1
|
Zhu L, Liu B, Hu Y, Wang M, Furtado JD, Rimm EB, Grandjean P, Sun Q. Per- and polyfluoroalkyl substances, apolipoproteins and the risk of coronary heart disease in US men and women. Environ Health 2024; 23:108. [PMID: 39627728 PMCID: PMC11613683 DOI: 10.1186/s12940-024-01147-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/22/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND Existing evidence for associations of per- and polyfluoroalkyl substances (PFASs) with blood lipids, lipoproteins and apolipoproteins (apo), and coronary heart disease (CHD) risk is limited and inconsistent. This study aims to explore associations between plasma PFASs, blood lipoprotein subspecies defined by apolipoproteins, and CHD risk. METHODS A case-control study of CHD was conducted in the Health Professionals Follow-Up Study (HPFS) and Nurses' Health Study (NHS). Among participants initially free of cardiovascular disease at blood collection in 1994 (HPFS) or 1990 (NHS), 101 participants who developed non-fatal myocardial infarction or fatal CHD were identified and confirmed. A healthy control was matched to each case for age, smoking status, and date of blood draw. Plasma levels of perfluorohexane sulfonic acid (PFHxS), perfluorooctanoic acid (PFOA), total perfluorooctane sulfonic acid (PFOS), branched PFOS (brPFOS), linear PFOS (nPFOS), perfluorononanoic acid (PFNA), and perfluorodecanoic acid (PFDA) were measured. Conditional logistic regression and cubic spline regression models were used to examine associations between baseline PFASs and CHD risk. Linear regression models were applied to study PFAS associations with lipids and their subfractions. RESULTS After multivariate adjustments, total PFOS, brPFOS and nPFOS were significantly associated with increased risk of developing CHD, and HRs (95% CIs) per log(ng/mL) increment of PFASs were 3.66 (1.36-9.89), 3.68 (1.55-8.76), and 3.01 (1.16-7.86), respectively. Significant positive dose-response relationships were identified for these PFASs (Plinearity = 0.01, 0.002, 0.02, respectively). Other PFASs were not associated with CHD risk. PFNA and PFDA were positively associated with total apoE levels among HDL particles with or without apoC-III. No associations were observed for other PFASs with blood lipid subspecies. Blood lipid subfractions did not explain the association between PFOS and CHD risk. CONCLUSIONS Plasma PFOS and its isomers were positively associated with CHD risk. These findings suggest that PFOS exposure causes public health risks that are greater than hitherto believed.
Collapse
Grants
- HL035464, HL060712 NHLBI NIH HHS
- DK119268, DK126698, DK120870, DK129670 NIDDK NIH HHS
- ES022981, ES036206 NIEHS NIH HHS
- ES027706 NIEHS NIH HHS
- UL1 TR002541 NCATS NIH HHS
- U01CA167552, R01HL35464, UM1 CA186107, R01 HL034594 and R01 CA49449 NIH HHS
- R01 ES036206 NIEHS NIH HHS
- National Institute of Environmental Health Sciences
- National Heart, Lung, and Blood Institute
- National Institute of Diabetes and Digestive and Kidney Diseases
- NIH
- Harvard Catalyst
Collapse
Affiliation(s)
- Lu Zhu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Binkai Liu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Yang Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jeremy D Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Philippe Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Department of Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Robarts DR, Paine-Cabrera D, Kotulkar M, Venneman KK, Gunewardena S, Foquet L, Bial G, Apte U. Identifying novel mechanisms of per- and polyfluoroalkyl substance-induced hepatotoxicity using FRG humanized mice. Arch Toxicol 2024; 98:3063-3075. [PMID: 38782768 DOI: 10.1007/s00204-024-03789-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) such as perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) and perfluoro-2-methyl-3-oxahexanoic acid (GenX), the new replacement PFAS, are major environmental contaminants. In rodents, these PFAS induce several adverse effects on the liver, including increased proliferation, hepatomegaly, steatosis, hypercholesterolemia, nonalcoholic fatty liver disease and liver cancers. Activation of peroxisome proliferator receptor alpha by PFAS is considered the primary mechanism of action in rodent hepatocyte-induced proliferation. However, the human relevance of this mechanism is uncertain. We investigated human-relevant mechanisms of PFAS-induced adverse hepatic effects using FRG liver-chimeric humanized mice with livers repopulated with functional human hepatocytes. Male FRG humanized mice were treated with 0.067 mg/L of PFOA, 0.145 mg/L of PFOS, or 1 mg/L of GenX in drinking water for 28 days. PFOS caused a significant decrease in total serum cholesterol and LDL/VLDL, whereas GenX caused a significant elevation in LDL/VLDL with no change in total cholesterol and HDL. All three PFAS induced significant hepatocyte proliferation. RNA-sequencing with alignment to the human genome showed a total of 240, 162, and 619 differentially expressed genes after PFOA, PFOS, and GenX exposure, respectively. Upstream regulator analysis revealed that all three PFAS induced activation of p53 and inhibition of androgen receptor and NR1D1, a transcriptional repressor important in circadian rhythm. Further biochemical studies confirmed NR1D1 inhibition and in silico modeling indicated potential interaction of all three PFAS with the DNA-binding domain of NR1D1. In conclusion, our studies using FRG humanized mice have revealed new human-relevant molecular mechanisms of PFAS including their previously unknown effect on circadian rhythm.
Collapse
Affiliation(s)
- Dakota R Robarts
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Diego Paine-Cabrera
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Manasi Kotulkar
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Kaitlyn K Venneman
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Greg Bial
- Yecuris Corporation, Tualatin, OR, USA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA.
| |
Collapse
|
3
|
Kashobwe L, Sadrabadi F, Brunken L, Coelho ACMF, Sandanger TM, Braeuning A, Buhrke T, Öberg M, Hamers T, Leonards PEG. Legacy and alternative per- and polyfluoroalkyl substances (PFAS) alter the lipid profile of HepaRG cells. Toxicology 2024; 506:153862. [PMID: 38866127 DOI: 10.1016/j.tox.2024.153862] [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: 03/05/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals used in various industrial and consumer products. They have gained attention due to their ubiquitous occurrence in the environment and potential for adverse effects on human health, often linked to immune suppression, hepatotoxicity, and altered cholesterol metabolism. This study aimed to explore the impact of ten individual PFAS, 3 H-perfluoro-3-[(3-methoxypropoxy) propanoic acid] (PMPP/Adona), ammonium perfluoro-(2-methyl-3-oxahexanoate) (HFPO-DA/GenX), perfluorobutanoic acid (PFBA), perfluorobutanesulfonic acid (PFBS), perfluorodecanoic acid (PFDA), perfluorohexanoic acid (PFHxA), perfluorohexanesulfonate (PFHxS), perfluorononanoic acid (PFNA), perfluorooctanoic acid (PFOA), and perfluorooctanesulfonic acid (PFOS) on the lipid metabolism in human hepatocyte-like cells (HepaRG). These cells were exposed to different concentrations of PFAS ranging from 10 µM to 5000 µM. Lipids were extracted and analyzed using liquid chromatography coupled with mass spectrometry (LC- MS-QTOF). PFOS at 10 µM and PFOA at 25 µM increased the levels of ceramide (Cer), diacylglycerol (DAG), N-acylethanolamine (NAE), phosphatidylcholine (PC), and triacylglycerol (TAG) lipids, while PMPP/Adona, HFPO-DA/GenX, PFBA, PFBS, PFHxA, and PFHxS decreased the levels of these lipids. Furthermore, PFOA and PFOS markedly reduced the levels of palmitic acid (FA 16.0). The present study shows distinct concentration-dependent effects of PFAS on various lipid species, shedding light on the implications of PFAS for essential cellular functions. Our study revealed that the investigated legacy PFAS (PFOS, PFOA, PFBA, PFDA, PFHxA, PFHxS, and PFNA) and alternative PFAS (PMPP/Adona, HFPO-DA/GenX and PFBS) can potentially disrupt lipid homeostasis and metabolism in hepatic cells. This research offers a comprehensive insight into the impacts of legacy and alternative PFAS on lipid composition in HepaRG cells.
Collapse
Affiliation(s)
- Lackson Kashobwe
- Vrije Universiteit Amsterdam, Amsterdam Institute for Life and Environment (A-LIFE), De Boelelaan 1105, Amsterdam, Netherlands
| | - Faezeh Sadrabadi
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Lars Brunken
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ana Carolina M F Coelho
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Torkjel M Sandanger
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Thorsten Buhrke
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Mattias Öberg
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Timo Hamers
- Vrije Universiteit Amsterdam, Amsterdam Institute for Life and Environment (A-LIFE), De Boelelaan 1105, Amsterdam, Netherlands
| | - Pim E G Leonards
- Vrije Universiteit Amsterdam, Amsterdam Institute for Life and Environment (A-LIFE), De Boelelaan 1105, Amsterdam, Netherlands.
| |
Collapse
|
4
|
Ren W, Wang Z, Guo H, Gou Y, Dai J, Zhou X, Sheng N. GenX analogs exposure induced greater hepatotoxicity than GenX mainly via activation of PPARα pathway while caused hepatomegaly in the absence of PPARα in female mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123314. [PMID: 38218542 DOI: 10.1016/j.envpol.2024.123314] [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: 10/30/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Despite their use as substitutes for perfluorooctanoic acid, the potential toxicities of hexafluoropropylene oxide dimer acid (HFPO-DA, commercial name: GenX) and its analogs (PFDMOHxA, PFDMO2HpA, and PFDMO2OA) remain poorly understood. To assess the hepatotoxicity of these chemicals on females, each chemical was orally administered to female C57BL/6 mice at the dosage of 0.5 mg/kg/d for 28 d. The contribution of peroxisome proliferator-activated receptors (PPARα and γ) and other nuclear receptors involving in these toxic effects of GenX and its analogs were identified by employing two PPAR knockout mice (PPARα-/- and PPARγΔHep) in this study. Results showed that the hepatotoxicity of these chemicals increased in the order of GenX < PFDMOHxA < PFDMO2HpA < PFDMO2OA. The increases of relative liver weight and liver injury markers were significantly much lower in PPARα-/- mice than in PPARα+/+ mice after GenX analog exposure, while no significant differences were observed between PPARγΔHep and its corresponding wildtype groups (PPARγF/F mice), indicating that GenX analog induce hepatotoxicity mainly via PPARα instead of PPARγ. The PPARα-dependent complement pathways were inhibited in PFDMO2HpA and PFDMO2OA exposed PPARα+/+ mice, which might be responsible for the observed liver inflammation. In PPARα-/- mice, hepatomegaly and increased liver lipid content were observed in PFDMO2HpA and PFDMO2OA treated groups. The activated pregnane X receptor (PXR) and constitutive activated receptor (CAR) pathways in the liver of PPARα-/- mice, which were highlighted by bioinformatics analysis, provided a reasonable explanation for hepatomegaly in the absence of PPARα. Our results indicate that GenX analogs could induce more serious hepatotoxicity than GenX whether there is a PPARα receptor or not. These chemicals, especially PFDMO2HpA and PFDMO2OA, may not be appropriate PFOA alternatives.
Collapse
Affiliation(s)
- Wanlan Ren
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiru Wang
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Guo
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yong Gou
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xuming Zhou
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nan Sheng
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Annunziato M, Bashirova N, Eeza MNH, Lawson A, Fernandez-Lima F, Tose LV, Matysik J, Alia A, Berry JP. An Integrated Metabolomics-Based Model, and Identification of Potential Biomarkers, of Perfluorooctane Sulfonic Acid Toxicity in Zebrafish Embryos. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2024. [PMID: 38411227 DOI: 10.1002/etc.5824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/28/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024]
Abstract
Known for their high stability and surfactant properties, per- and polyfluoroalkyl substances (PFAS) have been widely used in a range of manufactured products. Despite being largely phased out due to concerns regarding their persistence, bioaccumulation, and toxicity, legacy PFAS such as perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid continue to persist at high levels in the environment, posing risks to aquatic organisms. We used high-resolution magic angle spinning nuclear magnetic resonance spectroscopy in intact zebrafish (Danio rerio) embryos to investigate the metabolic pathways altered by PFOS both before and after hatching (i.e., 24 and 72 h post fertilization [hpf], respectively). Assessment of embryotoxicity found embryo lethality in the parts-per-million range with no significant difference in mortality between the 24- and 72-hpf exposure groups. Metabolic profiling revealed mostly consistent changes between the two exposure groups, with altered metabolites generally associated with oxidative stress, lipid metabolism, energy production, and mitochondrial function, as well as specific targeting of the liver and central nervous system as key systems. These metabolic changes were further supported by analyses of tissue-specific production of reactive oxygen species, as well as nontargeted mass spectrometric lipid profiling. Our findings suggest that PFOS-induced metabolic changes in zebrafish embryos may be mediated through previously described interactions with regulatory and transcription factors leading to disruption of mitochondrial function and energy metabolism. The present study proposes a systems-level model of PFOS toxicity in early life stages of zebrafish, and also identifies potential biomarkers of effect and exposure for improved environmental biomonitoring. Environ Toxicol Chem 2024;00:1-19. © 2024 SETAC.
Collapse
Affiliation(s)
- Mark Annunziato
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| | - Narmin Bashirova
- Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - Muhamed N H Eeza
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Ariel Lawson
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - Francisco Fernandez-Lima
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| | - Lilian V Tose
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| | - Jörg Matysik
- Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - A Alia
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - John P Berry
- Institute of Environment, Florida International University, Miami, Florida, USA
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Science Institute, Florida International University, Miami, Florida, USA
| |
Collapse
|
6
|
Sabovic I, Lupo MG, Rossi I, Pedrucci F, Di Nisio A, Dall’Acqua S, Ferri N, Ferlin A, Foresta C, De Toni L. Legacy perfluoro-alkyl substances impair LDL-cholesterol uptake independently from PCSK9-function. Toxicol Rep 2023; 11:288-294. [PMID: 37818225 PMCID: PMC10560979 DOI: 10.1016/j.toxrep.2023.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023] Open
Abstract
Perfluoro-alkyl substances (PFAS) are pollutants, whose exposure was associated with altered levels of low-density lipoproteins (LDL) in humans. Here we investigated this clinical outcome in two groups of young male adults residing in areas of respectively low and high environmental exposure to perfluoro-octanoic-acid (PFOA). From the Regional Authority data on pollution areas, 38 not-exposed and 59 exposed age-matched participants were evaluated for serum levels of total cholesterol (Total-Chol), LDL-Chol, high-density lipoprotein cholesterol (HDL-Chol), triglycerides (Tgl) and chromatography quantified PFOA. Human hepato-carcinoma cell line HepG2 was exposed to PFOA or perfluoro-octane-sulfonate (PFOS), as legacy PFAAs, and C6O4 as new generation compound. Fluorimetry was used to evaluate the cell-uptake of labelled-LDL. Proprotein Convertase Subtilisin/Kexin 9 (PCSK9)-mediated LDL-receptor (LDL-R) degradation and sub-cellular localization of LDL-R were evaluated by western blot analysis. Serum levels of PFOA, were positively and significantly correlated with Total-Chol (ρ = 0.312, P = 0.002), LDL-Chol (ρ = 0.333, P = 0.001) and Tgl (ρ = 0.375, P < 0.001). Participants with high serum LDL-Chol and Tgl levels, according to the cardiovascular risk, were more prevalent in exposed compared to not-exposed subjects (respectively: 23.7% vs 5.3%, P = 0.023 and 18,6% vs 0%, P = 0.006). Exposure of HepG2 cells to PFOA or C6O4 100 ng/mL was associated with a significantly lower LDL uptake than controls but no major impact of any PFAAs on PCSK9-mediated LDL-R degradation was observed. Compared to controls, exposure to PFAS showed an unbalanced LDL-R partition between membrane and cytoplasm. Endocytosis inducer sphingosine restored LDL-R partition only in samples exposed to C6O4. These data suggest a novel endocytosis-based mechanism of altered lipid trafficking associated with the exposure to legacy PFAS.
Collapse
Affiliation(s)
- Iva Sabovic
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | | | - Ilaria Rossi
- Department of Medicine, University of Padova, Padova, Italy
| | - Federica Pedrucci
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Andrea Di Nisio
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Stefano Dall’Acqua
- Department of Pharmacological Sciences, University of Padova, Padova, Italy
| | - Nicola Ferri
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Alberto Ferlin
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Carlo Foresta
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Luca De Toni
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
7
|
Yang J, Zhang K, Shen C, Tang P, Tu S, Li J, Chen L, Yang W. The Association of Hypertension with Perfluoroalkyl and Polyfluoroalkyl Substances. Int Heart J 2023; 64:1079-1087. [PMID: 37967990 DOI: 10.1536/ihj.23-036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Perfluoroalkyl and polyfluoroalkyl substance (PFAS) is a large group of fluorinated synthetic chemicals, e.g., perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS), perfluorohexanesulfonic acid (PFHxS), perfluorodecanoic acid (PFDA), and perfluorononanoic acid (PFNA). Many epidemiological studies have found that PFAS exposure is associated with hypertension risk, but others possess a different opinion. Overall, the relationship between PFASs and hypertension risk remains controversial. We sought to conduct a systematic review and meta-analysis to clarify the association between PFAS exposure and human risk of hypertension.We conducted a meta-analysis based on population-involving studies published from 1975 to 2023, which we collected from Web of Science, PubMed, and Embase databases. The odds ratio (OR) and standardized mean difference (SMD), with their 95% confidence interval (CI), were used to assess the risk of hypertension with PFAS exposure. The statistical heterogeneity among studies was assessed with the Q-test and I2 statistics. Research publications related to our meta-analysis topic were systematically reviewed.Fourteen studies involving 71,663 participants, in which 26,281 suffered hypertension, met the inclusion criteria. Our analyses suggest that exposure to general PFAS (OR = 1.09, 95% CI = 1.04-1.14) or PFOS (OR = 1.17, 95% CI = 1.05-1.30) is associated with hypertension risk. Specifically, elevated levels of general PFAS (SMD = 0.25, 95% CI = 0.08-0.42), PFHxS (SMD = 0.17, 95% CI = 0.07-0.27), and PFDA (SMD = 0.08, 95% CI = 0.02-0.13) are associated with a high risk of hypertension.Our meta-analysis indicates that PFAS exposure is a risk factor for hypertension, and increased hypertension risk is associated with higher PFAS levels. Further study may eventually provide a better and more comprehensive elucidation of the potential mechanism of this association.
Collapse
Affiliation(s)
- Jingxuan Yang
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| | - Kui Zhang
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| | - Chengchen Shen
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| | - Peng Tang
- Department of Crop Science, College of Agriculture, Shanxi Agricultural University
| | - Shasha Tu
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| | - Jiangyun Li
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| | - Li Chen
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| | - Wenxing Yang
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University
| |
Collapse
|
8
|
Robarts DR, Dai J, Lau C, Apte U, Corton JC. Hepatic Transcriptome Comparative In Silico Analysis Reveals Similar Pathways and Targets Altered by Legacy and Alternative Per- and Polyfluoroalkyl Substances in Mice. TOXICS 2023; 11:963. [PMID: 38133364 PMCID: PMC10748317 DOI: 10.3390/toxics11120963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
Per- and poly-fluoroalkyl substances (PFAS) are a large class of fluorinated carbon chains that include legacy PFAS, such as perfluorooctane sulfonate (PFOS), perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), and perfluorohexane sulfonate (PFHxS). These compounds induce adverse health effects, including hepatotoxicity. Potential alternatives to the legacy PFAS (HFPO-DA (GenX), HFPO4, HFPO-TA, F-53B, 6:2 FTSA, and 6:2 FTCA), as well as a byproduct of PFAS manufacturing (Nafion BP2), are increasingly being found in the environment. The potential hazards of these new alternatives are less well known. To better understand the diversity of molecular targets of the PFAS, we performed a comparative toxicogenomics analysis of the gene expression changes in the livers of mice exposed to these PFAS, and compared these to five activators of PPARα, a common target of many PFAS. Using hierarchical clustering, pathway analysis, and predictive biomarkers, we found that most of the alternative PFAS modulate molecular targets that overlap with legacy PFAS. Only three of the 11 PFAS tested did not appreciably activate PPARα (Nafion BP2, 6:2 FTSA, and 6:2 FTCA). Predictive biomarkers showed that most PFAS (PFHxS, PFOA, PFOS, PFNA, HFPO-TA, F-53B, HFPO4, Nafion BP2) activated CAR. PFNA, PFHxS, PFOA, PFOS, HFPO4, HFPO-TA, F-53B, Nafion BP2, and 6:2 FTSA suppressed STAT5b, activated NRF2, and activated SREBP. There was no apparent relationship between the length of the carbon chain, type of head group, or number of ether linkages and the transcriptomic changes. This work highlights the similarities in molecular targets between the legacy and alternative PFAS.
Collapse
Affiliation(s)
- Dakota R. Robarts
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Christopher Lau
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - J. Christopher Corton
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
9
|
Hu L, Xu T, Wang X, Qian M, Jin Y. Exposure to the fungicide prothioconazole and its metabolite prothioconazole-desthio induced hepatic metabolism disorder and oxidative stress in mice. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 193:105452. [PMID: 37248020 DOI: 10.1016/j.pestbp.2023.105452] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023]
Abstract
Prothioconazole (PTC), as a popular triazole fungicide, with its main metabolite prothioconazole desthio (PTC-d), have attracted widespread concern due to their widely use and toxicological effects on non-target organisms. However, toxic effects of study analyzed PTC and PTC-d on the hepatic metabolism of mammalian still remains unclear. In this study, we conducted the study of the C57BL/6 mice which oral exposure to 30 mg/kg PTC and PTC-d via metabolomic analysis. In the liver, the metabolomics profile unveiled that exposure to 30 mg/kg PTC and PTC-d led to significantly altered 13 and 28 metabolites respectively, with 6 metabolites in common including significant decreased d-Fructose, Glutathione, showing the change of carbohydrate, lipid and amino acid metabolism. Via the further exploration of genes related to hepatic glycolipid metabolism and the biomarkers of oxidative stress, we found that liver was potentially damaged after exposure to 5 and 30 mg/kg PTC and PTC-d. Particularly, it was proved that PTC-d caused more adverse effect than its parent compound PTC on hepatotoxicity, and high concentration PTC or PTC-d exposure is more harmful than low concentration exposure.
Collapse
Affiliation(s)
- Lingyu Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy, Zhejiang Shuren University, Hangzhou 310015, China
| | - Ting Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaofang Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy, Zhejiang Shuren University, Hangzhou 310015, China
| | - Mingrong Qian
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
10
|
Kim OJ, Kim S, Park EY, Oh JK, Jung SK, Park S, Hong S, Jeon HL, Kim HJ, Park B, Park B, Kim S, Kim B. Exposure to serum perfluoroalkyl substances and biomarkers of liver function: The Korean national environmental health survey 2015-2017. CHEMOSPHERE 2023; 322:138208. [PMID: 36822523 DOI: 10.1016/j.chemosphere.2023.138208] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Exposure to perfluoroalkyl substances (PFAS) may increase the risk of liver disease by disrupting cholesterol and lipid synthesis/metabolism, leading to higher liver-enzyme concentrations. However, most studies assessing association between PFAS and liver enzymes focused on individual PFAS. Moreover, PFAS concentrations differ based on sex and obesity status, and it remains unclear whether these factors affect associations with liver function. Therefore, we examined the association between exposure to both individual and combined PFAS and liver-function biomarkers and assessed sex and obesity as effect modifiers in Korean adults. METHODS We measured serum concentrations of the five most abundant PFAS (PFOA, PFOS, PFHxS, PFDA, PFNA) and three liver enzymes (alanine transaminase [ALT], aspartate aminotransferase [AST], γ-glutamyl transferase [GGT]) in 1404 adults from the Korean National Environmental Health Survey Cycle 3, 2015-2017. We used linear regression to evaluate associations between individual PFAS and liver-function biomarkers, assessing sex and obesity as possible effect modifiers, and performed Bayesian kernel machine regression and quantile g-computation to evaluate the overall effect of PFAS mixture on biomarkers of liver function. RESULTS Among 1404 Korean adults, all five PFAS were detected. Geometric mean concentration was highest for PFOS (16.11 μg/L), followed by PFOA (5.83 μg/L), PFHxS (2.21 μg/L), PFNA (2.03 μg/L), and PFDA (1.06 μg/L). In multivariable linear regression, all PFAS were positively associated with ALT, AST, and GGT; 2-fold increase in each PFAS was associated with 3.4-8.6% higher ALT, 2.4-4.6% higher AST, and 4.6-11.1% higher GGT (all p < 0.05). Positive associations for PFOA, PFDA, and PFNA with AST were stronger in men, and positive associations for PFOS with ALT and GGT were stronger in women. Compared to obese participants, nonobese participants had higher average percent changes in each enzyme, particularly GGT, when individual PFAS concentration doubled. Additionally, increased exposure to PFAS mixtures was associated with higher ALT, AST, and GGT. In quantile g-computations, simultaneous quartile increase in all PFAS was significantly associated with 6.9% (95%CI: 3.7, 10.2) higher ALT, 4.5% (95%CI: 2.4, 6.6) higher AST, and 8.3% (95%CI: 3.7, 13.1) higher GGT levels, on average. CONCLUSIONS Exposure to individual and combined PFAS is associated with higher liver enzymes in Korean adults, providing additional evidence for the association between PFAS exposure and risk of liver disease.
Collapse
Affiliation(s)
- Ok-Jin Kim
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, 22689, Republic of Korea
| | - Seyoung Kim
- National Cancer Control Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Eun Young Park
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jin Kyoung Oh
- National Cancer Control Institute, National Cancer Center, Goyang, 10408, Republic of Korea; Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Sun Kyoung Jung
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, 22689, Republic of Korea
| | - Soyoung Park
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, 22689, Republic of Korea
| | - Sooyeon Hong
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, 22689, Republic of Korea
| | - Hye Li Jeon
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, 22689, Republic of Korea
| | - Hyun-Jin Kim
- National Cancer Control Institute, National Cancer Center, Goyang, 10408, Republic of Korea; Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Bohyun Park
- National Cancer Control Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Bomi Park
- Department of Preventive Medicine, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Suejin Kim
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, 22689, Republic of Korea.
| | - Byungmi Kim
- National Cancer Control Institute, National Cancer Center, Goyang, 10408, Republic of Korea; Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea.
| |
Collapse
|
11
|
Liu B, Zhu L, Wang M, Sun Q. Associations between Per- and Polyfluoroalkyl Substances Exposures and Blood Lipid Levels among Adults-A Meta-Analysis. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:56001. [PMID: 37141244 PMCID: PMC10159273 DOI: 10.1289/ehp11840] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
BACKGROUND Associations between per- and polyfluoroalkyl substances (PFAS) and blood lipid levels in humans were mixed. OBJECTIVES The objective of this meta-analysis was to summarize associations between PFAS and blood lipids in adults. METHODS A literature search was conducted on PubMed and Web of Science for articles published through 13 May 2022 that examined associations between PFAS and blood lipids, including total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triacylglycerols (TGs). Inclusion criteria included the presence of associations between five PFAS (PFOA, PFOS, PFHxS, PFDA, and PFNA) and four blood lipid measures (TC, HDL-C, LDL-C, and TGs) in adults. Data on study characteristics and PFAS-lipid associations were extracted. Assessments of individual study quality were performed. Associations of changes of blood lipid levels corresponding to 1 interquartile range (IQR)-unit increase of blood PFAS levels were pooled using random effects models. Dose-response relationships were examined. RESULTS Twenty-nine publications were included in the present analyses. Every IQR increase of PFOA was significantly associated with a 2.1 -mg / dL increase in TC (95% CI: 1.2, 3.0), a 1.3 -mg / dL increase in TGs (95% CI: 0.1, 2.4), and a 1.4 -mg / dL increase in LDL-C (95% CI: 0.6, 2.2). PFOS was also significantly associated with TC and LDL-C levels, and the corresponding values were 2.6 (95% CI: 1.5, 3.6) and 1.9 (95% CI: 0.9, 3.0), respectively. Associations of PFOS and PFOA with HDL-C levels were largely null. For minor PFAS species, PFHxS was significantly associated with higher levels of HDL-C [0.8 (95% CI: 0.5, 1.2)]. Inverse associations were observed between PFDA and TGs [- 5.0 (95% CI: - 8.1 , - 1.9 )] and between PFNA and TGs [- 1.7 (95% CI: - 3.5 , - 0.02 )], whereas a positive association was observed between PFDA and HDL-C [1.4 (95% CI: 0.1, 2.7)]. Nonsignificant nonlinear dose-response relationships were identified for associations of PFOA and PFOS with certain blood lipids. DISCUSSION PFOA and PFOS were significantly associated with TC and LDL-C levels in adults. Whether these findings may translate into an elevated cardiovascular disease risk associated with PFAS exposure warrants further investigation. https://doi.org/10.1289/EHP11840.
Collapse
Affiliation(s)
- Binkai Liu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Lu Zhu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Qi Sun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Robarts DR, Paine-Cabrera D, Kotulkar M, Venneman KK, Gunewardena S, Corton JC, Lau C, Foquet L, Bial G, Apte U. Identifying Human Specific Adverse Outcome Pathways of Per- and Polyfluoroalkyl Substances Using Liver-Chimeric Humanized Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526711. [PMID: 36778348 PMCID: PMC9915685 DOI: 10.1101/2023.02.01.526711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Per- and polyfluoroalkyl substances (PFAS) are persistent organic pollutants with myriad adverse effects. While perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) are the most common contaminants, levels of replacement PFAS, such as perfluoro-2-methyl-3-oxahexanoic acid (GenX), are increasing. In rodents, PFOA, PFOS, and GenX have several adverse effects on the liver, including nonalcoholic fatty liver disease. Objective We aimed to determine human-relevant mechanisms of PFAS induced adverse hepatic effects using FRG liver-chimeric humanized mice with livers repopulated with functional human hepatocytes. Methods Male humanized mice were treated with 0.067 mg/L of PFOA, 0.145 mg/L of PFOS, or 1 mg/L of GenX in drinking water for 28 days. Liver and serum were collected for pathology and clinical chemistry, respectively. RNA-sequencing coupled with pathway analysis was used to determine molecular mechanisms. Results PFOS caused a significant decrease in total serum cholesterol and LDL/VLDL, whereas GenX caused a significant elevation in LDL/VLDL with no change in total cholesterol and HDL. PFOA had no significant changes in serum LDL/VLDL and total cholesterol. All three PFAS induced significant hepatocyte proliferation. RNA-sequencing with alignment to the human genome showed a total of 240, 162, and 619 differentially expressed genes after PFOA, PFOS, and GenX exposure, respectively. Upstream regulator analysis revealed inhibition of NR1D1, a transcriptional repressor important in circadian rhythm, as the major common molecular change in all PFAS treatments. PFAS treated mice had significant nuclear localization of NR1D1. In silico modeling showed PFOA, PFOS, and GenX potentially interact with the DNA-binding domain of NR1D1. Discussion These data implicate PFAS in circadian rhythm disruption via inhibition of NR1D1. These studies show that FRG humanized mice are a useful tool for studying the adverse outcome pathways of environmental pollutants on human hepatocytes in situ.
Collapse
Affiliation(s)
- Dakota R. Robarts
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Diego Paine-Cabrera
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Manasi Kotulkar
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Kaitlyn K. Venneman
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - J. Christopher Corton
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC
| | - Christopher Lau
- Center for Public Health and Environmental Assessment, Office of Research and Development, US EPA, Research Triangle Park, NC
| | | | | | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
13
|
Nilsson S, Smurthwaite K, Aylward LL, Kay M, Toms LM, King L, Marrington S, Kirk MD, Mueller JF, Bräunig J. Associations between serum perfluoroalkyl acid (PFAA) concentrations and health related biomarkers in firefighters. ENVIRONMENTAL RESEARCH 2022; 215:114370. [PMID: 36174755 DOI: 10.1016/j.envres.2022.114370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Firefighters who used aqueous film forming foam in the past have experienced elevated exposures to perfluoroalkyl acids (PFAAs). The objective of this study was to examine the associations between clinical chemistry endpoints and serum concentrations of perfluorooctanoic acid (PFOA), perfluorohexane sulfonate (PFHxS), perfluoroheptane sulfonate (PFHpS) and perfluorooctane sulfonate (PFOS) in firefighters. Multiple linear regression was used to assess relationships between PFAA serum concentrations and biochemical markers for cardiovascular disease, kidney-, liver- and thyroid function, in a cross-sectional survey of 783 firefighters with elevated levels of PFHxS, PFHpS and PFOS in relation to the most recently reported levels in the general Australian population. Linear logistic regression was used to assess the odds ratios for selected self-reported health outcomes. Repeated measures linear mixed models were further used to assess relationships between PFAAs and biomarkers for cardiovascular disease and kidney function longitudinally in a subset of the firefighters (n = 130) where serum measurements were available from two timepoints, five years apart. In the cross-sectional analysis, higher levels of all PFAAs were significantly associated with higher levels of biomarkers for cardiovascular disease (total-cholesterol, and LDL-cholesterol). For example, doubling in PFOS serum concentration were associated with increases in total cholesterol (β:0.111, 95% confidence interval (95%CI): 0.026, 0.195 mmol/L) and LDL-cholesterol (β: 0.104, 95%CI:0.03, 0.178 mmol/L). Doubling in PFOA concentration, despite not being elevated in the study population, were additionally positively associated with kidney function marker urate (e.g., β: 0.010, 95%CI; 0.004, 0.016 mmol/L) and thyroid function marker TSH (e.g., β: 0.087, 95%CI: 0.014, 0.161 mIU/L). PFAAs were not associated with any assessed self-reported health conditions. No significant relationships were observed in the longitudinal analysis. Findings support previous studies, particularly on the association between PFAAs and serum lipids.
Collapse
Affiliation(s)
- Sandra Nilsson
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, 20 Cornwall Street, Woolloongabba, 4102, QLD, Australia.
| | - Kayla Smurthwaite
- National Centre for Epidemiology and Population Health, The Australian National University, Cnr of Eggleston and Mills Roads Acton 2600, Australia
| | - Lesa L Aylward
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, 20 Cornwall Street, Woolloongabba, 4102, QLD, Australia; Summit Toxicology, LLP, La Quinta, 92253, CA, USA
| | - Margaret Kay
- General Practice Clinical Unit, Faculty of Medicine, The University of Queensland, Health Sciences Building, RBWH Complex, Herston, 4029, QLD, Australia
| | - Leisa-Maree Toms
- School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Musk Avenue, Kelvin Grove, 4059, QLD, Australia
| | - Leisa King
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, 20 Cornwall Street, Woolloongabba, 4102, QLD, Australia
| | - Shelby Marrington
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, 20 Cornwall Street, Woolloongabba, 4102, QLD, Australia
| | - Martyn D Kirk
- National Centre for Epidemiology and Population Health, The Australian National University, Cnr of Eggleston and Mills Roads Acton 2600, Australia
| | - Jochen F Mueller
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, 20 Cornwall Street, Woolloongabba, 4102, QLD, Australia
| | - Jennifer Bräunig
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, 20 Cornwall Street, Woolloongabba, 4102, QLD, Australia
| |
Collapse
|
14
|
Feng Z, McLamb F, Vu JP, Gong S, Gersberg RM, Bozinovic G. Physiological and transcriptomic effects of hexafluoropropylene oxide dimer acid in Caenorhabditis elegans during development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114047. [PMID: 36075119 DOI: 10.1016/j.ecoenv.2022.114047] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are chemicals resistant to degradation. While such a feature is desirable in consumer and industrial products, some PFAS, including perfluorooctanoic acid (PFOA), are toxic and bioaccumulate. Hexafluoropropylene oxide dimer acid (HFPO-DA), an emerging PFAS developed to replace PFOA, has not been extensively studied. To evaluate the potential toxicity of HFPO-DA with a cost- and time-efficient approach, we exposed C. elegans larvae for 48 h to 4 × 10-9-4 g/L HFPO-DA in liquid media and measured developmental, behavioral, locomotor, and transcriptional effects at various exposure levels. Worms exposed to 1.5-4 g/L HFPO-DA were developmentally delayed, and progeny production was significantly delayed (p < 0.05) in worms exposed to 2-4 g/L HFPO-DA. Statistically significant differential gene expression was identified in all fourteen HFPO-DA exposure groups ranging from 1.25 × 10-5 to 4 g/L, except for 6.25 × 10-5 g/L. Among 10298 analyzed genes, 2624 differentially expressed genes (DEGs) were identified in the developmentally delayed 4 g/L group only, and 78 genes were differentially expressed in at least one of the thirteen groups testing 1.25 × 10-5-2 g/L HFPO-DA exposures. Genes encoding for detoxification enzymes including cytochrome P450 and UDP glucuronosyltransferases were upregulated in 0.25-4 g/L acute exposure groups. DEGs were also identified in lower exposure level groups, though they did not share biological functions except for six ribosomal protein-coding genes. While our transcriptional data is inconclusive to infer mechanisms of toxicity, the significant gene expression differences at 1.25 × 10-5 g/L, the lowest concentration tested for transcriptional changes, calls for further targeted analyses of low-dose HFPO-DA exposure effects.
Collapse
Affiliation(s)
- Zuying Feng
- Boz Life Science Research and Teaching Institute, 3030 Bunker Hill Street, San Diego, CA, USA; School of Public Health, San Diego State University, 5500 Campanile Drive, San Diego, CA, USA.
| | - Flannery McLamb
- Boz Life Science Research and Teaching Institute, 3030 Bunker Hill Street, San Diego, CA, USA; Division of Extended Studies, University of California San Diego, 9600N. Torrey Pines Road, La Jolla, CA, USA.
| | - Jeanne P Vu
- Boz Life Science Research and Teaching Institute, 3030 Bunker Hill Street, San Diego, CA, USA; School of Public Health, San Diego State University, 5500 Campanile Drive, San Diego, CA, USA; Division of Extended Studies, University of California San Diego, 9600N. Torrey Pines Road, La Jolla, CA, USA.
| | - Sylvia Gong
- Boz Life Science Research and Teaching Institute, 3030 Bunker Hill Street, San Diego, CA, USA; School of Public Health, San Diego State University, 5500 Campanile Drive, San Diego, CA, USA; Division of Extended Studies, University of California San Diego, 9600N. Torrey Pines Road, La Jolla, CA, USA.
| | - Richard M Gersberg
- School of Public Health, San Diego State University, 5500 Campanile Drive, San Diego, CA, USA.
| | - Goran Bozinovic
- Boz Life Science Research and Teaching Institute, 3030 Bunker Hill Street, San Diego, CA, USA; School of Public Health, San Diego State University, 5500 Campanile Drive, San Diego, CA, USA; Division of Biological Sciences, University of California San Diego, 9500 Gilman Dr., La Jolla, CA, USA.
| |
Collapse
|
15
|
Davidsen N, Ramhøj L, Lykkebo CA, Kugathas I, Poulsen R, Rosenmai AK, Evrard B, Darde TA, Axelstad M, Bahl MI, Hansen M, Chalmel F, Licht TR, Svingen T. PFOS-induced thyroid hormone system disrupted rats display organ-specific changes in their transcriptomes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119340. [PMID: 35460815 DOI: 10.1016/j.envpol.2022.119340] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 06/14/2023]
Abstract
Perfluorooctanesulfonic acid (PFOS) is a persistent anthropogenic chemical that can affect the thyroid hormone system in humans and animals. In adults, thyroid hormones (THs) are regulated by the hypothalamic-pituitary-thyroid (HPT) axis, but also by organs such as the liver and potentially the gut microbiota. PFOS and other xenobiotics can therefore disrupt the TH system at various locations and through different mechanisms. To start addressing this, we exposed adult male rats to 3 mg PFOS/kg/day for 7 days and analysed effects on multiple organs and pathways simultaneously by transcriptomics. This included four primary organs involved in TH regulation, namely hypothalamus, pituitary, thyroid, and liver. To investigate a potential role of the gut microbiota in thyroid hormone regulation, two additional groups of animals were dosed with the antibiotic vancomycin (8 mg/kg/day), either with or without PFOS. PFOS exposure decreased thyroxine (T4) and triiodothyronine (T3) without affecting thyroid stimulating hormone (TSH), resembling a state of hypothyroxinemia. PFOS exposure resulted in 50 differentially expressed genes (DEGs) in the hypothalamus, 68 DEGs in the pituitary, 71 DEGs in the thyroid, and 181 DEGs in the liver. A concomitant compromised gut microbiota did not significantly change effects of PFOS exposure. Organ-specific DEGs did not align with TH regulating genes; however, genes associated with vesicle transport and neuronal signaling were affected in the hypothalamus, and phase I and phase II metabolism in the liver. This suggests that a decrease in systemic TH levels may activate the expression of factors altering trafficking, metabolism and excretion of TH. At the transcriptional level, little evidence suggests that the pituitary or thyroid gland is involved in PFOS-induced TH system disruption.
Collapse
Affiliation(s)
- Nichlas Davidsen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Louise Ramhøj
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Claus Asger Lykkebo
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Indusha Kugathas
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Rikke Poulsen
- Department of Environmental Science, Aarhus University, Roskilde, DK-4000, Denmark
| | | | - Bertrand Evrard
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | | | - Marta Axelstad
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Martin Hansen
- Department of Environmental Science, Aarhus University, Roskilde, DK-4000, Denmark
| | - Frederic Chalmel
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark.
| |
Collapse
|
16
|
Chen Q, Chou WC, Lin Z. Integration of Toxicogenomics and Physiologically Based Pharmacokinetic Modeling in Human Health Risk Assessment of Perfluorooctane Sulfonate. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:3623-3633. [PMID: 35194992 DOI: 10.1021/acs.est.1c06479] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Toxicogenomics and physiologically based pharmacokinetic (PBPK) models are useful approaches in chemical risk assessment, but the methodology to incorporate toxicogenomic data into a PBPK model to inform risk assessment remains to be developed. This study aimed to develop a probabilistic human health risk assessment approach by integrating toxicogenomic dose-response data and PBPK modeling using perfluorooctane sulfonate (PFOS) as a case study. Based on the available human in vitro and mouse in vivo toxicogenomic data, we identified the differentially expressed genes (DEGs) at each exposure paradigm/duration. Kyoto Encyclopedia of Genes and Genomes and disease ontology enrichment analyses were conducted on the DEGs to identify significantly enriched pathways and diseases. The dose-response data of DEGs were analyzed using the Bayesian benchmark dose (BMD) method. Using a previously published PBPK model, the gene BMDs were converted to human equivalent doses (HEDs), which were summarized to pathway and disease HEDs and then extrapolated to reference doses (RfDs) by considering an uncertainty factor of 30 for mouse in vivo data and 10 for human in vitro data. The results suggested that the median RfDs at different exposure paradigms were similar to the 2016 U.S. Environmental Protection Agency's recommended RfD, while the RfDs for the most sensitive pathways and diseases were closer to the recent European Food Safety Authority's guidance values. In conclusion, genomic dose-response data and PBPK modeling can be integrated to become a useful alternative approach in risk assessment of environmental chemicals. This approach considers multiple endpoints, provides toxicity mechanistic insights, and does not rely on apical toxicity endpoints.
Collapse
Affiliation(s)
- Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida 32610, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32608, United States
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida 32610, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32608, United States
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida 32610, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32608, United States
| |
Collapse
|
17
|
Su S, Billy LJ, Chang S, Gonzalez FJ, Patterson AD, Peters JM. The role of mouse and human peroxisome proliferator-activated receptor-α in modulating the hepatic effects of perfluorooctane sulfonate in mice. Toxicology 2022; 465:153056. [PMID: 34861291 PMCID: PMC10292111 DOI: 10.1016/j.tox.2021.153056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022]
Abstract
Perfluorooctane sulfonate (PFOS) is a stable environmental contaminant that can activate peroxisome proliferator-activated receptor alpha (PPARα). In the present work, the specific role of mouse and human PPARα in mediating the hepatic effects of PFOS was examined in short-term studies using wild type, Ppara-null and PPARA-humanized mice. Mice fed 0.006 % PFOS for seven days (∼10 mg/kg/day), or 0.003 % PFOS for twenty-eight days (∼5 mg/kg/day), exhibited higher liver and serum PFOS concentrations compared to controls. Relative liver weights were also higher following exposure to dietary PFOS in all three genotypes as compared vehicle fed control groups. Histopathological examination of liver sections from mice treated for twenty-eight days with 0.003 % PFOS revealed a phenotype consistent with peroxisome proliferation, in wild-type and PPARA-humanized mice that was not observed in Ppara-null mice. With both exposures, expression of the PPARα target genes, Acox1, Cyp4a10, was significantly increased in wild type mice but not in Ppara-null or PPARA-humanized mice. By contrast, expression of the constitutive androstane receptor (CAR) target gene, Cyp2b10, and the pregnane X receptor (PXR) target gene, Cyp3a11, were higher in response to PFOS administration in all three genotypes compared to controls for both exposure periods. These results indicate that mouse PPARα can be activated in the liver by PFOS causing increased expression of Acox1, Cyp4a10 and histopathological changes in the liver. While histopathological analyses indicated the presence of mouse PPARα-dependent hepatic peroxisome proliferation in wild-type (a response associated with activation of PPARα) and a similar phenotype in PPARA-humanized mice, the lack of increased Acox1 and Cyp4a10 mRNA by PFOS in PPARA-humanized mice indicates that the human PPARα was not as responsive to PFOS as mouse PPARα with this dose regimen. Moreover, results indicate that hepatomegaly caused by PFOS does not require mouse or human PPARα and could be due to effects induced by activation of CAR and/or PXR.
Collapse
Affiliation(s)
- Shengzhong Su
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States.
| | - Laura J Billy
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| | - Sue Chang
- Corporate Occupational Medicine, 3M Company, St. Paul, MN, United States
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD, United States
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
18
|
Pan Y, Qin H, Zheng L, Guo Y, Liu W. Disturbance in transcriptomic profile, proliferation and multipotency in human mesenchymal stem cells caused by hexafluoropropylene oxides. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 292:118483. [PMID: 34763017 DOI: 10.1016/j.envpol.2021.118483] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/24/2021] [Accepted: 11/07/2021] [Indexed: 06/13/2023]
Abstract
As alternatives to perfluorooctanoic acid (PFOA), hexafluoropropylene oxide dimer acid (HFPO-DA) and hexafluoropropylene oxide trimer acid (HFPO-TA) have raised concerns of their potential health risks. Human bone marrow mesenchymal stem cell was employed as an in vitro model to investigate the molecular targets and the adverse effects of HFPOs in stem cells in concentrations range starting at human relevant levels. Unsupervised transcriptomic analysis identified 1794 and 1429 DEGs affected by HFPO-TA and HFPO-DA, respectively. Cell cycle-associated biological processes were commonly altered by both chemicals. 18 and 35 KEGG pathways were enriched in HFPO-TA and HFPO-DA treatment group, respectively, among which multiple pathways were related to cancer and pluripotency. Few genes in PPAR signalling pathway were disturbed by HFPOs suggesting the involvement of PPAR-independent toxic mechanism. HFPO-TA promoted cell proliferation with significance at 1 μM mRNA levels of CDK and MYC were down-regulated by HFPOs, suggesting the negative feedback regulation to the abnormal cell proliferation. Decreased expression of CD44 protein, and ENG and THY1 mRNA levels demonstrated HFPOs-caused changes of hBMSCs phenotype. The osteogenic differentiation was also inhibited by HFPOs with reduced formation of calcium deposition. Furthermore, gene and protein expression of core pluripotency regulators NANOG was enhanced by HFPO-TA. The present study provides human relevant mechanistic evidence for health risk assessment of HFPOs, prioritizing comprehensive carcinogenicity assessment of this type of PFOA alternatives.
Collapse
Affiliation(s)
- Yifan Pan
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China
| | - Hui Qin
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China
| | - Lu Zheng
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Lu, Shanghai, 200032, China
| | - Wei Liu
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China.
| |
Collapse
|
19
|
Azimzadeh O, Subramanian V, Sievert W, Merl-Pham J, Oleksenko K, Rosemann M, Multhoff G, Atkinson MJ, Tapio S. Activation of PPARα by Fenofibrate Attenuates the Effect of Local Heart High Dose Irradiation on the Mouse Cardiac Proteome. Biomedicines 2021; 9:biomedicines9121845. [PMID: 34944662 PMCID: PMC8698387 DOI: 10.3390/biomedicines9121845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 01/13/2023] Open
Abstract
Radiation-induced cardiovascular disease is associated with metabolic remodeling in the heart, mainly due to the inactivation of the transcription factor peroxisome proliferator-activated receptor alpha (PPARα), thereby inhibiting lipid metabolic enzymes. The objective of the present study was to investigate the potential protective effect of fenofibrate, a known agonist of PPARα on radiation-induced cardiac toxicity. To this end, we compared, for the first time, the cardiac proteome of fenofibrate- and placebo-treated mice 20 weeks after local heart irradiation (16 Gy) using label-free proteomics. The observations were further validated using immunoblotting, enzyme activity assays, and ELISA. The analysis showed that fenofibrate restored signalling pathways that were negatively affected by irradiation, including lipid metabolism, mitochondrial respiratory chain, redox response, tissue homeostasis, endothelial NO signalling and the inflammatory status. The results presented here indicate that PPARα activation by fenofibrate attenuates the cardiac proteome alterations induced by irradiation. These findings suggest a potential benefit of fenofibrate administration in the prevention of cardiovascular diseases, following radiation exposure.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection, 85764 Neuherberg, Germany
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Correspondence: ; Tel.: +49-030/18333-2242
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA
| | - Wolfgang Sievert
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, 80939 Munich, Germany;
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Gabriele Multhoff
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Michael J. Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Chair of Radiation Biology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| |
Collapse
|
20
|
Bjork JA, Dawson DA, Krogstad JO, Wallace KB. Transcriptional effects of binary combinations of PFAS in FaO cells. Toxicology 2021; 464:152997. [PMID: 34695511 DOI: 10.1016/j.tox.2021.152997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 11/25/2022]
Abstract
Per- and polyfluoroalkyl substances (PFAS) represent a large class of structurally diverse chemicals of increasing public concern, mostly due to their chemical stability and undetermined toxicity profiles. In laboratory animals, adverse effects implicated for certain PFAS, perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) in particular, include liver toxicity and the associated metabolic dysregulation, immune and thyroid alterations, reproductive toxicity, and selected tumors. The broad commercialization and environmental distribution of PFAS has drawn attention to the need for understanding risks associated with combined exposure to multiple PFAS in complex mixtures. The purpose of this investigation is to determine whether binary combinations of PFAS elicit a molecular response that is either greater than or less than the sum of the individual responses. Exposure of FaO rat hepatoma cells for 24 h to 25 μM-200 μM of the 4- and 8-carbon perfluorocarboxylic acids (PFBA and PFOA) or the 4, 6, and 8-carbon perfluorosulfonic acids (PFBS, PFHxS, and PFOS, respectively) individually caused a dose-dependent increase in PPARα-regulated expression of peroxisomal bifunctional enzyme (Ehhadh). Potency increased with carbon number, with the carboxylates eliciting a greater transcriptional response than the corresponding sulfonates. Combined exposure to PFOA and PFBA produced an effect that was significantly less than the sum of the individual responses. The response to the combination of PFOA and PFOS produced a summative effect at concentrations that were not cytotoxic. Combined exposures to PFOS and either PFBS or PFHxS at low noncytotoxic concentrations produced a transcriptional effect that was significantly less than the sum of the individual effects. The results demonstrate that among the five structurally related perfluoroalkyl acids included in this investigation, PPARα transcriptional activation in response to combined binary exposures is consistently at or below that predicted by the sum of the individual effects.
Collapse
Affiliation(s)
- James A Bjork
- University of Minnesota Medical School, Department of Biomedical Sciences, 1035 University Drive, Duluth, MN, 55812, United States
| | - Douglas A Dawson
- Department of Biology/Toxicology, 318 Kettering Science Center, Ashland University, Ashland, OH, United States
| | - Jacob O Krogstad
- University of Minnesota Medical School, Department of Biomedical Sciences, 1035 University Drive, Duluth, MN, 55812, United States
| | - Kendall B Wallace
- University of Minnesota Medical School, Department of Biomedical Sciences, 1035 University Drive, Duluth, MN, 55812, United States.
| |
Collapse
|
21
|
Goodrich JA, Alderete TL, Baumert BO, Berhane K, Chen Z, Gilliland FD, Goran MI, Hu X, Jones DP, Margetaki K, Rock S, Stratakis N, Valvi D, Walker DI, Conti DV, Chatzi L. Exposure to Perfluoroalkyl Substances and Glucose Homeostasis in Youth. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:97002. [PMID: 34468161 PMCID: PMC8409228 DOI: 10.1289/ehp9200] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
BACKGROUND Exposure to per- and polyfluoroalkyl substances (PFAS), a prevalent class of persistent pollutants, may increase the risk of type 2 diabetes. OBJECTIVE We examined associations between PFAS exposure and glucose metabolism in youth. METHODS Overweight/obese adolescents from the Study of Latino Adolescents at Risk of Type 2 Diabetes (SOLAR; n=310) participated in annual visits for an average of 3.3±2.9y. Generalizability of findings were tested in young adults from the Southern California Children's Health Study (CHS; n=135) who participated in a clinical visit with a similar protocol. At each visit, oral glucose tolerance tests were performed to estimate glucose metabolism and β-cell function via the insulinogenic index. Four PFAS were measured at baseline using liquid chromatography-high-resolution mass spectrometry; high levels were defined as concentrations >66th percentile. RESULTS In females from the SOLAR, high perfluorohexane sulfonate (PFHxS) levels (≥2.0 ng/mL) were associated with the development of dysregulated glucose metabolism beginning in late puberty. The magnitude of these associations increased postpuberty and persisted through 18 years of age. For example, postpuberty, females with high PFHxS levels had 25-mg/dL higher 60-min glucose (95% CI: 12, 39mg/dL; p<0.0001), 15-mg/dL higher 2-h glucose (95% CI: 1, 28mg/dL; p=0.04), and 25% lower β-cell function (p=0.02) compared with females with low levels. Results were largely consistent in the CHS, where females with elevated PFHxS levels had 26-mg/dL higher 60-min glucose (95% CI: 6.0, 46mg/dL; p=0.01) and 19-mg/dL higher 2-h glucose, which did not meet statistical significance (95% CI: -1, 39mg/dL; p=0.08). In males, no consistent associations between PFHxS and glucose metabolism were observed. No consistent associations were observed for other PFAS and glucose metabolism. DISCUSSION Youth exposure to PFHxS was associated with dysregulated glucose metabolism in females, which may be due to changes in β-cell function. These associations appeared during puberty and were most pronounced postpuberty. https://doi.org/10.1289/EHP9200.
Collapse
Affiliation(s)
- Jesse A. Goodrich
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Brittney O. Baumert
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Kiros Berhane
- Department of Biostatistics, Columbia University, New York, New York, USA
| | - Zhanghua Chen
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Frank D. Gilliland
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Michael I. Goran
- Department of Pediatrics, Keck School of Medicine, Los Angeles, California, USA
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Xin Hu
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dean P. Jones
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Katerina Margetaki
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Sarah Rock
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Nikos Stratakis
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Douglas I. Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David V. Conti
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Leda Chatzi
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
22
|
Rowan-Carroll A, Reardon A, Leingartner K, Gagné R, Williams A, Meier MJ, Kuo B, Bourdon-Lacombe J, Moffat I, Carrier R, Nong A, Lorusso L, Ferguson SS, Atlas E, Yauk C. High-Throughput Transcriptomic Analysis of Human Primary Hepatocyte Spheroids Exposed to Per- and Polyfluoroalkyl Substances as a Platform for Relative Potency Characterization. Toxicol Sci 2021; 181:199-214. [PMID: 33772556 DOI: 10.1093/toxsci/kfab039] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Per- and poly-fluoroalkyl substances (PFAS) are widely found in the environment because of their extensive use and persistence. Although several PFAS are well studied, most lack toxicity data to inform human health hazard and risk assessment. This study focused on 4 model PFAS: perfluorooctanoic acid (PFOA; 8 carbon), perfluorobutane sulfonate (PFBS; 4 carbon), perfluorooctane sulfonate (PFOS; 8 carbon), and perfluorodecane sulfonate (PFDS; 10 carbon). Human primary liver cell spheroids (pooled from 10 donors) were exposed to 10 concentrations of each PFAS and analyzed at 4 time points. The approach aimed to: (1) identify gene expression changes mediated by the PFAS, (2) identify similarities in biological responses, (3) compare PFAS potency through benchmark concentration analysis, and (4) derive bioactivity exposure ratios (ratio of the concentration at which biological responses occur, relative to daily human exposure). All PFAS induced transcriptional changes in cholesterol biosynthesis and lipid metabolism pathways, and predicted PPARα activation. PFOS exhibited the most transcriptional activity and had a highly similar gene expression profile to PFDS. PFBS induced the least transcriptional changes and the highest benchmark concentration (ie, was the least potent). The data indicate that these PFAS may have common molecular targets and toxicities, but that PFOS and PFDS are the most similar. The transcriptomic bioactivity exposure ratios derived here for PFOA and PFOS were comparable to those derived using rodent apical endpoints in risk assessments. These data provide a baseline level of toxicity for comparison with other known PFAS using this testing strategy.
Collapse
Affiliation(s)
- Andrea Rowan-Carroll
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Anthony Reardon
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Karen Leingartner
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Remi Gagné
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Matthew J Meier
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Byron Kuo
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Julie Bourdon-Lacombe
- Water and Air Quality Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Ivy Moffat
- Water and Air Quality Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Richard Carrier
- Water and Air Quality Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Andy Nong
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Luigi Lorusso
- Chemicals and Environmental Health Management Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Stephen S Ferguson
- U.S. National Institute of Environmental Health Sciences (NIEHS), Ottawa, Ontario K1N 6N5, Canada
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada
| | - Carole Yauk
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, Ontario K1N 6N5, Canada.,Department of Biology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
23
|
Salter DM, Wei W, Nahar PP, Marques E, Slitt AL. Perfluorooctanesulfonic Acid (PFOS) Thwarts the Beneficial Effects of Calorie Restriction and Metformin. Toxicol Sci 2021; 182:82-95. [PMID: 33844015 DOI: 10.1093/toxsci/kfab043] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A combination of calorie restriction (CR), dietary modification, and exercise is the recommended therapy to reverse obesity and nonalcoholic fatty liver disease. In the liver, CR shifts hepatic metabolism from lipid storage to lipid utilization pathways, such as AMP-activated protein kinase (AMPK). Perfluorooctanesulfonic acid (PFOS), a fluorosurfactant previously used in stain repellents and anti-stick materials, can increase hepatic lipids in mice following relatively low-dose exposures. To test the hypothesis that PFOS administration interferes with CR, adult male C57BL/6N mice were fed ad libitum or a 25% reduced calorie diet concomitant with either vehicle (water) or 100 μg PFOS/kg/day via oral gavage for 6 weeks. CR alone improved hepatic lipids and glucose tolerance. PFOS did not significantly alter CR-induced weight loss, white adipose tissue mass, or liver weight over 6 weeks. However, PFOS increased hepatic triglyceride accumulation, in both mice fed ad libitum and subjected to CR. This was associated with decreased phosphorylated AMPK expression in liver. Glucagon (100 nM) treatment induced glucose production in hepatocytes, which was further upregulated with PFOS (2.5 μM) co-treatment. Next, to explore whether the observed changes were related to AMPK signaling, HepG2 cells were treated with metformin or AICAR alone or in combination with PFOS (25 μM). PFOS interfered with glucose-lowering effects of metformin, and AICAR treatment partially impaired PFOS-induced increase in glucose production. In 3T3-L1 adipocytes, metformin was less effective with PFOS co-treatment. Overall, PFOS administration disrupted hepatic lipid and glucose homeostasis and interfered with beneficial glucose-lowering effects of CR and metformin.
Collapse
Affiliation(s)
- Deanna M Salter
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Wei Wei
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Pragati P Nahar
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Emily Marques
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| |
Collapse
|
24
|
Pfohl M, Ingram L, Marques E, Auclair A, Barlock B, Jamwal R, Anderson D, Cummings BS, Slitt AL. Perfluorooctanesulfonic Acid and Perfluorohexanesulfonic Acid Alter the Blood Lipidome and the Hepatic Proteome in a Murine Model of Diet-Induced Obesity. Toxicol Sci 2021; 178:311-324. [PMID: 32991729 DOI: 10.1093/toxsci/kfaa148] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Perfluoroalkyl substances (PFAS) represent a family of environmental toxicants that have infiltrated the living world. This study explores diet-PFAS interactions and the impact of perfluorooctanesulfonic acid (PFOS) and perfluorohexanesulfonic (PFHxS) on the hepatic proteome and blood lipidomic profiles. Male C57BL/6J mice were fed with either a low-fat diet (10.5% kcal from fat) or a high fat (58% kcal from fat) high carbohydrate (42 g/l) diet with or without PFOS or PFHxS in feed (0.0003% wt/wt) for 29 weeks. Lipidomic, proteomic, and gene expression profiles were determined to explore lipid outcomes and hepatic mechanistic pathways. With administration of a high-fat high-carbohydrate diet, PFOS and PFHxS increased hepatic expression of targets involved in lipid metabolism and oxidative stress. In the blood, PFOS and PFHxS altered serum phosphatidylcholines, phosphatidylethanolamines, plasmogens, sphingomyelins, and triglycerides. Furthermore, oxidized lipid species were enriched in the blood lipidome of PFOS and PFHxS treated mice. These data support the hypothesis that PFOS and PFHxS increase the risk of metabolic and inflammatory disease induced by diet, possibly by inducing dysregulated lipid metabolism and oxidative stress.
Collapse
Affiliation(s)
- Marisa Pfohl
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Lishann Ingram
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602.,Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland 21218
| | - Emily Marques
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Adam Auclair
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Benjamin Barlock
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Rohitash Jamwal
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Dwight Anderson
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Brian S Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602.,Interdisciplinary Toxicology Program, College of Pharmacy, University of Georgia, Athens, Georgia 30602
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881
| |
Collapse
|
25
|
Andersen ME, Hagenbuch B, Apte U, Corton JC, Fletcher T, Lau C, Roth WL, Staels B, Vega GL, Clewell HJ, Longnecker MP. Why is elevation of serum cholesterol associated with exposure to perfluoroalkyl substances (PFAS) in humans? A workshop report on potential mechanisms. Toxicology 2021; 459:152845. [PMID: 34246716 PMCID: PMC9048712 DOI: 10.1016/j.tox.2021.152845] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/25/2021] [Accepted: 06/22/2021] [Indexed: 01/09/2023]
Abstract
Serum concentrations of cholesterol are positively correlated with exposure to perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) in humans. The associated change in cholesterol is small across a broad range of exposure to PFOA and PFOS. Animal studies generally have not indicated a mechanism that would account for the association in humans. The extent to which the relationship is causal is an open question. Nonetheless, the association is of particular importance because increased serum cholesterol has been considered as an endpoint to derive a point of departure in at least one recent risk assessment. To gain insight into potential mechanisms for the association, both causal and non-causal, an expert workshop was held Oct 31 and Nov 1, 2019 to discuss relevant data and propose new studies. In this report, we summarize the relevant background data, the discussion among the attendees, and their recommendations for further research.
Collapse
Affiliation(s)
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd - MS 1018, Kansas City, KS 66160, USA.
| | - Udayan Apte
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd - MS 1018, Kansas City, KS 66160, USA.
| | - J Christopher Corton
- Advanced Experimental Toxicology Models Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 T.W. Alexander Dr., MD B105-03, Research Triangle Park, NC 27711, USA.
| | - Tony Fletcher
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | - Christopher Lau
- Reproductive and Developmental Toxicology Branch, Public Health and Integrated Toxicology Division, Mail Code B105-04, Center for Public Health and Environmental Assessment, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - William L Roth
- U.S. Food and Drug Administration (Retired), Numerical Animals, 16005 Frontier Rd., Reno, NV 89508, USA.
| | - Bart Staels
- Univ. Lille, Inserm, CHU de Lille, Institut Pasteur de Lille, U1011-EGID, F-59019 Lille, France.
| | - Gloria L Vega
- Center for Human Nutrition, Dallas, TX, USA; Department of Clinical Nutrition, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9052, USA.
| | - Harvey J Clewell
- Ramboll US Consulting, Inc., 3214 Charles B. Root Wynd, Suite 130, Raleigh, NC 27612, USA.
| | - Matthew P Longnecker
- Ramboll US Consulting, Inc., 3214 Charles B. Root Wynd, Suite 130, Raleigh, NC 27612, USA.
| |
Collapse
|
26
|
Hamilton MC, Heintz MM, Pfohl M, Marques E, Ford L, Slitt AL, Baldwin WS. Increased toxicity and retention of perflourooctane sulfonate (PFOS) in humanized CYP2B6-Transgenic mice compared to Cyp2b-null mice is relieved by a high-fat diet (HFD). Food Chem Toxicol 2021; 152:112175. [PMID: 33838175 PMCID: PMC8154739 DOI: 10.1016/j.fct.2021.112175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 01/11/2023]
Abstract
PFOS is a persistent, fluorosurfactant used in multiple products. Murine Cyp2b's are induced by PFOS and high-fat diets (HFD) and therefore we hypothesized that human CYP2B6 may alleviate PFOS-induced steatosis. Cyp2b-null and hCYP2B6-Tg mice were treated with 0, 1, or 10 mg/kg/day PFOS by oral gavage for 21-days while provided a chow diet (ND) or HFD. Similar to murine Cyp2b10, CYP2B6 is inducible by PFOS. Furthermore, three ND-fed hCYP2B6-Tg females treated with 10 mg/kg/day PFOS died during the exposure period; neither Cyp2b-null nor HFD-fed mice died. hCYP2B6-Tg mice retained more PFOS in serum and liver than Cyp2b-null mice presumably causing the observed toxicity. In contrast, serum PFOS retention was reduced in the HFD-fed hCYP2B6-Tg mice; the opposite trend observed in HFD-fed Cyp2b-null mice. Hepatotoxicity biomarkers, ALT and ALP, were higher in PFOS-treated mice and repressed by a HFD. However, PFOS combined with a HFD exacerbated steatosis in all mice, especially in the hCYP2B6-Tg mice with significant disruption of key lipid metabolism genes such as Srebp1, Pparg, and Hmgcr. In conclusion, CYP2B6 is induced by PFOS but does not alleviate PFOS toxicity presumably due to increased retention. CYP2B6 protects from PFOS-mediated steatosis in ND-fed mice, but increases steatosis when co-treated with a HFD.
Collapse
Affiliation(s)
- Matthew C Hamilton
- Environmental Toxicology Program, Clemson University, Clemson, SC, 29634, USA
| | - Melissa M Heintz
- Environmental Toxicology Program, Clemson University, Clemson, SC, 29634, USA
| | - Marisa Pfohl
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Emily Marques
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Lucie Ford
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Angela L Slitt
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - William S Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
27
|
Sant KE, Annunziato K, Conlin S, Teicher G, Chen P, Venezia O, Downes GB, Park Y, Timme-Laragy AR. Developmental exposures to perfluorooctanesulfonic acid (PFOS) impact embryonic nutrition, pancreatic morphology, and adiposity in the zebrafish, Danio rerio. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 275:116644. [PMID: 33581636 PMCID: PMC8101273 DOI: 10.1016/j.envpol.2021.116644] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/13/2021] [Accepted: 01/30/2021] [Indexed: 05/17/2023]
Abstract
Perfluorooctanesulfonic acid (PFOS) is a persistent environmental contaminant previously found in consumer surfactants and industrial fire-fighting foams. PFOS has been widely implicated in metabolic dysfunction across the lifespan, including diabetes and obesity. However, the contributions of the embryonic environment to metabolic disease remain uncharacterized. This study seeks to identify perturbations in embryonic metabolism, pancreas development, and adiposity due to developmental and subchronic PFOS exposures and their persistence into later larval and juvenile periods. Zebrafish embryos were exposed to 16 or 32 μM PFOS developmentally (1-5 days post fertilization; dpf) or subchronically (1-15 dpf). Embryonic fatty acid and macronutrient concentrations and expression of peroxisome proliferator-activated receptor (PPAR) isoforms were quantified in embryos. Pancreatic islet morphometry was assessed at 15 and 30 dpf, and adiposity and fish behavior were assessed at 15 dpf. Concentrations of lauric (C12:0) and myristic (C14:0) saturated fatty acids were increased by PFOS at 4 dpf, and PPAR gene expression was reduced. Incidence of aberrant islet morphologies, principal islet areas, and adiposity were increased in 15 dpf larvae and 30 dpf juvenile fish. Together, these data suggest that the embryonic period is a susceptible window of metabolic programming in response to PFOS exposures, and that these early exposures alone can have persisting effects later in the lifecourse.
Collapse
Affiliation(s)
- Karilyn E Sant
- Division of Environmental Health, San Diego State University School of Public Health, San Diego, CA, 92182, USA; Department of Environmental Health Sciences, University of Massachusetts School of Public Health and Health Sciences, Amherst, MA, 01003, USA.
| | - Kate Annunziato
- Department of Environmental Health Sciences, University of Massachusetts School of Public Health and Health Sciences, Amherst, MA, 01003, USA
| | - Sarah Conlin
- Department of Environmental Health Sciences, University of Massachusetts School of Public Health and Health Sciences, Amherst, MA, 01003, USA
| | - Gregory Teicher
- Biology Department, University of Massachusetts, Amherst, MA, 01003, USA
| | - Phoebe Chen
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| | - Olivia Venezia
- Department of Environmental Health Sciences, University of Massachusetts School of Public Health and Health Sciences, Amherst, MA, 01003, USA
| | - Gerald B Downes
- Biology Department, University of Massachusetts, Amherst, MA, 01003, USA
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, University of Massachusetts School of Public Health and Health Sciences, Amherst, MA, 01003, USA
| |
Collapse
|
28
|
Fragki S, Dirven H, Fletcher T, Grasl-Kraupp B, Bjerve Gützkow K, Hoogenboom R, Kersten S, Lindeman B, Louisse J, Peijnenburg A, Piersma AH, Princen HMG, Uhl M, Westerhout J, Zeilmaker MJ, Luijten M. Systemic PFOS and PFOA exposure and disturbed lipid homeostasis in humans: what do we know and what not? Crit Rev Toxicol 2021; 51:141-164. [PMID: 33853480 DOI: 10.1080/10408444.2021.1888073] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Associations between per- and polyfluoroalkyl substances (PFASs) and increased blood lipids have been repeatedly observed in humans, but a causal relation has been debated. Rodent studies show reverse effects, i.e. decreased blood cholesterol and triglycerides, occurring however at PFAS serum levels at least 100-fold higher than those in humans. This paper aims to present the main issues regarding the modulation of lipid homeostasis by the two most common PFASs, PFOS and PFOA, with emphasis on the underlying mechanisms relevant for humans. Overall, the apparent contrast between human and animal data may be an artifact of dose, with different molecular pathways coming into play upon exposure to PFASs at very low versus high levels. Altogether, the interpretation of existing rodent data on PFOS/PFOA-induced lipid perturbations with respect to the human situation is complex. From a mechanistic perspective, research on human liver cells shows that PFOS/PFOA activate the PPARα pathway, whereas studies on the involvement of other nuclear receptors, like PXR, are less conclusive. Other data indicate that suppression of the nuclear receptor HNF4α signaling pathway, as well as perturbations of bile acid metabolism and transport might be important cellular events that require further investigation. Future studies with human-relevant test systems would help to obtain more insight into the mechanistic pathways pertinent for humans. These studies shall be designed with a careful consideration of appropriate dosing and toxicokinetics, so as to enable biologically plausible quantitative extrapolations. Such research will increase the understanding of possible perturbed lipid homeostasis related to PFOS/ PFOA exposure and the potential implications for human health.
Collapse
Affiliation(s)
- Styliani Fragki
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Hubert Dirven
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Tony Fletcher
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England (PHE), Chilton, UK
| | - Bettina Grasl-Kraupp
- Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, Vienna, Austria
| | | | - Ron Hoogenboom
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Birgitte Lindeman
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jochem Louisse
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Ad Peijnenburg
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.,Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Hans M G Princen
- Metabolic Health Research, The Netherlands Organization of Applied Scientific Research (TNO), Gaubius Laboratory, Leiden, The Netherlands
| | - Maria Uhl
- Environment Agency Austria (EAA), Vienna, Austria
| | - Joost Westerhout
- Risk Analysis for Products In Development, The Netherlands Organization of Applied Scientific Research (TNO), Utrecht, The Netherlands
| | - Marco J Zeilmaker
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
29
|
Li D, Jiang L, Hong Y, Cai Z. Multilayered glycoproteomic analysis reveals the hepatotoxic mechanism in perfluorooctane sulfonate (PFOS) exposure mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115774. [PMID: 33143982 DOI: 10.1016/j.envpol.2020.115774] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is one of the most widely used and distributed perfluorinated compounds proven to cause adverse health outcomes. Datasets of ecotoxico-genomics and proteomics have given greater insights for PFOS toxicological effect. However, the molecular mechanisms of hepatotoxicity of PFOS on post-translational modifications (PTMs) regulation, which is most relevant for regulating the activity of proteins, are not well elucidated. Protein glycosylation is one of the most ubiquitous PTMs associated with diverse cellular functions, which are critical towards the understanding of the multiple biological processes and toxic mechanisms exposed to PFOS. Here, we exploit the multilayered glycoproteomics to quantify the global protein expression levels, glycosylation sites, and glycoproteins in PFOS exposure and wild-type mouse livers. The identified 2439 proteins, 1292 glycosites, and 799 glycoproteins were displayed complex heterogeneity in PFOS exposure mouse livers. Quantification results reveal that 241 dysregulated proteins (fold change ≥ 2, p < 0.05) in PFOS exposure mouse livers were involved in the lipid and xenobiotic metabolism. While, 16 overexpressed glycoproteins were exclusively related to neutrophil degranulation, cellular responses to stress, protein processing in endoplasmic reticulum (ER). Moreover, the interactome and functional network analysis identified HP and HSP90AA1 as the potential glycoprotein biomarkers. These results provide unique insights into a deep understanding of the mechanisms of PFOS induced hepatotoxicity and liver disease. Our platform of multilayered glycoproteomics can be adapted to diverse ecotoxicological research.
Collapse
Affiliation(s)
- Dapeng Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China
| | - Lilong Jiang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Yanjun Hong
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
30
|
Roth K, Imran Z, Liu W, Petriello MC. Diet as an Exposure Source and Mediator of Per- and Polyfluoroalkyl Substance (PFAS) Toxicity. FRONTIERS IN TOXICOLOGY 2020; 2:601149. [PMID: 35296120 PMCID: PMC8915917 DOI: 10.3389/ftox.2020.601149] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/06/2020] [Indexed: 01/09/2023] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) are ubiquitously found in the environment due to their widespread commercial use and high chemical stability. Humans are exposed primarily through ingestion of contaminated water and food and epidemiological studies over the last several decades have shown that PFAS levels are associated with adverse chronic health effects, including cardiometabolic disorders such as hyperlipidemia and non-alcoholic fatty liver disease. Perhaps the most well-established effects, as demonstrated in animal studies and human epidemiological studies, are the metabolic alterations PFAS exposure can lead to, especially on lipid homeostasis and signaling. This altered lipid metabolism has often been linked to conditions such as dyslipidemia, leading to fatty liver disease and steatosis. Western diets enriched in high fat and high cholesterol containing foods may be an important human exposure route of PFAS and may also act as an important modulator of associated toxicities. In fact, the chemical structure of PFAS resemble fatty acids and may activate some of the same signaling cascades critical for endogenous metabolism. In this review we aim to outline known dietary exposure sources of PFAS, describe the detrimental metabolic health effects associated with PFAS exposure, and focus on studies examining emerging interaction of dietary effects with PFAS exposure that further alter the dysregulated metabolic state.
Collapse
Affiliation(s)
- Katherine Roth
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States
| | - Zunaira Imran
- Department of Chemistry, Wayne State University, Detroit, MI, United States
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, Wayne State University, Detroit, MI, United States
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Michael C. Petriello
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, United States
- *Correspondence: Michael C. Petriello
| |
Collapse
|
31
|
Liu J, Liu S, Huang Z, Fu Y, Fei J, Liu X, He Z. Associations between the serum levels of PFOS/PFOA and IgG N-glycosylation in adult or children. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114285. [PMID: 32806420 DOI: 10.1016/j.envpol.2020.114285] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Perfluorooctanoic acid (PFOA) and perfluorooctanoic sulfonate (PFOS) have been shown to be associated with disease development. Immunoglobulin G (IgG) N-glycosylation plays a vital role in human immune system and inflammatory activities. Altered IgG glycosylation was one of the molecular markers of various disorders. However, whether the chemicals affect IgG glycosylation has not been investigated. METHODS Serum samples of 190 individuals including 95 adults and 95 children were selected based on the sex, age and PFOA/PFOS concentration. IgG N-glycome profile was obtained from glycan release, derivatization, and MALDI-MS analysis. One-factor ANOVA test was performed to analyze the association between different levels of PFOS/PFOA and IgG glycosylation changes. Evaluation of the diagnostic performance of significantly changed IgG glycosylation was performed by receiver operating characteristic curve. PFOS/PFOA concentrations were studied in relation to IgG glycosylation by 3D-nonlinear regression analysis. RESULTS 10 of the 28 individual IgG glycans were significantly altered between different levels of PFOS/PFOA in adult serum. Among children with high serum levels of PFOS or PFOA, a total of 12 IgG N-glycans were markedly different from those with lower serum PFOS/PFOA. The glycan derived traits for adults with higher serum PFOS or PFOA were marked by significant alterations in IgG digalactosylation, agalactosylation, fucosylation, fucosylated sialylation, and disialylation. Similarly, pronounced changes in agalactosylation, digalactosylation, mono-sialylation and total sialylation, as well as neutral and sialo bisection, were associated with elevated serum PFOS or PFOA in children. Several glycans gained moderately accurate scores of area under the curve for diagnosis of PFOS or PFOA pollution. Nonlinear surface fitting showed the independent or coordinate effect of PFOS or PFOA on the expression of IgG glycosylation. CONCLUSIONS High levels of PFOS or PFOA in human serum were strongly associated with altered IgG glycosylation and therefore are a potential risk factor for the development of diseases.
Collapse
Affiliation(s)
- Junling Liu
- Wuhan Centers for Disease Prevention and Control, Wuhan, 430015, China
| | - Si Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhiwen Huang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yang Fu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jian Fei
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Zhenyu He
- Wuhan Centers for Disease Prevention and Control, Wuhan, 430015, China.
| |
Collapse
|
32
|
Marques E, Pfohl M, Auclair A, Jamwal R, Barlock BJ, Sammoura FM, Goedken M, Akhlaghi F, Slitt AL. Perfluorooctanesulfonic acid (PFOS) administration shifts the hepatic proteome and augments dietary outcomes related to hepatic steatosis in mice. Toxicol Appl Pharmacol 2020; 408:115250. [PMID: 32979393 DOI: 10.1016/j.taap.2020.115250] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/01/2020] [Accepted: 09/21/2020] [Indexed: 01/09/2023]
Abstract
Hepatic steatosis increases risk of fatty liver and cardiovascular disease. Perfluorooctanesulfonic acid (PFOS) is a persistent, bio-accumulative pollutant that has been used in industrial and commercial applications. PFOS administration induces hepatic steatosis in rodents and increases lipogenic gene expression signatures in cultured hepatocytes. We hypothesized that PFOS treatment interferes with lipid loss when switching from a high fat diet (HFD) to a standard diet (SD), and augments HFD-induced hepatic steatosis. Male C57BL/6 N mice were fed standard chow diet or 60% kCal high-fat diet (HFD) for 4 weeks to increase body weight. Then, some HFD mice were switched to SD and mice were further divided to diet only or diet containing 0.0003% PFOS, for six treatment groups: SD, HFD to SD (H-SD), HFD, SD + PFOS, H-SD + PFOS, or HFD + PFOS. After 10 weeks on study, blood and livers were collected. HFD for 14 weeks increased body weight and hepatic steatosis, whereas H-SD mice returned to SD measures. PFOS administration reduced body weight in mice fed a SD, but not H-SD or HFD. PFOS administration increased liver weight in H-SD + PFOS and HFD + PFOS mice. PFOS increased hepatic steatosis in H-SD and HFD groups. Hepatic mRNA expression and SWATH-MS proteomic analysis revealed that PFOS induced lipid and xenobiotic transporters, as well as metabolism pathways. Overall, the findings herein suggest that PFOS treatment did interfere with lipid loss associated with switch to a SD and similarly augmented hepatic lipid accumulation in mice established on an HFD.
Collapse
Affiliation(s)
- Emily Marques
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Marisa Pfohl
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Adam Auclair
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Rohitash Jamwal
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Benjamin J Barlock
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Ferass M Sammoura
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Michael Goedken
- Rutgers Translational Sciences, Rutgers University, 33 Knightsbridge Road, Piscataway, NJ 08854, USA
| | - Fatemeh Akhlaghi
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Rd, Kingston, RI 02881, USA.
| |
Collapse
|
33
|
Chappell GA, Thompson CM, Wolf JC, Cullen JM, Klaunig JE, Haws LC. Assessment of the Mode of Action Underlying the Effects of GenX in Mouse Liver and Implications for Assessing Human Health Risks. Toxicol Pathol 2020; 48:494-508. [PMID: 32138627 PMCID: PMC7153225 DOI: 10.1177/0192623320905803] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
GenX is an alternative to environmentally persistent long-chain perfluoroalkyl and polyfluoroalkyl substances. Mice exposed to GenX exhibit liver hypertrophy, elevated peroxisomal enzyme activity, and other apical endpoints consistent with peroxisome proliferators. To investigate the potential role of peroxisome proliferator-activated receptor alpha (PPARα) activation in mice, and other molecular signals potentially related to observed liver changes, RNA sequencing was conducted on paraffin-embedded liver sections from a 90-day subchronic toxicity study of GenX conducted in mice. Differentially expressed genes were identified for each treatment group, and gene set enrichment analysis was conducted using gene sets that represent biological processes and known canonical pathways. Peroxisome signaling and fatty acid metabolism were among the most significantly enriched gene sets in both sexes at 0.5 and 5 mg/kg GenX; no pathways were enriched at 0.1 mg/kg. Gene sets specific to the PPARα subtype were significantly enriched. These findings were phenotypically anchored to histopathological changes in the same tissue blocks: hypertrophy, mitoses, and apoptosis. In vitro PPARα transactivation assays indicated that GenX activates mouse PPARα. These results indicate that the liver changes observed in GenX-treated mice occur via a mode of action (MOA) involving PPARα, an important finding for human health risk assessment as this MOA has limited relevance to humans.
Collapse
Affiliation(s)
| | | | | | - John M. Cullen
- North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - James E. Klaunig
- Indiana University, School of Public Health, Bloomington, IN, USA
| | | |
Collapse
|
34
|
Wan HT, Lai KP, Wong CKC. Comparative Analysis of PFOS and PFOA Toxicity on Sertoli Cells. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:3465-3475. [PMID: 32119782 DOI: 10.1021/acs.est.0c00201] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Perfluoroalkyl chemicals induce male reproductive toxicity. Current evidence showed the effects of the chemical exposure on the deterioration of testicular functions, and reduction in epididymal sperm counts. Previous studies showed that PFOA and PFOS displayed a high correlation with each other in seminal plasma levels, but induced different effects on semen variables. In this study, we focused on the comparative toxicity analysis of PFOA and PFOS, using a rat primary Sertoli cell model. Our transcriptomic data showed that PFOA and PFOS treatments (40 μM) perturbed global gene expression. While PFOS induced higher toxicity in affecting cytoskeleton signaling, Sertoli cell-cell junction, and inflammation, underlined by Ingenuity pathway analysis. Immunocytochemical staining revealed that PFOS treatment (40 and 80 μM) induced truncated actin filament and disorganized bundled configuration in the cell cytoplasm. Moreover, disorganized distribution of N-cadherin (N-cad) and β-catenin (β-cat), and defragmentation of ZO-1 at the Sertoli cell-cell interface was evident. At 80 μM of PFOS, cytoplasmic distribution of N-cad, β-cat, and ZO-1 were observed. We then examined whether resveratrol, a polyphenol antioxidant, was able to protect the cells from PFOS toxicity. The pretreatment of Sertoli cells with 10 μM resveratrol prevented the formation of truncated actin filament and dis-localization of β-cat. Western blot analysis showed that Res pretreatment increased the levels of basal ES proteins (N-cad and β-cat), tight junction proteins (ZO-1 and occludin), and gap junction protein, versus control.
Collapse
Affiliation(s)
- Hin Ting Wan
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| | - Keng Po Lai
- Guanxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, P. R. China
| | - Chris Kong Chu Wong
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| |
Collapse
|
35
|
Pouwer MG, Pieterman EJ, Chang SC, Olsen GW, Caspers MPM, Verschuren L, Jukema JW, Princen HMG. Dose Effects of Ammonium Perfluorooctanoate on Lipoprotein Metabolism in APOE*3-Leiden.CETP Mice. Toxicol Sci 2020; 168:519-534. [PMID: 30657992 PMCID: PMC6432869 DOI: 10.1093/toxsci/kfz015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Epidemiological studies have reported positive associations between serum perfluorooctanoic acid (PFOA) and total and non-high-density lipoprotein cholesterol (non-HDL-C) although the magnitude of effect of PFOA on cholesterol lacks consistency. The objectives of this study were to evaluate the effect of PFOA on plasma cholesterol and triglyceride metabolism at various plasma PFOA concentrations relevant to humans, and to elucidate the mechanisms using APOE*3-Leiden.CETP mice, a model with a human-like lipoprotein metabolism. APOE*3-Leiden.CETP mice were fed a Western-type diet with PFOA (10, 300, 30 000 ng/g/d) for 4-6 weeks. PFOA exposure did not alter plasma lipids in the 10 and 300 ng/g/d dietary PFOA dose groups. At 30 000 ng/g/d, PFOA decreased plasma triglycerides (TG), total cholesterol (TC), and non-HDL-C, whereas HDL-C was increased. The plasma lipid alterations could be explained by decreased very low-density lipoprotein (VLDL) production and increased VLDL clearance by the liver through increased lipoprotein lipase activity. The concomitant increase in HDL-C was mediated by decreased cholesteryl ester transfer activity and changes in gene expression of proteins involved in HDL metabolism. Hepatic gene expression and pathway analysis confirmed the changes in lipoprotein metabolism that were mediated for a major part through activation of the peroxisome proliferator-activated receptor (PPAR)α. Our data confirmed the findings from a phase 1 clinical trial in humans that demonstrated high serum or plasma PFOA levels resulted in lower cholesterol levels. The study findings do not show an increase in cholesterol at environmental or occupational levels of PFOA exposure, thereby indicating these findings are associative rather than causal.
Collapse
Affiliation(s)
- Marianne G Pouwer
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, 2333 CK, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Elsbet J Pieterman
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, 2333 CK, Leiden, The Netherlands
| | | | - Geary W Olsen
- Medical Department, 3M Company, Saint Paul, Minnesota 55144
| | - Martien P M Caspers
- The Netherlands Organization of Applied Scientific Research (TNO) - Microbiology and Systems Biology, 3704 HE Zeist, The Netherlands
| | - Lars Verschuren
- The Netherlands Organization of Applied Scientific Research (TNO) - Microbiology and Systems Biology, 3704 HE Zeist, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Hans M G Princen
- The Netherlands Organization of Applied Scientific Research (TNO), Metabolic Health Research, Gaubius Laboratory, 2333 CK, Leiden, The Netherlands
| |
Collapse
|
36
|
Qi W, Clark JM, Timme-Laragy AR, Park Y. Perfluorobutanesulfonic Acid (PFBS) Induces Fat Accumulation in HepG2 Human Hepatoma. TOXICOLOGICAL AND ENVIRONMENTAL CHEMISTRY 2020; 102:585-606. [PMID: 33762794 PMCID: PMC7986581 DOI: 10.1080/02772248.2020.1808894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Per- and poly-fluoroalkyl substances, especially perfluorooctanesulfonic acid, have been extensively used for over 50 years. A growing body of evidence has emerged demonstrating the potential adverse effects of these substances, including its effect on the development of non-alcoholic fatty liver disease, as one of the most prevalent chronic liver diseases. Nonetheless, there is no report of effects of perfluorobutanesulfonic acid, the major replacement for perfluorooctanesulfonic acid, on non-alcoholic fatty liver disease. Therefore, the effects of perfluorobutanesulfonic acid exposure on fat accumulation in a human hepatoma cell line were examined. Cells were exposed to perfluorobutanesulfonic acid with or without 300 μmol/L fatty acid mixture (oleic acid:palmitic acid = 2:1) conjugated by bovine serum albumin as an inducer of steatosis for 48 hours. Perfluorobutanesulfonic acid at 200 μmol/L significantly increased the triglyceride level in the presence of fatty acid compared to the control, but not without fatty acid, which was abolished by a specific peroxisome proliferator-activated receptor gamma antagonist. Perfluorobutanesulfonic acid upregulated key genes controlling lipogenesis and fatty acid uptake. Perfluorobutanesulfonic acid treatment also promoted the production of reactive oxygen species, an endoplasmic reticulum stress marker and cytosolic calcium. In conclusion, perfluorobutanesulfonic acid increased fat accumulation, in part, via peroxisome proliferator-activated receptor gamma-mediated pathway in hepatoma cells.
Collapse
Affiliation(s)
- Weipeng Qi
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, United States
| | - John M. Clark
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, United States
| | - Alicia R. Timme-Laragy
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, 01003, United States
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, United States
- Corresponding author: Phone (413) 545-1018,
| |
Collapse
|
37
|
Attanasio R. Sex differences in the association between perfluoroalkyl acids and liver function in US adolescents: Analyses of NHANES 2013-2016. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:113061. [PMID: 31454574 DOI: 10.1016/j.envpol.2019.113061] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 05/26/2023]
Abstract
Perfluoroalkyl acids (PFAAs) are persistent in the environment, highly bio-accumulative in the body, and likely hepatotoxic in humans. There is evidence of sex-specific physiological responses to PFAA exposure. However, epidemiological studies seldom stratify the analyses by sex. Given the high prevalence of liver disease in general population adolescents, this study was designed to determine whether or not there is association between exposure to PFAAs and biomarkers of liver function in adolescent participants of the 2013-2016 National Health and Nutrition Examination Survey, and whether or not such association is sex-specific. Multivariate linear regressions were performed to examine the association between single PFAAs [perfluorooctane sulfonic acid (PFOS); linear form of perfluorooctanoic acid (PFOA); perfluorohexane sulfonic acid (PFHxS); perfluorononanoic acid (PFNA)], and biomarkers of liver function - gamma glutamyltransferase (GGT), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and total bilirubin. Multivariate logistic regressions were performed to estimate adjusted odd ratios (aOR) of elevated ALT, AST and GGT. The study results show that, in females, there was a positive association of the highest PFOA quartile with increased ALT, AST and GGT, and the highest PFNA quartile with increased ALT and AST. Conversely, in male adolescents there was an association of the highest linear PFOA quartile with decreased ALT, and the highest PFNA quartile with ALT and AST. Females had higher odds of clinically-defined elevated ALT with increased PFOA (aOR = 1.79; 95% CI: 1.05, 3.04) or PFNA (aOR = 2.28; 95% CI: 1.08, 2.28), whereas males had decreased odds of clinically-defined elevated ALT with increased n-PFOA (aOR = 0.43; 95% CI: 0.20, 0.93) or PFNA (aOR = 0.5; 95% CI: 0.28, 0.89). In conclusion, there were sex differences in the association between serum PFAA levels and biomarkers of liver function. These results may provide support for analyzing sex-based adverse effects of PFAAs.
Collapse
Affiliation(s)
- Roberta Attanasio
- Department of Biology, Georgia State University, Petit Science Center - Room 495, 100 Piedmont Ave., Atlanta, GA, USA.
| |
Collapse
|
38
|
Chen A, Jandarov R, Zhou L, Calafat AM, Zhang G, Urbina EM, Sarac J, Augustin DH, Caric T, Bockor L, Petranovic MZ, Novokmet N, Missoni S, Rudan P, Deka R. Association of perfluoroalkyl substances exposure with cardiometabolic traits in an island population of the eastern Adriatic coast of Croatia. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 683:29-36. [PMID: 31129329 PMCID: PMC6581612 DOI: 10.1016/j.scitotenv.2019.05.250] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 05/31/2023]
Abstract
BACKGROUND Exposure to perfluoroalkyl substances (PFAS), ubiquitous environmental contaminants, may be related to cardiometabolic diseases in adults. Studies in European populations to examine the association of PFAS exposure and comprehensive cardiometabolic traits and metabolic syndrome (MetS) are limited. METHODS In this pilot cross-sectional study of a well-characterized adult population of the island of Hvar, situated off the eastern Adriatic coast of Croatia, we measured PFAS concentrations in plasma samples collected during 2007-2008 and examined their cross-sectional associations with cardiometabolic traits and MetS after adjustment of covariates (n = 122). PFAS investigated in this study included perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorohexane sulfonic acid (PFHxS), and perfluorononanoic acid (PFNA). RESULTS The geometric mean (range) was 8.91 (2.36, 33.67) ng/mL for PFOS, 2.87 (1.03, 8.02) ng/mL for PFOA, 0.77 (0.25, 2.40) ng/mL for PFHxS, and 1.29 (0.48, 3.46) ng/mL for PFNA, with frequency of detection at 100%, 100%, 95.9%, and 100%, respectively. PFOS, PFOA, and PFNA concentrations were positively associated with the risk of MetS as defined by the Adult Treatment Panel III (ATP III) criteria, with estimated odds ratios and 95% confidence intervals at 1.89 (0.93, 3.86), 2.19 (0.88, 5.44), and 2.95 (1.12, 7.80), respectively, with only PFNA reaching statistical significance. PFNA concentrations were associated with increased risk of overweight or obesity. CONCLUSIONS Background exposure to PFOS, PFOA, and PFNA was marginally associated with increased risk of MetS in this small study, and these results should be confirmed with a larger sample size and longitudinal follow-up.
Collapse
Affiliation(s)
- Aimin Chen
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Roman Jandarov
- Division of Biostatistics and Bioinformatics, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Li Zhou
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ge Zhang
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Elaine M Urbina
- Heart Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jelena Sarac
- Institute for Anthropological Research, Zagreb, Croatia
| | | | - Tonko Caric
- Institute for Anthropological Research, Zagreb, Croatia
| | - Luka Bockor
- Institute for Anthropological Research, Zagreb, Croatia
| | | | | | - Sasa Missoni
- Institute for Anthropological Research, Zagreb, Croatia; Josip Juraj Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia
| | - Pavao Rudan
- Institute for Anthropological Research, Zagreb, Croatia; Croatian Academy of Sciences and Arts, Zagreb, Croatia
| | - Ranjan Deka
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
39
|
Lv N, Yuan J, Ji A, Shi L, Gao M, Cui L, Jiang Q. Perfluorooctanoic acid-induced toxicities in chicken embryo primary cardiomyocytes: Roles of PPAR alpha and Wnt5a/Frizzled2. Toxicol Appl Pharmacol 2019; 381:114716. [PMID: 31445018 DOI: 10.1016/j.taap.2019.114716] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/25/2019] [Accepted: 08/18/2019] [Indexed: 01/06/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a widespread persistent organic pollutant and may induce developmental toxicities, including developmental cardiotoxicity. To explore the potential mechanism of developmental cardiotoxicity induced by PFOA exposure, chicken embryo primary cardiomyocytes were extracted either from chicken embryos pretreated with PFOA (2 mg/kg), or from untreated embryos and then directly exposed cells to PFOA (1, 10, 30 or 100 μg/ml) in culture. Additionally, peroxisome proliferator activated receptor alpha (PPAR alpha) silencing lentivirus was applied to the embryos on embryonic day (ED2). Cell viability was measured with CCK-8 kit, morphology was assessed with hematoxylin and eosin staining, and intracellular Ca2+ concentrations were determined with Fluo-4 AM probe. Western blotting was utilized to confirm PPAR alpha silencing efficiency and the protein abundance of Wnt5a and Frizzled2. The results indicated that both PFOA pretreatment and direct exposure decreased primary cardiomyocyte viability, altered cell morphology and increased intracellular Ca2+ concentrations. While l-carnitine co-treatment effectively abolished such changes, PPAR alpha silencing only abolished most of the changes in PFOA pretreatment group, but not in cells directly exposed to relatively high doses of PFOA. The protein abundance of Wnt5a and Frizzled2 was increased by PFOA pretreatment, while direct exposure to PFOA increased Frizzled2 abundance but decreased Wnt5a abundance. PPAR alpha silencing resulted in over 50% decrease of PPAR alpha expression level, which abolished the Wnt5a/Frizzled2 expression alterations following PFOA exposure. In conclusion, PFOA-induced primary cardiomyocyte toxicity is associated with PPAR alpha and Wnt5a/Frizzled2, in which PPAR alpha seems to play regulatory roles towards Wnt5a/Frizzled2.
Collapse
Affiliation(s)
- Na Lv
- Department of Pharmacology, School of Pharmacy, Qingdao University, China
| | - Junhua Yuan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, China
| | - Andong Ji
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Limei Shi
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Mengyu Gao
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Lianhua Cui
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, China.
| |
Collapse
|
40
|
Wang C, Zhang Y, Deng M, Wang X, Tu W, Fu Z, Jin Y. Bioaccumulation in the gut and liver causes gut barrier dysfunction and hepatic metabolism disorder in mice after exposure to low doses of OBS. ENVIRONMENT INTERNATIONAL 2019; 129:279-290. [PMID: 31146162 DOI: 10.1016/j.envint.2019.05.056] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 05/23/2023]
Abstract
The compound sodium ρ-perfluorous nonenoxybenzene sulfonate (OBS), a new kind of perfluoroalkyl and polyfluoroalkyl compound, is a surfactant for increasing oil production, and it has been widely detected in various organisms. Because of its wide use, OBS is detectable in the environment. However, knowledge about the biological toxicity of OBS to animals is very limited. Here, male mice were exposed to 0, 0.1, 1 or 10 μg/L of OBS for 6 weeks via drinking water. It was demonstrated that OBS was highly bioaccumulated both in the liver and gut in the mice after low doses of OBS exposure. Curiously, a low dose of OBS exposure also caused gut barrier dysfunction by decreasing mucus secretion and altering Ionic transport in the gut via the CFTR pathway. In addition, liver function was influenced by OBS at both the histopathological and physiological levels. Hepatic transcriptomics and metabolomics analysis showed a total of 1157 genes, and multiple metabolites changed significantly in the livers of mice exposed to low-dose OBS for 6 weeks. The functions of these changed genes and metabolites are tightly related to glycolysis, fatty acid synthesis, fatty acid transport, and β-oxidation. All these results indicate that the liver and gut are important target tissues for OBS exposure. Importantly, it is possible that high levels of bioaccumulation of OBS in the gut and liver might directly cause gut barrier dysfunction and hepatic metabolism disorder in mice.
Collapse
Affiliation(s)
- Caiyun Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yi Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Mi Deng
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China
| | - Xia Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
41
|
Wen X, Baker AA, Klaassen CD, Corton JC, Richardson JR, Aleksunes LM. Hepatic carboxylesterases are differentially regulated in PPARα-null mice treated with perfluorooctanoic acid. Toxicology 2019; 416:15-22. [PMID: 30685356 DOI: 10.1016/j.tox.2019.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/06/2018] [Accepted: 01/18/2019] [Indexed: 01/09/2023]
Abstract
Hepatic carboxylesterases (Ces) catalyze the metabolism of drugs, environmental toxicants, and endogenous lipids and are known to be regulated by multiple nuclear receptors. Perfluorooctanoic acid (PFOA) is a synthetic fluorochemical that has been associated with dyslipidemia in exposed populations. In liver, PFOA can activate nuclear receptors such as PPARα, and alter the metabolism and excretion of chemicals. Here, we sought to test the ability of PFOA to modulate Ces expression and activity in the presence and absence of the PPARα receptor. For this purpose, male C57BL/6 NCrl mice were administered PFOA (1 or 3 mg/kg, po, 7 days) and livers collected for assessment of Ces expression and activity. PFOA increased Ces1 and 2 protein and activity. Notably, PFOA increased Ces1d, 1e, 1f, 1 g, 2c, and 2e mRNAs between 1.5- and 2.5-fold, while it decreased Ces1c and 2b. Activation of PPARα by PFOA was confirmed by up-regulation of Cyp4a14 mRNA. In a separate study of PFOA-treated wild-type (WT) and PPARα-null mice, induction of Ces 1e and 1f mRNA and in turn, Ces1 protein, was PPARα-dependent. Interestingly, in PPARα-null mice, Ces1c, 1d, 1 g, 2a, 2b, and 2e mRNAs and Ces2 protein were up-regulated by PFOA which contributed to sustained up-regulation of Ces activity, although to a lower extent than observed in WT mice. Activation of the CAR and PXR receptors likely accounted for up-regulation of select Ces1 and 2 subtypes in PPARα-null mice. In conclusion, the environmental contaminant PFOA modulates the expression and function of hepatic Ces enzymes, in part through PPARα.
Collapse
Affiliation(s)
- Xia Wen
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, 08854, USA
| | - Angela A Baker
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, 08854, USA
| | - Curtis D Klaassen
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - J Christopher Corton
- Integrated Systems Toxicology Division, NHEERL/ORD, US-EPA, Research Triangle Park, NC, 27711, USA
| | - Jason R Richardson
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, 08854, USA; Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
42
|
Sakuma A, Wasada Ochi H, Yoshioka M, Yamanaka N, Ikezawa M, Guruge KS. Changes in hepato-renal gene expression in microminipigs following a single exposure to a mixture of perfluoroalkyl acids. PLoS One 2019; 14:e0210110. [PMID: 30608957 PMCID: PMC6319762 DOI: 10.1371/journal.pone.0210110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/16/2018] [Indexed: 01/09/2023] Open
Abstract
It is evident that some perfluoroalkyl acids (PFAAs), a group of globally dispersed pollutants, have long biological half-lives in humans and farm animals. However, the effects of PFAAs in domestic animals have not been fully elucidated. The present study investigated how exposure to a single dose of a mixture of 10 PFAAs influenced hepatic and renal gene expression and histopathology, as well as plasma clinical biochemistry, in microminipigs (MMPigs) over 21 days. In animals treated with PFAAs, the mRNA expression of twelve genes related to fatty acid metabolism was upregulated in the kidney, while only few of these genes were induced in the liver. The expression of several kidney injury-associated genes such as, IGFBP1, IGFBP6, GCLC X2, GCLC X3, MSGT1, OLR1 was upregulated in the kidney. Interestingly, the expression of IGFBP-genes was differentially altered in the liver and kidney. Our findings thus identified hepato-renal gene expression changes in MMPigs that were associated with various molecular pathways including peroxisome proliferation, lipid metabolism, kidney injury, and apoptosis. Furthermore, serum HDL levels were significantly decreased following exposure to PFAAs, whereas no significant histopathological changes were detected, as compared to the vehicle group. Taken together, the present study provided the first indication that a single exposure to a mixture of PFAAs can produce changes in MMPig renal gene expression that were observed three weeks post exposure, suggesting that more attention should be paid to the kidney as a primary target organ of PFAAs.
Collapse
Affiliation(s)
- Akiko Sakuma
- Miyagi Prefectural Sendai Livestock Hygiene Service Center, Anyoji, Miyagino, Sendai, Miyagi, Japan
- Cellular Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Aramaki Aza Aoba, Aoba, Sendai, Miyagi, Japan
| | - Haruyo Wasada Ochi
- Kumamoto Prefectural Central Livestock Hygiene Service Center, Jyounan-mahi Shizume, Minami, Kumamoto, Kumamoto, Japan
| | - Miyako Yoshioka
- National Institute of Animal Health, National Agriculture and Food Research Organization, Kannondai, Tsukuba, Ibaraki, Japan
| | - Noriko Yamanaka
- National Institute of Animal Health, National Agriculture and Food Research Organization, Kannondai, Tsukuba, Ibaraki, Japan
| | - Mitsutaka Ikezawa
- National Institute of Animal Health, National Agriculture and Food Research Organization, Kannondai, Tsukuba, Ibaraki, Japan
| | - Keerthi S. Guruge
- National Institute of Animal Health, National Agriculture and Food Research Organization, Kannondai, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka, Japan
- Centre for Crop Health, University of Southern Queensland, Toowoomba Campus, Queensland, Australia
| |
Collapse
|
43
|
Zhang H, Zhou X, Sheng N, Cui R, Cui Q, Guo H, Guo Y, Sun Y, Dai J. Subchronic Hepatotoxicity Effects of 6:2 Chlorinated Polyfluorinated Ether Sulfonate (6:2 Cl-PFESA), a Novel Perfluorooctanesulfonate (PFOS) Alternative, on Adult Male Mice. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:12809-12818. [PMID: 30256107 DOI: 10.1021/acs.est.8b04368] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The compound 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFESA), an alternative to perfluorooctanesulfonate (PFOS) in the metal-plating industry, has been widely detected in various environmental matrices. However, its hepatotoxicity has yet to be clarified. Here, male mice were exposed to 0.04, 0.2, or 1 mg/kg/day of 6:2 Cl-PFESA for 56 days. Results demonstrated that relative liver weight increased significantly in the 0.2 and 1 mg/kg/day 6:2 Cl-PFESA groups, whereas liver lipid accumulation increased in all 6:2 Cl-PFESA groups. Serum enzyme activities of alanine transaminase and alkaline phosphatase were increased. Serum triglycerides and low-density lipoprotein cholesterol both increased, whereas serum total cholesterol and high-density lipoprotein cholesterol decreased following 6:2 Cl-PFESA exposure. A total of 264 differentially expressed proteins (127 up-regulated and 137 down-regulated), mainly involved in lipid metabolism, xenobiotic metabolism, and ribosome biogenesis, were identified by quantitative proteomics. Bioinformatics analysis highlighted the de-regulation of PPAR and PXR, which may contribute to the hepatotoxicity of 6:2 Cl-PFESA. Additionally, 6:2 Cl-PFESA induced both cell apoptosis and proliferation in the mouse liver. Compared to the overt toxicity of PFOS, 6:2 Cl-PFESA exhibited more-serious hepatotoxicity. Thus, caution should be exercised in the application of 6:2 Cl-PFESA as a replacement alternative to PFOS in industrial areas.
Collapse
Affiliation(s)
- Hongxia Zhang
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| | - Xiujuan Zhou
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| | - Nan Sheng
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| | - Ruina Cui
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| | - Qianqian Cui
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| | - Hua Guo
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry , Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences , Shanghai 200032 , China
| | - Yan Sun
- Key Laboratory of Organofluorine Chemistry , Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences , Shanghai 200032 , China
| | - Jiayin Dai
- Key Laboratory of Animal Ecology and Conservation Biology , Institute of Zoology, Chinese Academy of Sciences , Beijing 100101 , China
| |
Collapse
|
44
|
Huck I, Beggs K, Apte U. Paradoxical Protective Effect of Perfluorooctanesulfonic Acid Against High-Fat Diet-Induced Hepatic Steatosis in Mice. Int J Toxicol 2018; 37:383-392. [PMID: 30134762 PMCID: PMC6150807 DOI: 10.1177/1091581818790934] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Perfluorooctanesulfonic acid (PFOS) is a persistent organic pollutant with worldwide bioaccumulation due to a very long half-life. Perfluorooctanesulfonic acid exposure results in significant hepatic effects including steatosis, proliferation, hepatomegaly, and in rodents, carcinogenesis. The objective of this study was to determine whether PFOS exposure exacerbates nonalcoholic fatty liver disease and nonalcoholic steatohepatitis pathogenesis. Eight-week-old male C57BL/6 J mice (n = 5 per group) were fed ad libitum normal chow diet (ND) alone, 60% high-fat diet (HFD) alone, ND + PFOS, and HFD + PFOS (0.0001% w/w (1 mg/kg) of PFOS) for 6 weeks. Both HFD alone and the ND + PFOS treatment induced significant adiposity and hepatomegaly, but the HFD + PFOS treatment showed a marked protection. Oil Red O staining and quantitative analysis of hepatic lipid content revealed increased hepatic steatosis in ND + PFOS and in HFD alone fed mice, which was prevented in HFD + PFOS treatment. Further studies revealed that ND + PFOS treatment significantly affected expression of lipid trafficking genes to favor steatosis, but these changes were absent in HFD + PFOS group. Specifically, expression of CD36, the major lipid importer in the cells, and peroxisome proliferator-activated receptor gamma (PPARγ), its major regulator, were induced in HFD + no treatment (NT) and ND + PFOS-fed mice but remained unchanged in HFD + PFOS mice. In conclusion, these data indicate that coadministration of PFOS with HFD mitigates steatosis and hepatomegaly induced by HFD and that by PFOS fed in ND diet via regulation of cellular lipid import machinery. These findings suggest dietary lipid content be considered when performing risk management of PFOS in humans and the elucidation of PFOS-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ian Huck
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kevin Beggs
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Udayan Apte
- 1 Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
45
|
Jacobsen AV, Nordén M, Engwall M, Scherbak N. Effects of perfluorooctane sulfonate on genes controlling hepatic fatty acid metabolism in livers of chicken embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:23074-23081. [PMID: 29860686 PMCID: PMC6096545 DOI: 10.1007/s11356-018-2358-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/22/2018] [Indexed: 04/15/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are synthetic surfactants with a wide variety of applications; however, due to their stability, they are particularly resistant to degradation and, as such, are classed as persistent organic pollutants. Perfluorooctane sulfonate (PFOS) is one such PFAS that is still detectable in a range of different environmental settings, despite its use now being regulated in numerous countries. Elevated levels of PFOS have been detected in various avian species, and the impact of this on avian health is of interest when determining acceptable levels of PFOS in the environment. Due to its similarities to naturally occurring fatty acids, PFOS has potential to disrupt a range of biological pathways, particularly those associated with lipid metabolism, and this has been shown in various species. In this study, we have investigated how in ovo exposure to environmentally relevant levels of PFOS affects expression of genes involved in lipid metabolism of developing chicken embryos. We have found a broad suppression of transcription of genes involved in fatty acid oxidation and PPAR-mediated transcription with more significant effects apparent at lower doses of PFOS. These results highlight the need for more research investigating the biological impacts of low levels of PFAS to properly inform environmental policy governing their regulation.
Collapse
Affiliation(s)
- Annette V Jacobsen
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, Australia
- The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
- The Walter and Eliza Hall Institute, Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Marcus Nordén
- MTM Research Center, School of Science and Technology, Örebro University, Örebro, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Magnus Engwall
- MTM Research Center, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Nikolai Scherbak
- The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden.
- MTM Research Center, School of Science and Technology, Örebro University, Örebro, Sweden.
| |
Collapse
|
46
|
Bangma JT, Reiner JL, Lowers RH, Cantu TM, Scott J, Korte JE, Scheidt DM, McDonough C, Tucker J, Back B, Adams DH, Bowden JA. Perfluorinated alkyl acids and fecundity assessment in striped mullet (Mugil cephalus) at Merritt Island national wildlife refuge. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 619-620:740-747. [PMID: 29161599 PMCID: PMC7707152 DOI: 10.1016/j.scitotenv.2017.11.141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 05/22/2023]
Abstract
This study investigated wild caught striped mullet (Mugil cephalus) at Merritt Island National Wildlife Refuge (MINWR) for levels of 15 perfluoroalkyl acids (PFAA) in tandem with individual fecundity measurements (Oocyte sub-stage 2 late, n=42) and oocyte reproductive stages (Stages 1-5, n=128). PFAA measurements were quantified in striped mullet liver (n=128), muscle (n=49), and gonad (n=10). No significant negative impacts of liver PFAA burden on wild-caught, mullet fecundity endpoints were observed in this study; however, changes in PFAA were observed in the liver as mullet progressed through different sub-stages of oocyte development. Of the PFAA with significant changes by sub-stage of oocyte development, the carboxylic acids (perfluorooctanoic acid, perfluorononanoic acid, and perfluorotridecanoic acid) increased in the liver with increasing sub-stage while the sulfonic acid and its precursor (perfluorooctanesulfonic acid (PFOS) and perfluorooctanesulfonamide, respectively) decreased in the liver with increasing sub-stage of oocyte development. This is a unique find and suggests PFAA change location of compartmentalization as mullet progress towards spawning. Investigations also revealed higher than expected median muscle and gonad levels of PFOS in striped mullet collected at MINWR (9.01ng/g and 80.2ng/g, respectively).
Collapse
Affiliation(s)
- Jacqueline T Bangma
- Medical University of South Carolina, Department of Obstetrics and Gynecology, 221 Fort Johnson Road, Charleston, SC 29412, USA
| | - Jessica L Reiner
- National Institute of Standards and Technology, Chemical Sciences Division, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA
| | - Russell H Lowers
- Integrated Mission Support Service (IMSS), Kennedy Space Center, FL, USA
| | - Theresa M Cantu
- Medical University of South Carolina, Department of Obstetrics and Gynecology, 221 Fort Johnson Road, Charleston, SC 29412, USA
| | - Jacob Scott
- Medical University of South Carolina, Department of Obstetrics and Gynecology, 221 Fort Johnson Road, Charleston, SC 29412, USA
| | - Jeffrey E Korte
- Medical University of South Carolina, Charleston, Department of Public Health Sciences, 135 Cannon Street, SC 29412, USA
| | - Doug M Scheidt
- Integrated Mission Support Service (IMSS), Kennedy Space Center, FL, USA
| | - Chris McDonough
- Marine Resources Division, South Carolina Department of Natural Resources, Charleston, SC, USA
| | - Jonathan Tucker
- Marine Resources Division, South Carolina Department of Natural Resources, Charleston, SC, USA
| | - Brenton Back
- Integrated Mission Support Service (IMSS), Kennedy Space Center, FL, USA
| | - Douglas H Adams
- Florida Fish & Wildlife Conservation Commission, Fish & Wildlife Research Institute, Melbourne, FL, USA
| | - John A Bowden
- National Institute of Standards and Technology, Chemical Sciences Division, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA.
| |
Collapse
|
47
|
Frawley RP, Smith M, Cesta MF, Hayes-Bouknight S, Blystone C, Kissling GE, Harris S, Germolec D. Immunotoxic and hepatotoxic effects of perfluoro-n-decanoic acid (PFDA) on female Harlan Sprague–Dawley rats and B6C3F1/N mice when administered by oral gavage for 28 days. J Immunotoxicol 2018. [DOI: 10.1080/1547691x.2018.1445145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Rachel P. Frawley
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Matthew Smith
- Richard Bland College of William & Mary, South Prince George, VA, USA
| | - Mark F. Cesta
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Chad Blystone
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Grace E. Kissling
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Shawn Harris
- Social & Scientific Systems, Inc., Durham, NC, USA
| | - Dori Germolec
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
48
|
Corton JC, Peters JM, Klaunig JE. The PPARα-dependent rodent liver tumor response is not relevant to humans: addressing misconceptions. Arch Toxicol 2017; 92:83-119. [PMID: 29197930 DOI: 10.1007/s00204-017-2094-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
A number of industrial chemicals and therapeutic agents cause liver tumors in rats and mice by activating the nuclear receptor peroxisome proliferator-activated receptor α (PPARα). The molecular and cellular events by which PPARα activators induce rodent hepatocarcinogenesis have been extensively studied elucidating a number of consistent mechanistic changes linked to the increased incidence of liver neoplasms. The weight of evidence relevant to the hypothesized mode of action (MOA) for PPARα activator-induced rodent hepatocarcinogenesis is summarized here. Chemical-specific and mechanistic data support concordance of temporal and dose-response relationships for the key events associated with many PPARα activators. The key events (KE) identified in the MOA are PPARα activation (KE1), alteration in cell growth pathways (KE2), perturbation of hepatocyte growth and survival (KE3), and selective clonal expansion of preneoplastic foci cells (KE4), which leads to the apical event-increases in hepatocellular adenomas and carcinomas (KE5). In addition, a number of concurrent molecular and cellular events have been classified as modulating factors, because they potentially alter the ability of PPARα activators to increase rodent liver cancer while not being key events themselves. These modulating factors include increases in oxidative stress and activation of NF-kB. PPARα activators are unlikely to induce liver tumors in humans due to biological differences in the response of KEs downstream of PPARα activation. This conclusion is based on minimal or no effects observed on cell growth pathways and hepatocellular proliferation in human primary hepatocytes and absence of alteration in growth pathways, hepatocyte proliferation, and tumors in the livers of species (hamsters, guinea pigs and cynomolgus monkeys) that are more appropriate human surrogates than mice and rats at overlapping dose levels. Despite this overwhelming body of evidence and almost universal acceptance of the PPARα MOA and lack of human relevance, several reviews have selectively focused on specific studies that, as discussed, contradict the consensus opinion and suggest uncertainty. In the present review, we systematically address these most germane suggested weaknesses of the PPARα MOA.
Collapse
Affiliation(s)
- J Christopher Corton
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr, MD-B105-03, Research Triangle Park, NC, 27711, USA.
| | - Jeffrey M Peters
- The Department of Veterinary and Biomedical Sciences and Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, 16803, USA
| | - James E Klaunig
- Department of Environmental Health, Indiana University, Bloomington, IN, 47402, USA
| |
Collapse
|
49
|
Lai KP, Lee JCY, Wan HT, Li JW, Wong AYM, Chan TF, Oger C, Galano JM, Durand T, Leung KS, Leung CC, Li R, Wong CKC. Effects of in Utero PFOS Exposure on Transcriptome, Lipidome, and Function of Mouse Testis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:8782-8794. [PMID: 28654245 DOI: 10.1021/acs.est.7b02102] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Transcriptomic and LC-MS/MS-based targeted lipidomic analyses were conducted to identify the effects of in utero PFOS exposure on neonatal testes and its relation to testicular dysfunction in adult offspring. Pregnant mice were orally administered 0.3 and 3 μg PFOS/g body weight until term. Neonatal testes (P1) were collected for the detection of PFOS, and were subjected to omics study. Integrated pathway analyses using DAVID, KEGG, and IPA underlined the effects of PFOS exposure on lipid metabolism, oxidative stress and cell junction signaling in testes. LC-MS/MS analysis showed that the levels of adrenic acid and docosahexaenoic acid (DHA) in testes were significantly reduced in the PFOS treatment groups. A significant linear decreasing trend in eicosapentaenoic acid and DHA with PFOS concentrations was observed. Moreover, LOX-mediated 5-hydroxyeicosatetraenoic acids (HETE) and 15-HETE from arachidonic acid in the testes were significantly elevated and a linear increasing trend of 15-HETE concentrations was detected with doses of PFOS. The perturbations of lipid mediators suggested that PFOS has potential negative impacts on testicular functions. Postnatal analysis of male offspring at P63 showed significant reductions in serum testosterone and epididymal sperm count. This study sheds light into the as yet unrevealed action of PFOS on lipid mediators in affecting testicular functions.
Collapse
Affiliation(s)
- Keng Po Lai
- Department of Biology and Chemistry, City University of Hong Kong , Hong Kong
| | | | - Hin Ting Wan
- Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University , Hong Kong
| | - Jing Woei Li
- School of Life Sciences, The Chinese University of Hong Kong , Hong Kong
| | - Aman Yi-Man Wong
- Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University , Hong Kong
| | - Ting Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong , Hong Kong
| | - Camille Oger
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS, ENSCM, Université de Montpellier , Montpellier, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS, ENSCM, Université de Montpellier , Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS, ENSCM, Université de Montpellier , Montpellier, France
| | - Kin Sum Leung
- School of Biological Sciences, The University of Hong Kong , Hong Kong
| | - Cherry C Leung
- Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University , Hong Kong
| | - Rong Li
- Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University , Hong Kong
| | - Chris Kong-Chu Wong
- Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University , Hong Kong
| |
Collapse
|
50
|
Chiu WA, Guyton KZ, Martin MT, Reif DM, Rusyn I. Use of high-throughput in vitro toxicity screening data in cancer hazard evaluations by IARC Monograph Working Groups. ALTEX 2017; 35:51-64. [PMID: 28738424 PMCID: PMC5783793 DOI: 10.14573/altex.1703231] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023]
Abstract
Evidence regarding carcinogenic mechanisms serves a critical role in International Agency for Research on Cancer (IARC) Monograph evaluations. Three recent IARC Working Groups pioneered inclusion of the US Environmental Protection Agency (EPA) ToxCast program high-throughput screening (HTS) data to supplement other mechanistic evidence. In Monograph V110, HTS profiles were compared between perfluorooctanoic acid (PFOA) and prototypical activators across multiple nuclear receptors. For Monograph V112-113, HTS assays were mapped to 10 key characteristics of carcinogens identified by an IARC expert group, and systematically considered as an additional mechanistic data stream. Both individual assay results and ToxPi-based rankings informed mechanistic evaluations. Activation of multiple nuclear receptors in HTS assays showed that PFOA targets not only peroxisome proliferator activated receptors, but also other receptors. ToxCast assays substantially covered 5 of 10 key characteristics, corroborating literature evidence of "induces oxidative stress" and "alters cell proliferation, cell death or nutrient supply" and filling gaps for "modulates receptor-mediated effects." Thus, ToxCast HTS data were useful both in evaluating specific mechanistic hypotheses and in contributing to the overall evaluation of mechanistic evidence. However, additional HTS assays are needed to provide more comprehensive coverage of the 10 key characteristics of carcinogens that form the basis of current IARC mechanistic evaluations.
Collapse
Affiliation(s)
- Weihsueh A. Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Kathryn Z. Guyton
- Monographs Programme, International Agency for Research on Cancer, Lyon, France
| | - Matthew T. Martin
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - David M. Reif
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|