective dietary intervention for preventing liver injury caused by PFOS.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Dan Huang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Huanting Pei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jiarui Liu
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Siqi Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Shuman Zhen
- Department of Radiotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| |
Collapse
|
2
|
Ryu S, Burchett W, Zhang S, Jia X, Modaresi SMS, Agudelo Areiza J, Rodrigues D, Zhu H, Sunderland EM, Fischer FC, Slitt AL. Unbound Fractions of PFAS in Human and Rodent Tissues: Rat Liver a Suitable Proxy for Evaluating Emerging PFAS? ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:14641-14650. [PMID: 39161261 DOI: 10.1021/acs.est.4c04050] [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: 08/21/2024]
Abstract
Adverse health effects associated with exposures to perfluoroalkyl and polyfluoroalkyl substances (PFAS) are a concern for public health and are driven by their elimination half-lives and accumulation in specific tissues. However, data on PFAS binding in human tissues are limited. Accumulation of PFAS in human tissues has been linked to interactions with specific proteins and lipids in target organs. Additional data on PFAS binding and unbound fractions (funbound) in whole human tissues are urgently needed. Here, we address this gap by using rapid equilibrium dialysis to measure the binding and funbound of 16 PFAS with 3 to 13 perfluorinated carbon atoms (ηpfc = 3-13) and several functional headgroups in human liver, lung, kidney, heart, and brain tissue. We compare results to mouse (C57BL/6 and CD-1) and rat tissues. Results show that funbound decreases with increasing fluorinated carbon chain length and hydrophobicity. Among human tissues, PFAS binding was generally greatest in brain > liver ≈ kidneys ≈ heart > lungs. A correlation analysis among human and rodent tissues identified rat liver as a suitable surrogate for predicting funbound for PFAS in human tissues (R2 ≥ 0.98). The funbound data resulting from this work and the rat liver prediction method offer input parameters and tools for toxicokinetic models for legacy and emerging PFAS.
Collapse
Affiliation(s)
- Sangwoo Ryu
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Incorporated, Groton, Connecticut 06340, United States
| | - Woodrow Burchett
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Incorporated, Groton, Connecticut 06340, United States
| | - Sam Zhang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Incorporated, Groton, Connecticut 06340, United States
| | - Xuelian Jia
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
- Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, Louisiana 70112, United States
| | - Seyed Mohamad Sadegh Modaresi
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Juliana Agudelo Areiza
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - David Rodrigues
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Pfizer Incorporated, Groton, Connecticut 06340, United States
| | - Hao Zhu
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
- Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, Louisiana 70112, United States
| | - Elsie M Sunderland
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Fabian Christoph Fischer
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| |
Collapse
|
3
|
Zhou R, Peng J, Zhang L, Sun Y, Yan J, Jiang H. Association between the dietary inflammatory index and serum perfluoroalkyl and polyfluoroalkyl substance concentrations: evidence from NANHES 2007-2018. Food Funct 2024; 15:7375-7386. [PMID: 37779497 DOI: 10.1039/d3fo01487h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Diet is an important source of perfluoroalkyl and polyfluoroalkyl substance (PFAS) exposure, and the dietary inflammatory index (DII) is a tool used to assess the inflammatory potential of an individual's diet. However, limited research has explored the association between the DII and PFAS exposure in humans. This study is the first to analyze the association between the five PFASs and DII using the National Health and Nutrition Examination Survey (NHANES) 2007-2018 database. Additionally, we assessed the interaction between the DII and PFASs regarding oxidative stress and inflammatory markers, including alkaline phosphatase, albumin, neutrophil count, lymphocyte count, total bilirubin, and serum iron based on a previous study. A series of covariates were included in the analysis to reduce the confounding bias. The study included 7773 and 5933 participants based on the different models. The DII was significantly associated with serum perfluorooctanoic acid, perfluorononanoic acid, perfluorooctane sulfonic acid, and sum-PFAS. Some of the food parameters used to calculate the DII also showed associations with special PFAS serum concentrations. Specifically, dietary fiber, n-3 polyunsaturated fatty acids, energy intake, and vitamin D were associated with more than three PFASs. Higher DII levels in participants were linked to a more significant association between bilirubin (the interaction P-value is not significant), alkaline phosphatase, serum iron, neutrophil counts, and some PFASs. In conclusion, this study clarified the association between the three PFASs and DII, highlighting the diverse effects of PFASs on oxidative stress and inflammatory markers across different DII levels.
Collapse
Affiliation(s)
- Ren Zhou
- Department of Anesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Jiali Peng
- Department of Anesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Lei Zhang
- Department of Anesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Yu Sun
- Department of Anesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Jia Yan
- Department of Anesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Hong Jiang
- Department of Anesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| |
Collapse
|
4
|
Kaye E, Marques E, Agudelo Areiza J, Modaresi SMS, Slitt A. Exposure to a PFOA, PFOS and PFHxS Mixture during Gestation and Lactation Alters the Liver Proteome in Offspring of CD-1 Mice. TOXICS 2024; 12:348. [PMID: 38787127 PMCID: PMC11126053 DOI: 10.3390/toxics12050348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Perfluroalkyl substances (PFASs) are persistent man-made chemicals considered to be emerging pollutants, with Perfluorooctanoic acid (PFOA), Perfluorooctanesulfonic acid (PFOS), and Perfluorohexanesulphonic acid (PFHxS) being linked to hepatotoxicity and steatosis. PFOA, PFOS, and PFHxS can undergo placental and lactational transfer, which results in PFOA, PFOS, and PFHxS distribution to the neonatal liver. Moreover, in pregnant dams, exposure to a PFAS mixture, in combination with a high fat diet, increased hepatic steatosis in offspring at postnatal day 21, but the mechanisms have not been elucidated. It was hypothesized that gestational/lactational PFAS exposure would alter the pup liver proteome and biochemical/signaling pathways. Timed-pregnant CD-1 dams were fed a standard chow or 60% kcal high-fat diet. From GD1 until PND20, dams were dosed via oral gavage with vehicle (0.5% Tween 20), individual doses of PFOA, PFOS, PFHxS at 1 mg/kg, or a mixture (1 mg/kg each, totaling 3 mg/kg). Livers were collected from PND21 offspring and SWATH-MS proteomics was performed. IPA analysis revealed PFAS exposure modified disease and biological function pathways involved in liver damage, xenobiotics, and lipid regulation in the PND21 liver. These pathways included lipid and fatty acid transport, storage, oxidation, and synthesis, as well as xenobiotic metabolism and transport, and liver damage and inflammation. This indicates the pup liver proteome is altered via maternal exposure and predisposes the pup to metabolic dysfunctions.
Collapse
Affiliation(s)
- Emily Kaye
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA; (E.K.); (E.M.); (J.A.A.); (S.M.S.M.)
| | - Emily Marques
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA; (E.K.); (E.M.); (J.A.A.); (S.M.S.M.)
- Office of Pollution Prevention and Toxics, US EPA, 1200 Pennsylvania Ave. NW, Washington, DC 20460, USA
| | - Juliana Agudelo Areiza
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA; (E.K.); (E.M.); (J.A.A.); (S.M.S.M.)
| | - Seyed Mohamad Sadegh Modaresi
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA; (E.K.); (E.M.); (J.A.A.); (S.M.S.M.)
| | - Angela Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA; (E.K.); (E.M.); (J.A.A.); (S.M.S.M.)
| |
Collapse
|
5
|
Mao X, Liu Y, Wei Y, Li X, Liu Y, Su G, Wang X, Jia J, Yan B. Threats of per- and poly-fluoroalkyl pollutants to susceptible populations. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 921:171188. [PMID: 38395163 DOI: 10.1016/j.scitotenv.2024.171188] [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: 10/18/2023] [Revised: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Environmental exposure to per- and poly-fluoroalkyl substances (PFAS) has raised significant global health concerns due to potential hazards in healthy adults. However, the impact of PFAS on susceptible populations, including pregnant individuals, newborns, the older people, and those with underlying health conditions, has been overlooked. These susceptible groups often have physiological changes that make them less resilient to the same exposures. Consequently, there is an urgent need for a comprehensive understanding of the health risks posed by PFAS exposure to these populations. In this review, we delve into the potential health risks of PFAS exposure in these susceptible populations. Equally important, we also examine and discuss the molecular mechanisms that underlie this susceptibility. These mechanisms include the induction of oxidative stress, disruption of the immune system, impairment of cellular metabolism, and alterations in gut microbiota, all of which contribute to the enhanced toxicity of PFAS in susceptible populations. Finally, we address the primary research challenges and unresolved issues that require further investigation. This discussion aims to foster research for a better understanding of how PFAS affect susceptible populations and to pave the way for strategies to minimize their adverse effects.
Collapse
Affiliation(s)
- Xuan Mao
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yujiao Liu
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yongyi Wei
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Xiaodi Li
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Xiaohong Wang
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.
| | - Jianbo Jia
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.
| | - Bing Yan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
6
|
Zhang Y, Lv J, Fan YJ, Tao L, Xu J, Tang W, Sun N, Zhao LL, Xu DX, Huang Y. Evaluating the Effect of Gestational Exposure to Perfluorohexane Sulfonate on Placental Development in Mice Combining Alternative Splicing and Gene Expression Analyses. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:117011. [PMID: 37995155 PMCID: PMC10666825 DOI: 10.1289/ehp13217] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Perfluorohexane sulfonate (PFHxS) is a frequently detected per- and polyfluoroalkyl substance in most populations, including in individuals who are pregnant, a period critical for early life development. Despite epidemiological evidence of exposure, developmental toxicity, particularly at realistic human exposures, remains understudied. OBJECTIVES We evaluated the effect of gestational exposure to human-relevant body burden of PFHxS on fetal and placental development and explored mechanisms of action combining alternative splicing (AS) and gene expression (GE) analyses. METHODS Pregnant ICR mice were exposed to 0, 0.03, and 0.3 μ g / kg / day from gestational day 7 to day 17 via oral gavage. Upon euthanasia, PFHxS distribution was measured using liquid chromatography-tandem mass spectrometry. Maternal and fetal phenotypes were recorded, and histopathology was examined for placenta impairment. Multiomics was adopted by combining AS and GE analyses to unveil disruptions in mRNA quality and quantity. The key metabolite transporters were validated by quantitative real-time PCR (qRT-PCR) for quantification and three-dimensional (3D) structural simulation by AlphaFold2. Targeted metabolomics based on liquid chromatography-tandem mass spectrometry was used to detect amino acid and amides levels in the placenta. RESULTS Pups developmentally exposed to PFHxS exhibited signs of intrauterine growth restriction (IUGR), characterized by smaller fetal weight and body length (p < 0.01 ) compared to control mice. PFHxS concentration in maternal plasma was 5.01 ± 0.54 ng / mL . PFHxS trans-placenta distribution suggested dose-dependent transfer through placental barrier. Histopathology of placenta of exposed dams showed placental dysplasia, manifested with an attenuated labyrinthine layer area and deescalated blood sinus counts and placental vascular development index marker CD34. Combined GE and AS analyses pinpointed differences in genes associated with key biological processes of placental development, proliferation, metabolism, and transport in placenta of exposed dams compared to that of control dams. Further detection of placental key transporter gene expression, protein structure simulation, and amino acid and amide metabolites levels suggested that PFHxS exposure during pregnancy led to impairment of placental amino acid transportation. DISCUSSION The findings from this study suggest that exposure to human-relevant very-low-dose PFHxS during pregnancy in mice caused IUGR, likely via downregulating of placental amino acid transporters, thereby impairing placental amino acid transportation, resulting in impairment of placental development. Our findings confirm epidemiological findings and call for future attention on the health risk of this persistent yet ubiquitous chemical in the early developmental stage and provide a new approach for understanding gene expression from both quantitative and qualitative omics approaches in toxicological studies. https://doi.org/10.1289/EHP13217.
Collapse
Affiliation(s)
- Yihao Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jia Lv
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Yi-Jun Fan
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
- Department of Gynecology and Obstetrics, Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Lin Tao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jingjing Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Weitian Tang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Nan Sun
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Ling-Li Zhao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the PRC, Hefei, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the PRC, Hefei, China
| |
Collapse
|
7
|
Duh-Leong C, Maffini MV, Kassotis CD, Vandenberg LN, Trasande L. The regulation of endocrine-disrupting chemicals to minimize their impact on health. Nat Rev Endocrinol 2023; 19:600-614. [PMID: 37553404 DOI: 10.1038/s41574-023-00872-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 08/10/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) are substances generated by human industrial activities that are detrimental to human health through their effects on the endocrine system. The global societal and economic burden posed by EDCs is substantial. Poorly defined or unenforced policies can increase human exposure to EDCs, thereby contributing to human disease, disability and economic damage. Researchers have shown that policies and interventions implemented at both individual and government levels have the potential to reduce exposure to EDCs. This Review describes a set of evidence-based policy actions to manage, minimize or even eliminate the widespread use of these chemicals and better protect human health and society. A number of specific challenges exist: defining, identifying and prioritizing EDCs; considering the non-linear or non-monotonic properties of EDCs; accounting for EDC exposure effects that are latent and do not appear until later in life; and updating testing paradigms to reflect 'real-world' mixtures of chemicals and cumulative exposure. A sound strategy also requires partnering with health-care providers to integrate strategies to prevent EDC exposure in clinical care. Critical next steps include addressing EDCs within global policy frameworks by integrating EDC exposure prevention into emerging climate policy.
Collapse
Affiliation(s)
- Carol Duh-Leong
- Department of Pediatrics, New York University Grossman School of Medicine, New York, NY, USA
| | | | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Laura N Vandenberg
- Department of Environmental Health Sciences, University of Massachusetts - Amherst, Amherst, MA, USA
| | - Leonardo Trasande
- Department of Pediatrics, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, USA.
- New York University Wagner Graduate School of Public Service, New York, NY, USA.
| |
Collapse
|
8
|
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: 1.0] [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
|
9
|
Yao W, Xu J, Tang W, Gao C, Tao L, Yu J, Lv J, Wang H, Fan Y, Xu DX, Huang Y. Developmental toxicity of perfluorohexane sulfonate at human relevant dose during pregnancy via disruption in placental lipid homeostasis. ENVIRONMENT INTERNATIONAL 2023; 177:108014. [PMID: 37315490 DOI: 10.1016/j.envint.2023.108014] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
Perfluorohexyl sulfonate (PFHxS) is the third most abundant per- and polyfluoroalkyl substances and its developmental toxicity remains very poorly understood. Here, pregnant mice exposed to PFHxS at human relevant dose showed increased fetal death incidence in the high-dose PFHxS-H group (P < 0.01). Body distribution analyses suggested that PFHxS crossed the placental barrier reaching the fetus in a dose-dependent manner. Histopathological data demonstrated impairment in the placenta with reduced blood sinus volume, placental labyrinth area as well as thickness of labyrinthine layer. Further lipidomic and transcriptomic data together showed that PFHxS exposure caused significant disruption in placental lipid homeostasis, including total lipid accumulation in the placenta, and dysregulation in phospholipid and glycerol lipid metabolism. Gene expression analyses uncovered elevation in key placental fatty acid transporters including fabp2, whereas protein expression showed transporter specific disruptions following exposure. Together, gestational exposure to human relevant level of PFHxS may increase the incidence of fetal deaths and caused placental dysplasia via disruption in lipid metabolism homeostasis. These findings raise the concern regarding the highly prevalent and persistent chemical towards early sensitive developing stages and provide basis for further understanding of its effects on lipid metabolism and underlying mechanisms.
Collapse
Affiliation(s)
- Wencong Yao
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jingjing Xu
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Weitian Tang
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Chang Gao
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lin Tao
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jie Yu
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jia Lv
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Yijun Fan
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China; Department of Gynecology and Obstetrics, Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| |
Collapse
|
10
|
Pineda S, Lignell S, Gyllenhammar I, Lampa E, Benskin JP, Lundh T, Lindh C, Kiviranta H, Glynn A. Exposure of Swedish adolescents to elements, persistent organic pollutants (POPs), and rapidly excreted substances - The Riksmaten adolescents 2016-17 national survey. Int J Hyg Environ Health 2023; 251:114196. [PMID: 37279611 DOI: 10.1016/j.ijheh.2023.114196] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/08/2023]
Abstract
Adolescence is a period of significant physiological changes, and likely a sensitive window to chemical exposure. Few nation-wide population-based studies of chemical body burdens in adolescents have been published. In the national dietary survey Riksmaten Adolescents (RMA) 2016-17, over 13 chemical substance groups, including elements, chlorinated/brominated/fluorinated persistent organic pollutants (POPs) were analysed in blood, and in urine metabolites of phthalates/phthalate alternatives, phosphorous flame retardants, polycyclic aromatic hydrocarbons (PAHs), and pesticides, along with bisphenols and biocide/preservative/antioxidant/UV filter substances (N = 1082, ages 11-21). The aim was to characterize the body burdens in a representative population of adolescents in Sweden, and to compare results with human biomonitoring guidance values (HBM-GVs). Cluster analyses and Spearman's rank order correlations suggested that concentrations of substances with known common exposure sources and similar toxicokinetics formed obvious clusters and showed moderate to very strong correlations (r ≥ 0.4). No clusters were formed between substances from different matrices. Geometric mean (GM) concentrations of the substances were generally less than 3-fold different from those observed among adolescents in NHANES (USA 2015-16) and GerES V (Germany 2014-17). Notable exceptions were brominated diphenyl ethers (PBDEs) with >20-fold lower GM concentrations, and the biocide triclosan and ultraviolet (UV) filter benzophenone-3 with >15-fold lower mean concentrations in RMA compared to NHANES. Exceedance of the most conservative HBM-GVs were observed for aluminium (Al, 26% of subjects), perfluorooctanesulfonic acid (PFOS, 19%), perfluorooctanoic acid (PFOA, 12%), lead (Pb, 12%), MBP (dibutyl phthalate metabolite, 4.8%), hexachlorobenzene (HCB, 3.1%) and 3-phenoxybenzoic acid (PBA, pyrethroid metabolite, 2.2%). Males showed a higher proportion of exceedances than females for Pb, HCB and PFOS; otherwise no gender-related differences in exceedances were observed. A higher proportion of males than females had a Hazard Index (HI) of substances with liver and kidney toxicity and neurotoxicity >1. Industrialized countries with similarly high standards of living, with some exceptions, show comparable average body burdens of a variety of toxic chemicals among adolescents from the general population. The exceedances of HBM-GVs and HIs strongly suggests that further efforts to limit chemical exposure are warranted.
Collapse
Affiliation(s)
- Sebastian Pineda
- Department of Biomedicine and Veterinary Public Health Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Sanna Lignell
- Department of Risk and Benefit Assessment, Swedish National Food Agency, Uppsala, Sweden
| | - Irina Gyllenhammar
- Department of Risk and Benefit Assessment, Swedish National Food Agency, Uppsala, Sweden
| | - Erik Lampa
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonathan P Benskin
- Department of Environmental Science, Stockholm University, Stockholm, Sweden
| | - Thomas Lundh
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Christian Lindh
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Hannu Kiviranta
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anders Glynn
- Department of Biomedicine and Veterinary Public Health Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
11
|
Ducatman A, Tan Y, Nadeau B, Steenland K. Perfluorooctanoic Acid (PFOA) Exposure and Abnormal Alanine Aminotransferase: Using Clinical Consensus Cutoffs Compared to Statistical Cutoffs for Abnormal Values. TOXICS 2023; 11:toxics11050449. [PMID: 37235263 DOI: 10.3390/toxics11050449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFASs) including perfluorooctanoic acid (PFOA) are ubiquitous environmental contaminants. Prior analysis in the large "C8 Health Project" population defined abnormal alanine aminotransferase (ALT) with statistically derived cutoffs (>45 IU/L in men, >34 IU/L in women). OBJECTIVE To explore the degree to which PFOA was associated with modern, clinically predictive ALT biomarker cutoffs in obese and nonobese participants, excluding those with diagnosed liver disease. METHODS We reevaluated the relationship of serum PFOA to abnormal ALT using predictive cutoff recommendations including those of the American College of Gastroenterology (ACG). Evaluations modeled lifetime cumulative exposure and measured internal PFOA exposure. RESULTS ACG cutoff values (≥34 IU/L for males, ≥25 IU/L for females) classified 30% of males (3815/12,672) and 21% of females (3359/15,788) above ALT cutoff values. Odds ratios (OR) for above cutoff values were consistently associated with modeled cumulative and measured serum PFOA. Linear trends were highly significant. ORs by quintile showed near monotonic increases. Trends were stronger for the overweight and obese. However, all weight classes were affected. CONCLUSION Predictive cutoffs increase the OR for abnormal ALT results. Obesity increases ORs, yet association with abnormal ALT pertains to all weight classes. The results are discussed in context of current knowledge about the health implications of PFOA hepatotoxicity.
Collapse
Affiliation(s)
- Alan Ducatman
- School of Public Health, West Virginia University, Morgantown, WV 26506-9190, USA
| | - Youran Tan
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Brian Nadeau
- Department of Gastroenterology, William Beaumont Hospital, Royal Oak, MI 48173, USA
| | - Kyle Steenland
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
12
|
Conley JM, Lambright CS, Evans N, Medlock-Kakaley E, Dixon A, Hill D, McCord J, Strynar MJ, Ford J, Gray LE. Cumulative maternal and neonatal effects of combined exposure to a mixture of perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) during pregnancy in the Sprague-Dawley rat. ENVIRONMENT INTERNATIONAL 2022; 170:107631. [PMID: 36402036 PMCID: PMC9944680 DOI: 10.1016/j.envint.2022.107631] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/29/2022] [Accepted: 11/10/2022] [Indexed: 05/10/2023]
Abstract
Globally, biomonitoring data demonstrate virtually all humans carry residues of multiple per- and polyfluoroalkyl substances (PFAS). Despite pervasive co-exposure, limited mixtures-based in vivo PFAS toxicity research has been conducted. Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) are commonly detected PFAS in human and environmental samples and both produce adverse effects in laboratory animal studies, including maternal and offspring effects when orally administered during pregnancy and lactation. To evaluate the effects of combined exposure to PFOA and PFOS, we orally exposed pregnant Sprague-Dawley rats from gestation day 8 (GD8) to postnatal day 2 (PND2) to PFOA (10-250 mg/kg/d) or PFOS (0.1-5 mg/kg/d) individually to characterize effects and dose response curve parameters, followed by a variable-ratio mixture experiment with a constant dose of PFOS (2 mg/kg/d) mixed with increasing doses of PFOA (3-80 mg/kg/d). The mixture study design was intended to: 1) shift the PFOA dose response curves for endpoints shared with PFOS, 2) allow comparison of dose addition (DA) and response addition (RA) model predictions, 3) conduct relative potency factor (RPF) analysis for multiple endpoints, and 4) avoid overt maternal toxicity. Maternal serum and liver concentrations of PFOA and PFOS were consistent between the individual chemical and mixture experiments. Combined exposure with PFOS significantly shifted the PFOA dose response curves towards effects at lower doses compared to PFOA-only exposure for multiple endpoints and these effects were well predicted by dose addition. For endpoints amenable to mixture model analyses, DA produced equivalent or better estimates of observed data than RA. All endpoints evaluated were accurately predicted by RPF and DA approaches except for maternal gestational weight gain, which produced less-than-additive results in the mixture. Data support the hypothesis of cumulative effects on shared endpoints from PFOA and PFOS co-exposure and dose additive approaches for predictive estimates of mixture effects.
Collapse
Affiliation(s)
- Justin M Conley
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Christy S Lambright
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Nicola Evans
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Elizabeth Medlock-Kakaley
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Aaron Dixon
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Donna Hill
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - James McCord
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Environmental Measurement and Modeling, Research Triangle Park, NC, USA.
| | - Mark J Strynar
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Environmental Measurement and Modeling, Research Triangle Park, NC, USA.
| | - Jermaine Ford
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA.
| | - L Earl Gray
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| |
Collapse
|
13
|
Zhang S, Lei X, Zhang Y, Shi R, Zhang Q, Gao Y, Yuan T, Li J, Tian Y. Prenatal exposure to per- and polyfluoroalkyl substances and childhood adiposity at 7 years of age. CHEMOSPHERE 2022; 307:136077. [PMID: 36002061 DOI: 10.1016/j.chemosphere.2022.136077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND An increasing number of studies have reported that prenatal per- and polyfluoroalkyl substances (PFAS) exposure may increase childhood adiposity. However, limited data is available in China, and the overall effects of PFAS mixture remain unclear. OBJECTIVE To examine the association of prenatal exposure to individual PFAS and their mixture with childhood adiposity at 7 years of age. METHODS A total of 206 mother-infant pairs were recruited from the Laizhou Wan (Bay) Birth Cohort in China between 2010 and 2013. Ten PFAS were measured in maternal serum. The measurements of fat mass, body fat percentage, body mass index, waist circumference, waist-to-height ratio and overweight/obesity were used to assess adiposity in children aged 7. We fitted logistic regression, linear regression and weighted quantile sum (WQS) regression models to estimate the association of prenatal exposure to individual PFAS and their mixture with childhood adiposity. RESULTS We found negative associations of perfluoroheptanoic acid (PFHpA) and perfluorooctane sulfonamide (PFOSA) exposure with adiposity measurements in all children. The result from the WQS model consistently revealed that the PFAS mixture was inversely related to adiposity measurements. Each quartile increase of the PFAS mixture was associated with a 1.14 kg decrease (95% CI: -2.27, -0.02) in fat mass and a 2.32% decrease (95% CI: -4.51, -0.14) in body fat. Moreover, significant sex differences were found. PFAS mixture was negatively associated with five adiposity measurements in boys, but positively associated with all adiposity measurements except body fat percentage in girls. PFOSA, PFHpA and perfluorobutanesulfonate (PFBS) with weights >0.300 were the main contributors to the overall effects observed among all children, boys and girls, respectively. CONCLUSION This study suggests potential sex-specific associations of prenatal exposure to individual PFAS and their mixture with childhood adiposity, with the observed relationship being negative for boys but positive for girls.
Collapse
Affiliation(s)
- Shanyu Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoning Lei
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Shi
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianlong Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gao
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Yuan
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jiong Li
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Clinical Epidemiology-Department of Clinical Medicine, Arhus University Hospital, Aarhus, Denmark
| | - Ying Tian
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Gray LE, Conley JM, Lambright CS, Furr JR. In utero exposure to a mixture of the perfluoroalkyl-isopropyl pesticide pyrifluquinazon with dibutyl phthalate cumulatively disrupts male rat reproductive development via different mechanisms of action. Toxicol Sci 2022; 188:234-247. [PMID: 35642937 PMCID: PMC10269475 DOI: 10.1093/toxsci/kfac059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Administration of individual chemicals and mixtures during sexual differentiation that disrupt the androgen signaling pathway can induce reproductive abnormalities in male rats. In the current study, we co-administered the heptafluoroisopropyl pesticide pyrifluquinazon (PFQ), and dibutyl phthalate (DBP) to pregnant rats during sexual differentiation of the reproductive tract. Both chemicals have been shown to disrupt reproductive tract differentiation in a dose-related manner reducing male anogenital distance (AGD), permanently reducing androgen-dependent tissue weights and sperm counts, and inducing reproductive malformations in male offspring, albeit by different mechanisms of action that converge downstream in the androgen signaling pathway on a common key event. Rats were orally dosed from gestation days 14-18 with dilutions of PFQ and DBP at 0, 12.5, 25, 50, 75 and 100% of the top dose (100 mg/kg PFQ and 750 mg/kg DBP). The mixture ratio was selected such that each chemical would contribute equally to multiple effects on the male offspring reproductive tract and the dose range was designed to determine if the mixture produced additive effects predicted by dose addition or response addition models, or whether significant interactions occurred. Observed data were compared to dose and response addition model predictions. As hypothesized, the mixture reduced F1 male AGD, reproductive organ weights and sperm counts and induced hypospadias with dose addition consistently providing a better prediction of the observed effects than response addition. These results support our hypothesis that chemicals that disrupt the androgen signaling pathway induce dose-additive male reproductive abnormalities regardless of the specific mechanism of action.
Collapse
Affiliation(s)
- L Earl Gray
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, Research Triangle Park, 27711 North Carolina
| | - Justin M Conley
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, Research Triangle Park, 27711 North Carolina
| | - Christy S Lambright
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, Research Triangle Park, 27711 North Carolina
| | - Johnathan R Furr
- Inotiv, 13 Firstfield Road, Suite 110, Gaithersburg, Maryland, 20878
| |
Collapse
|
15
|
Wang P, Liu D, Yan S, Cui J, Liang Y, Ren S. Adverse Effects of Perfluorooctane Sulfonate on the Liver and Relevant Mechanisms. TOXICS 2022; 10:toxics10050265. [PMID: 35622678 PMCID: PMC9144769 DOI: 10.3390/toxics10050265] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent, widely present organic pollutant. PFOS can enter the human body through drinking water, ingestion of food, contact with utensils containing PFOS, and occupational exposure to PFOS, and can have adverse effects on human health. Increasing research shows that the liver is the major target of PFOS, and that PFOS can damage liver tissue and disrupt its function; however, the exact mechanisms remain unclear. In this study, we reviewed the adverse effects of PFOS on liver tissue and cells, as well as on liver function, to provide a reference for subsequent studies related to the toxicity of PFOS and liver injury caused by PFOS.
Collapse
|
16
|
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: 151] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
|
17
|
Liu H, Sun W, Zhou Y, Griffin N, Faulkner S, Wang L. iTRAQ-Based Quantitative Proteomics Analysis of Sprague-Dawley Rats Liver Reveals Perfluorooctanoic acid-induced Lipid Metabolism and Urea Cycle Dysfunction. Toxicol Lett 2021; 357:20-32. [PMID: 34958885 DOI: 10.1016/j.toxlet.2021.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 11/27/2021] [Accepted: 12/17/2021] [Indexed: 12/28/2022]
Abstract
Perfluorooctanoic acid (PFOA) is a typical C8 representative compound of perfluoroalkyl and polyfluoroalkyl substances (PFAS) widely used in industrial and domestic products. It is a persistent organic pollutant found in the environment as well as in the tissues of humans and wildlife. Despite emerging scientific and public interest, the precise mechanisms of PFOA toxicity remain unclear. In this study, male rats were exposed to 1.25, 5, and 20 mg PFOA/kg body weight/day for 14 days by gavage; food intake and bodyweight changes were recorded every day. After 14 days, blood was collected for sera biochemistry, livers were quickly stripped and weighed after execution. Part of the liver tissue was frozen by liquid nitrogen for iTRAQ-Based Quantitative Proteomics Analysis; and some was fixed in 4% paraformaldehyde (PFA) for histological section and hematoxylin-eosin (HE) staining. Urine samples were also collected and monitored by raising rats in metabolic cages. Real-time quantitative PCR and western blot was used to validate the proteomics assay after bioinformatics analysis. The results demonstrate that 20 mg/kg/d PFOA exposure cause body weight loss and significant liver swelling and reduced urea metabolism. The sera biochemistry assay shows that ALT, GGT, BILD and UREA levels have significant changes compared with normal control group and reference range of rat sera. The subsequent iTRAQ-based quantitative proteomics analysis of rat livers identified 3,327 non-redundant proteins of which 112 proteins were significantly upregulated and 80 proteins were downregulated. Gene ontology analysis revealed proteins are primarily involved in cellular, metabolic and single-organism processes. Among them, eight proteins (ACOX1, ACOX2, ACOX3, ACSL1, EHHADH, GOT2, MTOR and ACAA1) were related to oxidation of fatty acids and two proteins (ASS1 and CPS1) were found to be associated with urea cycle disorder. The downregulation of urea synthesis proteins ASS1 and CPS1 after exposure to PFOA was then confirmed through qPCR and western blot analysis. Together, these data demonstrate that PFOA exposure directly influences urea metabolism and provides insight into specific mechanisms of hepatotoxicity as a result of PFOA exposure.
Collapse
Affiliation(s)
- Hui Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, 233030, PR China.
| | - Weiqiang Sun
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, 233030, PR China
| | - Yongbing Zhou
- School of Public Health, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, 233030, PR China
| | - Nathan Griffin
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Sam Faulkner
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia
| | - Li Wang
- School of Public Health, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, 233030, PR China.
| |
Collapse
|
18
|
Merrill AK, Anderson T, Conrad K, Marvin E, James-Todd T, Cory-Slechta DA, Sobolewski M. Protracted Impairment of Maternal Metabolic Health in Mouse Dams Following Pregnancy Exposure to a Mixture of Low Dose Endocrine-Disrupting Chemicals, a Pilot Study. TOXICS 2021; 9:346. [PMID: 34941779 PMCID: PMC8706199 DOI: 10.3390/toxics9120346] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022]
Abstract
Pregnancy, a period of increased metabolic demands coordinated by fluctuating steroid hormones, is an understudied critical window of disease susceptibility for later-life maternal metabolic health. Epidemiological studies have identified associations between exposures to various endocrine-disrupting chemicals (EDCs) with an increased risk for metabolic syndrome, obesity, and diabetes. Whether such adverse outcomes would be heightened by concurrent exposures to multiple EDCs during pregnancy, consistent with the reality that human exposures are to EDC mixtures, was examined in the current pilot study. Mouse dams were orally exposed to relatively low doses of four EDCs: (atrazine (10 mg/kg), bisphenol-A (50 µg/kg), perfluorooctanoic acid (0.1 mg/kg), 2,3,7,8-tetrachlorodibenzo-p-dioxin (0.036 µg/kg)), or the combination (MIX), from gestational day 7 until birth or for an equivalent 12 days in non-pregnant females. Glucose intolerance, serum lipids, weight, and visceral adiposity were assessed six months later. MIX-exposed dams exhibited hyperglycemia with a persistent elevation in blood glucose two hours after glucose administration in a glucose tolerance test, whereas no such effects were observed in MIX-exposed non-pregnant females. Correspondingly, MIX dams showed elevated serum low-density lipoprotein (LDL). There were no statistically significant differences in weight or visceral adipose; MIX dams showed an average visceral adipose volume to body volume ratio of 0.09, while the vehicle dams had an average ratio of 0.07. Collectively, these findings provide biological plausibility for the epidemiological associations observed between EDC exposures during pregnancy and subsequent maternal metabolic dyshomeostasis, and proof of concept data that highlight the importance of considering complex EDC mixtures based of off common health outcomes, e.g., for increased risk for later-life maternal metabolic effects following pregnancy.
Collapse
Affiliation(s)
- Alyssa K. Merrill
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA; (A.K.M.); (T.A.); (K.C.); (E.M.); (D.A.C.-S.)
| | - Timothy Anderson
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA; (A.K.M.); (T.A.); (K.C.); (E.M.); (D.A.C.-S.)
| | - Katherine Conrad
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA; (A.K.M.); (T.A.); (K.C.); (E.M.); (D.A.C.-S.)
| | - Elena Marvin
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA; (A.K.M.); (T.A.); (K.C.); (E.M.); (D.A.C.-S.)
| | - Tamarra James-Todd
- Department of Environmental Health, Harvard University, Boston, MA 02115, USA;
| | - Deborah A. Cory-Slechta
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA; (A.K.M.); (T.A.); (K.C.); (E.M.); (D.A.C.-S.)
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA; (A.K.M.); (T.A.); (K.C.); (E.M.); (D.A.C.-S.)
| |
Collapse
|