1
|
Rodriguez-Carrillo A, Vela-Soria F, Smagulova F, Fernández MF, Freire C. Association between PFAS exposure and metabolic-related biomarkers in Spanish adolescents. ENVIRONMENTAL RESEARCH 2025; 273:121171. [PMID: 39978619 DOI: 10.1016/j.envres.2025.121171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) exert endocrine disruptive effects on the endocrine-metabolic axis. Emerging knowledge suggests that kisspeptin may play a key role in these effects. OBJECTIVE To assess the cross-sectional association of blood PFAS concentrations with kisspeptin levels, KISS1 gene DNA methylation, and metabolic-related biomarkers in adolescent males from the Spanish INMA-Granada cohort. METHODS Seven PFAS and twelve biomarkers (glucose-GLU, total cholesterol-TC, triglycerides, LDL, HDL, ALP, AST, ALT, GGT, total bilirubin-BILT, direct bilirubin-BILD, and urea) were measured in plasma and serum, respectively, from 129 adolescent males (15-17 yrs). Systolic and diastolic blood pressure (SBP, DBP), pulse, z-scored body mass index, kisspeptin protein levels (n = 104) and whole blood KISS1 DNA methylation (n = 117) were determined. Linear regression models, weighted quantile sum (WQS), and Bayesian kernel machine (BKMR) were fit. RESULTS PFHpA was associated with lower GLU levels [% change per log-unit increase in plasma concentrations (95%CI) = -4.73 (-8.98;-0.28)], and PFUnDA with higher GLU, TC, and HDL levels. In models adjusted by kisspeptin level, PFOS was associated with higher SBP [3.42 (-0.12; 7.09)]. Additionally, PFNA and total PFAS concentrations were associated with higher kisspeptin levels [3.91 (0.55; 7.37) and 6.14 (0.47; 12.13), respectively]. Mixture models showed that combined PFAS exposure was associated with higher HDL, lower hepatic biomarkers (ALT, BILD) and higher kisspeptin levels. CONCLUSION Certain PFAS (e.g. PFUnDA) and their mixture were associated with metabolic-related biomarkers, mainly GLU, HDL, and SBP. These associations may be influenced by kisspeptin levels. More studies are needed to verify these observations.
Collapse
Affiliation(s)
- Andrea Rodriguez-Carrillo
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, 2400, Belgium; Toxicological Centre, University of Antwerp, Universiteitsplein, 1, Wilrijk, 2610, Belgium; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| | - Fernando Vela-Soria
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain
| | - Fatima Smagulova
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, Rennes, F-35000, France
| | - Mariana F Fernández
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, Granada, 18016, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Carmen Freire
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Legal Medicine, Toxicology, and Physical Anthropology, School of Medicine, University of Granada, Granada, 18016, Spain.
| |
Collapse
|
2
|
Tu L, Yang Y, Lin Y, Wang X, Peng H, Chen B, Zhang R, Luan T. Time-Course Strategy Reveals a Dual Potential of Perfluorooctanoic Acid and Its Alternative in Adipocyte Differentiation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:56-68. [PMID: 39720916 DOI: 10.1021/acs.est.4c06425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Exposure to perfluorooctanoic acid (PFOA) and hexafluoropropylene oxide dimer acid (HFPO-DA) was associated with adipogenesis. However, potential mechanisms remain to be elucidated. Herein, a 3T3-L1 adipocyte model was used to explore the dynamic changes in adipocyte differentiation (2, 4, and 8 days) under PFOA and HFPO-DA exposure. PFOA and HFPO-DA increased the adipocyte formation rate and intracellular levels of triglycerides (TG). Meanwhile, adipocyte browning was induced by PFOA and HFPO-DA, which was characterized by small lipid droplets, low levels of TG per adipocyte, increased ATP levels, and elevated mitochondrial respiration activities with time-dependent differentiation. The browning potency indexes of PFOA and HFPO-DA were approximately 1.5 times higher than those of the controls. Time-course transcriptomics analysis showed that PFOA and HFPO-DA activated the biological process of adipocyte browning but different gene expression patterns regulated adipocyte browning. Only overexpressed hydroxymethylglutaryl-CoA synthase (Hmgcs2) was shared between PFOA and HFPO-DA groups from 2 to 8 days. Hmgcs2 could regulate adipocyte browning induced by PFOA and HFPO-DA, and this observation was lost when Hmgcs2 was knocked down. Our study suggests that PFOA and HFPO-DA could play dual roles in the differentiation of 3T3-L1 adipocytes, and Hmgcs2 might be a target of PFOA- and HFPO-DA-induced adipocyte browning.
Collapse
Affiliation(s)
- Lanyin Tu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yuanzhu Yang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yingshi Lin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Xucong Wang
- School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Hongyu Peng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200020, China
| | - Baowei Chen
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
| | - Ruijia Zhang
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, China
| | - Tiangang Luan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Jiangmen Key Laboratory of Synthetic Chemistry and Cleaner Production, School of Environmental and Chemical Engineering, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
3
|
Wu S, Wang L, Schlenk D, Liu J. Machine Learning-Based Toxicological Modeling for Screening Environmental Obesogens. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18133-18144. [PMID: 39359054 DOI: 10.1021/acs.est.4c05070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
The emerging presence of environmental obesogens, chemicals that disrupt energy balance and contribute to adipogenesis and obesity, has become a major public health challenge. Molecular initiating events (MIEs) describe biological outcomes resulting from chemical interactions with biomolecules. Machine learning models based on MIEs can predict complex toxic end points due to chemical exposure and improve the interpretability of models. In this study, a system was constructed that integrated six MIEs associated with adipogenesis and obesity. This system showed high accuracy in external validation, with an area under the receiver operating characteristic curve of 0.78. Molecular hydrophobicity (SlogP_VSA) and direct electrostatic interactions (PEOE_VSA) were identified as the two most critical molecular descriptors representing the obesogenic potential of chemicals. This system was further used to predict the obesogenic effects of chemicals on the candidate list of substances of very high concern (SVHCs). Results from 3T3-L1 adipogenesis assays verified that the system correctly predicted obesogenic or nonobesogenic effects of 10 of the 12 SVHCs tested, and identified four novel potential obesogens, including 2-benzotriazol-2-yl-4,6-ditert-butylphenol (UV-320), 4-(1,1,5-trimethylhexyl)phenol (p262-NP), 2-[4-(1,1,3,3-tetramethylbutyl)phenoxy]ethanol (OP1EO) and endosulfan. These validation data suggest that the screening system has good performance in adipogenic prediction.
Collapse
Affiliation(s)
- Siying Wu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Linping Wang
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Jing Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
4
|
Pederick JL, Frkic RL, McDougal DP, Bruning JB. A structural basis for the activation of peroxisome proliferator-activated receptor gamma (PPARγ) by perfluorooctanoic acid (PFOA). CHEMOSPHERE 2024; 354:141723. [PMID: 38494006 DOI: 10.1016/j.chemosphere.2024.141723] [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: 01/17/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Perfluorooctanoic acid (PFOA) is a widespread environmental pollutant of the perfluoroalkyl substance (PFAS) class that is extremely resistant to environmental and metabolic degradation, leading to bioaccumulation. PFOA exposure has been linked to many health effects including endocrine disruption and metabolic dysregulation, but our understanding of the molecular mechanisms resulting in these outcomes remains incomplete. One target affected by PFOA is the ligand regulated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) which plays a critical role in controlling metabolic homeostasis through regulating processes such as adipogenesis, glucose homeostasis, inflammation and osteogenesis. It has been previously established that PFOA activates PPARγ through binding to the PPARγ ligand binding domain (PPARγ LBD) leading to increased expression of PPARγ controlled target genes. However, the mechanism by which PFOA achieves this has remained elusive. Here, we employed a combination of X-ray crystallography and fluorescence polarization assays to provide a structural basis for PFOA mediated activation of PPARγ via binding to the PPARγ LBD. Using X-ray crystallography, the cocrystal structure of the PPARγ LBD:PFOA complex was solved. This revealed that PFOA occupies three distinct sites, two within the PPARγ LBD and one within the activation function 2 (AF2) on the protein surface. Structural comparison of PFOA binding with previously reported PPARγ:ligand complexes supports that PFOA activates PPARγ by a partial agonist mechanism at micromolar concentrations. Fluorescence polarization assays also revealed that PFOA binding to the AF2 is unlikely to occur in a cellular context and confirmed that PFOA behaves as a partial agonist in vitro, weakly recruiting a coactivator peptide to the AF2 of the PPARγ LBD. This discovery provides an advancement in understanding PFOA mediated regulation of PPARγ, giving new insight regarding regulation of PPARγ by PFAS and PFAS substitutes in general and can be applied to the design and assessment of safer PFAS.
Collapse
Affiliation(s)
- J L Pederick
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, SA, Australia
| | - R L Frkic
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, SA, Australia
| | - D P McDougal
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, SA, Australia
| | - J B Bruning
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, SA, Australia.
| |
Collapse
|
5
|
Chen LW, Ng S, Tint MT, Michael N, Sadananthan SA, Ong YY, Yuan WL, Chen ZY, Chen CY, Godfrey KM, Tan KH, Gluckman PD, Chong YS, Eriksson JG, Yap F, Lee YS, Fortier MV, Velan SS, Chan SY. Associations of cord plasma per- and polyfluoroakyl substances (PFAS) with neonatal and child body composition and adiposity: The GUSTO study. ENVIRONMENT INTERNATIONAL 2024; 183:108340. [PMID: 38043321 DOI: 10.1016/j.envint.2023.108340] [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: 08/29/2023] [Revised: 10/26/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND The influence of prenatal exposure to per- and poly- fluoroalkyl substances (PFAS) on birth size and offspring adiposity is unclear, especially for the newer, shorter-chained replacement PFAS. METHODS In the GUSTO multi-ethnic Singaporean mother-offspring cohort, 12 PFAS were measured in 783 cord plasma samples using ultra-performance-liquid chromatography-tandem-mass-spectrometer (UPLC-MS/MS). Outcomes included offspring anthropometry, other indicators of body composition/metabolic health, and MRI-derived abdominal adiposity (subset) at birth and 6 years of age. PFAS were modeled individually, in categories of long-chain and short-chain PFAS, and as scores of three principal components (PC) derived using PC analysis (PC1, PC2, and PC3 reflect predominant exposure patterns to "very-long-PFAS", "long-PFAS", and "short-PFAS", respectively). Associations with outcomes were assessed using multivariable linear regressions, adjusted for important covariates such as maternal sociodemographic and lifestyle factors. RESULTS Overall, cord PFAS levels showed either no or positive associations (mostly for long-chain PFAS) with birth weight, length and head circumference. In general, PFAS were associated with higher neonatal abdominal adiposity, driven by shorter-chain PFAS. Perfluoroheptanoic acid (PFHpA) was associated with higher volumes of superficial subcutaneous adipose tissue (sSAT) (3.75 [1.13, 6.37] mL per SD increase in PFAS) and internal adipose tissue (IAT) (1.39 [0.41, 2.38] mL). Higher levels of perfluorobutanesulfonic acid (PFBS), short-chain PFAS, and PC3 were associated with higher IAT volume (β range 1.22-1.41 mL/SD, all P < 0.02), especially in girls. Higher PC3 score was additionally associated with higher sSAT (3.12 [0.45, 5.80] mL) volume. At age 6 years, most observed associations did not persist. No consistent associations were observed between PFAS and whole-body adiposity measures. CONCLUSIONS Fetal exposure to emerging short-chain PFAS was associated with higher abdominal adiposity at birth but not at age 6 years. Further research is needed to replicate the findings and to determine if these effects may reappear beyond early childhood. Population exposure to newer PFAS and consequent health impact must be monitored.
Collapse
Affiliation(s)
- Ling-Wei Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, No. 17 Xu-Zhou Road, Taipei 10055, Taiwan; Master of Public Health Program, College of Public Health, National Taiwan University, No. 17 Xu-Zhou Road, Taipei 10055, Taiwan; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore
| | - Sharon Ng
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore
| | - Mya-Thway Tint
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore; Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore
| | - Yi Ying Ong
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore; Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Wen Lun Yuan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore; Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004 Paris, France
| | - Ze-Ying Chen
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17 Xu-Zhou Road, Taipei 10055, Taiwan
| | - Chia-Yang Chen
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17 Xu-Zhou Road, Taipei 10055, Taiwan
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre & NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Tremona Road, SO16 6YD Southampton, UK
| | - Kok Hian Tan
- Duke-NUS Medical School, 8 College Road, 169857, Singapore; Department of Reproductive Medicine, KK Women's and Children Hospital, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore; Liggins Institute, University of Auckland, 85 Park Rd, Grafton, Auckland 1023, New Zealand
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore; Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore; Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore; Department of General Practice and Primary Health Care, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland; Folkhälsan Research Center, Topeliusgatan 20, 00250 Helsinki, Finland
| | - Fabian Yap
- Duke-NUS Medical School, 8 College Road, 169857, Singapore; Department of Pediatric Endocrinology, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899, Singapore
| | - Yung Seng Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Health System, 1E Kent Ridge Road, 119228, Singapore
| | - Marielle V Fortier
- Department of Diagnostic & Interventional Imaging, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899, Singapore
| | - Sendhil S Velan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, 30 Medical Drive, 117609, Singapore; Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore.
| |
Collapse
|
6
|
Zheng L, Wang Z, Yang R, Chen W, Zhang J, Li R, Lv W, Lin B, Luo J. The interference between effects of PFAS exposure on thyroid hormone disorders and cholesterol levels: an NHANES analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:90949-90959. [PMID: 37468783 DOI: 10.1007/s11356-023-28739-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Studies have documented that per- and polyfluoroalkyl substance (PFAS) exposures are associated with thyroid hormones (TH) and lipid levels. This study investigates whether these effects interfere with each other. We analyzed data on 3954 adults in the US National Health and Nutrition Examination Survey (NHANES; 2007-2012). TH disorder was defined using thyroid hormones. Serum high-density lipoprotein (HDL) cholesterol, total cholesterol, and six types of PFAS were included. Weighted quantile sum (WQS) regression was used to estimate the overall effect of PFAS mixture on TH disorder and cholesterols, respectively. Potential confounders, including age, race, gender, education, household poverty, smoking, and alcohol drinking, were adjusted. PFAS mixture was associated increased risk for TH disorder (odds ratio = 1.21, 95% confidence interval (CI): 1.02, 1.43), higher HDL cholesterol (linear coefficient = 1.31, 95% CI: 0.50, 2.11), and higher total cholesterol (linear coefficient = 5.30, 95% CI: 3.40, 7.21). TH disorder was associated with higher HDL cholesterol (linear coefficient = 2.30, 95% CI: 0.50, 2.11), but not total cholesterol. When adjusted for TH disorder, the effect estimates of PFAS mixture remain roughly unchanged on HDL cholesterol (linear coefficient = 1.13, 95% CI: 0.28, 1.98) and total cholesterol (linear coefficient = 5.61, 95% CI: 3.58, 7.63). Sex modified the effect of PFAS mixture on HDL cholesterol (P for interaction: 0.04) but did not change the interaction between PFAS and TH disorder on cholesterols. We corroborated the adverse health effects of PFAS exposure on TH and lipids; however, these two effects appear to be independent of and not interfere with each other.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Thyroid Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Zhecun Wang
- Department of Vascular Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Rui Yang
- Department of Urology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Wanna Chen
- Department of Thyroid Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Jing Zhang
- Department of Thyroid Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Ruixia Li
- Department of Thyroid Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Weiming Lv
- Department of Thyroid Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Bo Lin
- Department of Thyroid Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Jiajun Luo
- Institute for Population and Precision Health, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
7
|
Wu S, Xie J, Zhao H, Sanchez O, Zhao X, Freeman JL, Yuan C. Pre-differentiation GenX exposure induced neurotoxicity in human dopaminergic-like neurons. CHEMOSPHERE 2023; 332:138900. [PMID: 37172627 DOI: 10.1016/j.chemosphere.2023.138900] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
GenX, also known as hexafluoropropylene oxide dimer acid (HFPO) was introduced as a safer alternative to perfluorooctanoic acid (PFOA) in 2009. After nearly two decades of applications there are increasing safety concerns about GenX due to its association with various organ damages. Few studies, however, have systematically assessed the molecular neurotoxicity of low-dose GenX exposure. Here, we evaluated the effects of pre-differentiation exposure of GenX on dopaminergic (DA) -like neurons using SH-SY5Y cell line; and assessed changes in epigenome, mitochondrion, and neuronal characteristics. Low dose GenX exposure at 0.4 and 4 μg/L prior to differentiation induces persistent changes in nuclear morphology and chromatin arrangements, manifested specifically in the facultative repressive marker H3K27me3. We also observed impaired neuronal network, increased calcium activity along with alterations in Tyrosine hydroxylase (TH) and α-Synuclein after prior exposure to GenX. Collectively, our results identified neurotoxicity of low-dose GenX exposure in human DA-like neurons following a developmental exposure scheme. The observed changes in neuronal characteristics suggest GenX as a potential neurotoxin and risk factor for Parkinson's disease.
Collapse
Affiliation(s)
- Shichen Wu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Han Zhao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Oscar Sanchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Xihui Zhao
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA.
| |
Collapse
|
8
|
Sun X, Xie Y, Zhang X, Song J, Wu Y. Estimation of Per- and Polyfluorinated Alkyl Substance Induction Equivalency Factors for Humpback Dolphins by Transactivation Potencies of Peroxisome Proliferator-Activated Receptors. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:3713-3721. [PMID: 36812292 DOI: 10.1021/acs.est.2c05044] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The potential risks of per- and polyfluoroalkyl substance (PFAS) accumulation in nearshore dolphins are not well understood. Here, transcriptional activities of 12 PFAS on peroxisome proliferator-activated receptors (PPAR-α, -β/δ, and -γ) in Indo-Pacific humpback dolphins (Sousa chinensis) were evaluated. All PFAS activated scPPAR-α in a dose-dependent manner. PFHpA exhibited the highest induction equivalency factors (IEFs). The order of IEFs for other PFAS was as follows: PFOA > PFNA > PFHxA > PFPeA > PFHxS > PFBA > PFOS > PFBuS ≈ PFDA ≫ PFUnDA and PFDoDA (not activated). The total induction equivalents (∑IEQs, 5537 ng/g wet weight) indicated that more attention should be paid to investigating contamination levels in dolphins, especially in PFOS (82.8% contribution to the ∑IEQs). The scPPAR-β/δ and -γ were not affected by any PFAS, except for PFOS, PFNA, and PFDA. Furthermore, PFNA and PFDA could induce higher PPAR-β/δ and PPAR-γ-mediated transcriptional activities than PFOA. Compared to human beings, PFAS might be more potent PPAR-α activators in humpback dolphins, suggesting that the dolphins may be more susceptible to the adverse effects of PFAS. Our results may be instructive for understanding the impacts of PFAS on marine mammal health due to the identical PPAR ligand-binding domain.
Collapse
Affiliation(s)
- Xian Sun
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-Sen University, Zhuhai 519082, China
| | - Yanqing Xie
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-Sen University, Zhuhai 519082, China
| | - Xiyang Zhang
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-Sen University, Zhuhai 519082, China
| | - Jiebing Song
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-Sen University, Zhuhai 519082, China
| | - Yuping Wu
- School of Marine Sciences, Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-Sen University, Zhuhai 519082, China
| |
Collapse
|
9
|
Qin W, Henneberger L, Huchthausen J, König M, Escher BI. Role of bioavailability and protein binding of four anionic perfluoroalkyl substances in cell-based bioassays for quantitative in vitro to in vivo extrapolations. ENVIRONMENT INTERNATIONAL 2023; 173:107857. [PMID: 36881956 DOI: 10.1016/j.envint.2023.107857] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 06/18/2023]
Abstract
Perfluoroalkyl substances (PFAS) are persistent and pose a risk to human health. High throughput screening (HTS) cell-based bioassays may inform risk assessment of PFAS provided that quantitative in vitro to in vivo extrapolation (QIVIVE) can be developed. The QIVIVE ratio is the ratio of nominal (Cnom) or freely dissolved concentration (Cfree) in human blood to Cnom or Cfree in the bioassays. Considering that the concentrations of PFAS in human plasma and in vitro bioassays may vary by orders of magnitude, we tested the hypothesis that anionic PFAS bind to proteins concentration-dependently and therefore the binding differs substantially between human plasma and bioassays, which has an impact on QIVIVE. Solid phase microextraction (SPME) with C18-coated fibers served to quantify the Cfree of four anionic PFAS (perfluorobutanoate (PFBA), perfluorooctanoate (PFOA), perfluorohexane sulfonate (PFHxS) and perfluorooctane sulfonate (PFOS)) in the presence of proteins and lipid, medium components, cells and human plasma over five orders of magnitude in concentrations. The C18-SPME method was used to quantify the non-linear binding to proteins, human plasma and medium, and the partition constants to cells. These binding parameters were used to predict Cfree of PFAS in cell bioassays and human plasma by a concentration-dependent mass balance model (MBM). The approach was illustrated with a reporter gene assay indicating activation of the peroxisome proliferator-activated receptor gamma (PPARγ-GeneBLAzer). Blood plasma levels were collected from literature for occupational exposure and the general population. The QIVIVEnom ratios were higher than the QIVIVEfree ratios due to the strong affinity to proteins and large differences in protein contents between human blood and bioassays. For human health risk assessment, the QIVIVEfree ratios of many in vitro assays need to be combined to cover all health relevant endpoints. If Cfree cannot be measured, they can be estimated with the MBM and concentration-dependent distribution ratios.
Collapse
Affiliation(s)
- Weiping Qin
- Department of Cell Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318 Leipzig, Germany; Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, Schnarrenbergstr, 94-96, DE-72076 Tübingen, Germany
| | - Luise Henneberger
- Department of Cell Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318 Leipzig, Germany
| | - Julia Huchthausen
- Department of Cell Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318 Leipzig, Germany; Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, Schnarrenbergstr, 94-96, DE-72076 Tübingen, Germany
| | - Maria König
- Department of Cell Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318 Leipzig, Germany
| | - Beate I Escher
- Department of Cell Toxicology, UFZ-Helmholtz Centre for Environmental Research, 04318 Leipzig, Germany; Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, Schnarrenbergstr, 94-96, DE-72076 Tübingen, Germany.
| |
Collapse
|
10
|
Abudayyak M, Karaman EF, Guler ZR, Ozden S. Effects of perfluorooctanoic acid on endoplasmic reticulum stress and lipid metabolism-related genes in human pancreatic cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104083. [PMID: 36804611 DOI: 10.1016/j.etap.2023.104083] [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: 02/25/2022] [Revised: 01/24/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Perfluorooctanoic acid (PFOA) is environmentally persistent and has been classified by The International Cancer Research Agency (IARC) as a possible human pancreatic carcinogen. In this study, the epigenetic alteration, the changes in the expression levels of endoplasmic reticulum stress-related and metabolism-related genes, as well as DNA methyltransferase expression were investigated using RT-PCR and ELISA assays. PFOA induced a significant increase in the methylation ratio (5-mC%), impacted DNA methylation maintenance gene expression and decreased lipid metabolism-related genes except for PPARγ (≥ 13-fold increase). While PFOA induced the expression of ATF4 (≥ 5.41-folds), CHOP (≥ 5.41-folds) genes, it inhibited the expression of ATF6 (≥ 67.2%), GRP78 (≥ 64.3%), Elf2α (≥ 95.8%), IRE1 (≥ 95.5%), and PERK (≥ 91.7%) genes. It is thought that epigenetic mechanisms together with disruption in the glucose-lipid metabolism and changes in endoplasmic reticulum stress-related genes may play a key role in PFOA-induced pancreatic toxicity.
Collapse
Affiliation(s)
- Mahmoud Abudayyak
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey
| | - Ecem Fatma Karaman
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Zeynep Rana Guler
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey
| | - Sibel Ozden
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
11
|
He X, Wu D, Xu Y, Zhang Y, Sun Y, Chang X, Zhu Y, Tang W. Perfluorooctanoic acid promotes pancreatic β cell dysfunction and apoptosis through ER stress and the ATF4/CHOP/TRIB3 pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:84532-84545. [PMID: 35788477 DOI: 10.1007/s11356-022-21188-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Perfluorooctanoic acid (PFOA), a widely used chemical substance, causes an increased risk of human type 2 diabetes (T2D), but its underlying mechanism is not well elucidated. The aim of the present study was to investigate whether PFOA regulates the functions of pancreatic β cells, which are specialized for the biosynthesis and secretion of insulin. The treatment of the mouse pancreatic β cell line (MIN6 cells) with PFOA caused a time- and dose-dependent inhibition of cell viability in CCK-8 assays. Annexin V/PI and TUNEL staining results confirmed that exposure to a high PFOA dose (500 μM) promoted apoptosis of β cells, while a low dose (300 μM) had no effects on β cell survival. PFOA treatment, even at a low dose, diminished glucose-stimulated insulin secretion (GSIS) in both primary islet perfusion and MIN6 cell experiments. RNA-sequencing data showed significantly increased expression of endoplasmic reticulum (ER) stress-associated genes, with tribbles homolog 3 (Trib3) ranking first among the altered genes. The activation of ER stress pathways was verified by qRT-PCR assays, and the ATF4/CHOP/TRIB3 pathway contributed to PFOA-induced β cell damage. The inhibition of TRIB3 expression significantly protected MIN6 cells from PFOA-induced GSIS defects and apoptosis by ameliorating ER stress. These findings reveal a link between ER stress and PFOA-induced β cell defects, opening up a new set of questions about the pathogenesis of T2D due to environmental chemicals.
Collapse
Affiliation(s)
- Xiaowei He
- Department of Endocrinology, Islet Cell Senescence and Function Research Laboratory, Nanjing Medical University Affiliated Geriatric Hospital/Jiangsu Province Geriatric Hospital, 30 Luojia Road, Nanjing, 210024, Jiangsu, China
| | - Dan Wu
- Department of Endocrinology, Islet Cell Senescence and Function Research Laboratory, Nanjing Medical University Affiliated Geriatric Hospital/Jiangsu Province Geriatric Hospital, 30 Luojia Road, Nanjing, 210024, Jiangsu, China
| | - Yanan Xu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Yuan, Nanjing, 210011, Jiangsu, China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Yue Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Wei Tang
- Department of Endocrinology, Islet Cell Senescence and Function Research Laboratory, Nanjing Medical University Affiliated Geriatric Hospital/Jiangsu Province Geriatric Hospital, 30 Luojia Road, Nanjing, 210024, Jiangsu, China.
| |
Collapse
|
12
|
Braun JM, Papandonatos GD, Li N, Sears CG, Buckley JP, Cecil KM, Chen A, Eaton CB, Kalkwarf HJ, Kelsey KT, Lanphear BP, Yolton K. Physical activity modifies the relation between gestational perfluorooctanoic acid exposure and adolescent cardiometabolic risk. ENVIRONMENTAL RESEARCH 2022; 214:114021. [PMID: 35952751 PMCID: PMC9637371 DOI: 10.1016/j.envres.2022.114021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 06/11/2023]
Abstract
OBJECTIVE Exposure to per- and polyfluoroalkyl substances (PFAS) - endocrine disrupting chemicals - may increase cardiometabolic risk. We evaluated whether adolescent lifestyle factors modified associations between gestational PFAS exposure and cardiometabolic risk using a prospective cohort study. METHODS In 166 mother-child pairs (HOME Study), we measured concentrations of four PFAS in maternal serum collected during pregnancy. When children were age 12 years, we calculated cardiometabolic risk scores from visceral adiposity area, blood pressure, and fasting serum biomarkers. We assessed adolescent physical activity and Healthy Eating Index scores using the Physical Activity Questionnaire for Older Children (PAQ-C), actigraphy, and 24-h diet recalls. Using multivariable linear regression and weighted quantile sum regression, we examined whether physical activity or diet modified covariate-adjusted associations of PFAS and their mixture with cardiometabolic risk scores. RESULTS Physical activity modified associations between perfluorooctanoic acid (PFOA) and cardiometabolic risk scores. Each doubling of PFOA was associated with worse cardiometabolic risk scores among children with PAQ-C scores < median (β:1.4; 95% CI:0.5, 2.2, n = 82), but not among those with PAQ-C scores ≥ median (β: 0.2; 95% CI: 1.2, 0.7, n = 84) (interaction p-value = 0.01). Associations were most prominent for insulin resistance, leptin-adiponectin ratio, and visceral fat area. We observed results suggesting that physical activity modified the association of PFAS mixture with cardiometabolic risk scores, insulin resistance, and visceral fat area (interaction p-values = 0.17, 0.07, and 0.10, respectively); however, the 95% CIs of the interaction terms included the null value. We observed similar, but attenuated patterns for PFOA and actigraphy-based measures of physical activity. Diet did not modify any associations. Physical activity or diet did not modify associations for other PFAS. CONCLUSIONS Childhood physical activity modified associations of prenatal serum PFOA concentrations with children's cardiometabolic risk in this cohort, indicating that lifestyle interventions may ameliorate the adverse effects of PFOA exposure.
Collapse
Affiliation(s)
- Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, 02912, Box G-S121, United States.
| | - George D Papandonatos
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI, United States
| | - Nan Li
- Department of Epidemiology, Brown University, Providence, RI, 02912, Box G-S121, United States
| | - Clara G Sears
- Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Jessie P Buckley
- Department of Environmental Health and Engineering, Johns Hopkins University School of Public Health, Baltimore, MD, United States
| | - Kim M Cecil
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Department of Radiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Charles B Eaton
- Department of Epidemiology, Brown University, Providence, RI, 02912, Box G-S121, United States; Department of Family Medicine, Warren Alpert Medical School of Brown University, Providence, RI, United States; Kent Memorial Hospital, Warwick, RI, United States
| | - Heidi J Kalkwarf
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, RI, 02912, Box G-S121, United States; Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Kimberly Yolton
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
13
|
Zhao H, Xie J, Wu S, Sánchez OF, Zhang X, Freeman JL, Yuan C. Pre-differentiation exposure of PFOA induced persistent changes in DNA methylation and mitochondrial morphology in human dopaminergic-like neurons. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 308:119684. [PMID: 35764183 DOI: 10.1016/j.envpol.2022.119684] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/10/2022] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
Perfluorooctanoic acid (PFOA) is abundant in environment due to its historical uses in consumer products and industrial applications. Exposure to low doses of PFOA has been associated with various disease risks, including neurological disorders. The underlying mechanism, however, remains poorly understood. In this study, we examined the effects of low dose PFOA exposure at 0.4 and 4 μg/L on the morphology, epigenome, mitochondrion, and neuronal markers of dopaminergic (DA)-like SH-SY5Y cells. We observed persistent decreases in H3K4me3, H3K27me3 and 5 mC markers in nucleus along with alterations in nuclear size and chromatin compaction percentage in DA-like neurons differentiated from SH-SY5Y cells exposed to 0.4 and 4 μg/L PFOA. Among the selected epigenetic features, DNA methylation pattern can be used to distinguish between PFOA-exposed and naïve populations, suggesting the involvement of epigenetic regulation. Moreover, DA-like neurons with pre-differentiation PFOA exposure exhibit altered network connectivity, mitochondrial volume, and TH expression, implying impairment in DA neuron functionality. Collectively, our results revealed the prolonged effects of developmental PFOA exposure on the fitness of DA-like neurons and identified epigenome and mitochondrion as potential targets for bearing long-lasting changes contributing to increased risks of neurological diseases later in life.
Collapse
Affiliation(s)
- Han Zhao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Shichen Wu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Oscar F Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Xinle Zhang
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA.
| |
Collapse
|
14
|
Kaiser AM, Zare Jeddi M, Uhl M, Jornod F, Fernandez MF, Audouze K. Characterization of Potential Adverse Outcome Pathways Related to Metabolic Outcomes and Exposure to Per- and Polyfluoroalkyl Substances Using Artificial Intelligence. TOXICS 2022; 10:toxics10080449. [PMID: 36006128 PMCID: PMC9412358 DOI: 10.3390/toxics10080449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 01/09/2023]
Abstract
Human exposure to per- and polyfluoroalkyl substances (PFAS) has been associated with numerous adverse health effects, depending on various factors such as the conditions of exposure (dose/concentration, duration, route of exposure, etc.) and characteristics associated with the exposed target (e.g., age, sex, ethnicity, health status, and genetic predisposition). The biological mechanisms by which PFAS might affect systems are largely unknown. To support the risk assessment process, AOP-helpFinder, a new artificial intelligence tool, was used to rapidly and systematically explore all available published information in the PubMed database. The aim was to identify existing associations between PFAS and metabolic health outcomes that may be relevant to support building adverse outcome pathways (AOPs). The collected information was manually organized to investigate linkages between PFAS exposures and metabolic health outcomes, including dyslipidemia, hypertension, insulin resistance, and obesity. Links between PFAS exposure and events from the existing metabolic-related AOPs were also retrieved. In conclusion, by analyzing dispersed information from the literature, we could identify some associations between PFAS exposure and components of existing AOPs. Additionally, we identified some linkages between PFAS exposure and metabolic outcomes for which only sparse information is available or which are not yet present in the AOP-wiki database that could be addressed in future research.
Collapse
Affiliation(s)
| | - Maryam Zare Jeddi
- National Institute for Public Health and Environment (RIVM), 3721 MA Bilthoven, The Netherlands
- Correspondence:
| | - Maria Uhl
- Environment Agency Austria, 1090 Vienna, Austria
| | - Florence Jornod
- Université Paris Cité, T3S, Inserm UMRS 1124, F-75006 Paris, France
| | - Mariana F. Fernandez
- Centre for Biomedical Research, E-18016 Granada, Spain
- Department of Radiology and Physical Medicine, School of Medicine, University of Granada, E-18071 Granada, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública, CIBERESP), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Karine Audouze
- Université Paris Cité, T3S, Inserm UMRS 1124, F-75006 Paris, France
| |
Collapse
|
15
|
Boyd RI, Ahmad S, Singh R, Fazal Z, Prins GS, Madak Erdogan Z, Irudayaraj J, Spinella MJ. Toward a Mechanistic Understanding of Poly- and Perfluoroalkylated Substances and Cancer. Cancers (Basel) 2022; 14:2919. [PMID: 35740585 PMCID: PMC9220899 DOI: 10.3390/cancers14122919] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 01/27/2023] Open
Abstract
Poly- and perfluoroalkylated substances (PFAS) are chemicals that persist and bioaccumulate in the environment and are found in nearly all human populations through several routes of exposure. Human occupational and community exposure to PFAS has been associated with several cancers, including cancers of the kidney, testis, prostate, and liver. While evidence suggests that PFAS are not directly mutagenic, many diverse mechanisms of carcinogenicity have been proposed. In this mini-review, we organize these mechanisms into three major proposed pathways of PFAS action-metabolism, endocrine disruption, and epigenetic perturbation-and discuss how these distinct but interdependent pathways may explain many of the proposed pro-carcinogenic effects of the PFAS class of environmental contaminants. Notably, each of the pathways is predicted to be highly sensitive to the dose and window of exposure which may, in part, explain the variable epidemiologic and experimental evidence linking PFAS and cancer. We highlight testicular and prostate cancer as models to validate this concept.
Collapse
Affiliation(s)
- Raya I. Boyd
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL 61802, USA; (R.I.B.); (R.S.); (Z.F.)
| | - Saeed Ahmad
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA; (S.A.); (J.I.)
| | - Ratnakar Singh
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL 61802, USA; (R.I.B.); (R.S.); (Z.F.)
| | - Zeeshan Fazal
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL 61802, USA; (R.I.B.); (R.S.); (Z.F.)
| | - Gail S. Prins
- Departments of Urology, Pathology and Physiology, College of Medicine, Chicago Center for Health and Environment, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Zeynep Madak Erdogan
- Department of Food Science and Human Nutrition, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA;
- Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute of Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Joseph Irudayaraj
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA; (S.A.); (J.I.)
- Department of Food Science and Human Nutrition, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA;
- Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute of Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Michael J. Spinella
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL 61802, USA; (R.I.B.); (R.S.); (Z.F.)
- Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
16
|
Heindel JJ, Howard S, Agay-Shay K, Arrebola JP, Audouze K, Babin PJ, Barouki R, Bansal A, Blanc E, Cave MC, Chatterjee S, Chevalier N, Choudhury M, Collier D, Connolly L, Coumoul X, Garruti G, Gilbertson M, Hoepner LA, Holloway AC, Howell G, Kassotis CD, Kay MK, Kim MJ, Lagadic-Gossmann D, Langouet S, Legrand A, Li Z, Le Mentec H, Lind L, Monica Lind P, Lustig RH, Martin-Chouly C, Munic Kos V, Podechard N, Roepke TA, Sargis RM, Starling A, Tomlinson CR, Touma C, Vondracek J, Vom Saal F, Blumberg B. Obesity II: Establishing causal links between chemical exposures and obesity. Biochem Pharmacol 2022; 199:115015. [PMID: 35395240 PMCID: PMC9124454 DOI: 10.1016/j.bcp.2022.115015] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a multifactorial disease with both genetic and environmental components. The prevailing view is that obesity results from an imbalance between energy intake and expenditure caused by overeating and insufficient exercise. We describe another environmental element that can alter the balance between energy intake and energy expenditure: obesogens. Obesogens are a subset of environmental chemicals that act as endocrine disruptors affecting metabolic endpoints. The obesogen hypothesis posits that exposure to endocrine disruptors and other chemicals can alter the development and function of the adipose tissue, liver, pancreas, gastrointestinal tract, and brain, thus changing the set point for control of metabolism. Obesogens can determine how much food is needed to maintain homeostasis and thereby increase the susceptibility to obesity. The most sensitive time for obesogen action is in utero and early childhood, in part via epigenetic programming that can be transmitted to future generations. This review explores the evidence supporting the obesogen hypothesis and highlights knowledge gaps that have prevented widespread acceptance as a contributor to the obesity pandemic. Critically, the obesogen hypothesis changes the narrative from curing obesity to preventing obesity.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA.
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, The Azrieli Faculty of Medicine, Bar Ilan University, Israel
| | - Juan P Arrebola
- Department of Preventive Medicine and Public Health University of Granada, Granada, Spain
| | - Karine Audouze
- Department of Systems Biology and Bioinformatics, University of Paris, INSERM, T3S, Paris France
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Etienne Blanc
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, USA
| | - Nicolas Chevalier
- Obstetrics and Gynecology, University of Cote d'Azur, Cote d'Azur, France
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisa Connolly
- The Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Xavier Coumoul
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Gabriella Garruti
- Department of Endocrinology, University of Bari "Aldo Moro," Bari, Italy
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Alison C Holloway
- McMaster University, Department of Obstetrics and Gynecology, Hamilton, Ontario, CA, USA
| | - George Howell
- Center for Environmental Health Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Mathew K Kay
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Sophie Langouet
- Univ Rennes, INSERM EHESP, IRSET UMR_5S 1085, 35000 Rennes, France
| | - Antoine Legrand
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Zhuorui Li
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Helene Le Mentec
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Occupational and Environmental Medicine, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California San Francisco, CA 94143, USA
| | | | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Normand Podechard
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Troy A Roepke
- Department of Animal Science, School of Environmental and Biological Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, Il 60612, USA
| | - Anne Starling
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Charbel Touma
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Frederick Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Völker J, Ashcroft F, Vedøy Å, Zimmermann L, Wagner M. Adipogenic Activity of Chemicals Used in Plastic Consumer Products. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022. [PMID: 35080176 DOI: 10.1101/2021.07.29.454199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Bisphenols and phthalates, chemicals frequently used in plastic products, promote obesity in cell and animal models. However, these well-known metabolism-disrupting chemicals (MDCs) represent only a minute fraction of all compounds found in plastics. To gain a comprehensive understanding of plastics as a source of exposure to MDCs, we characterized the chemicals present in 34 everyday products using nontarget high-resolution mass spectrometry and analyzed their joint adipogenic activities by high-content imaging. We detected 55,300 chemical features and tentatively identified 629 unique compounds, including 11 known MDCs. Importantly, the chemicals extracted from one-third of the products caused murine 3T3-L1 preadipocytes to proliferate, and differentiate into adipocytes, which were larger and contained more triglycerides than those treated with the reference compound rosiglitazone. Because the majority of plastic extracts did not activate the peroxisome proliferator-activated receptor γ and the glucocorticoid receptor, the adipogenic effects are mediated via other mechanisms and, thus, likely to be caused by unknown MDCs. Our study demonstrates that daily-use plastics contain potent mixtures of MDCs and can, therefore, be a relevant yet underestimated environmental factor contributing to obesity.
Collapse
Affiliation(s)
- Johannes Völker
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Felicity Ashcroft
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Åsa Vedøy
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Lisa Zimmermann
- Department of Aquatic Ecotoxicology, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Martin Wagner
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| |
Collapse
|
18
|
Aaseth J, Javorac D, Djordjevic AB, Bulat Z, Skalny AV, Zaitseva IP, Aschner M, Tinkov AA. The Role of Persistent Organic Pollutants in Obesity: A Review of Laboratory and Epidemiological Studies. TOXICS 2022; 10:65. [PMID: 35202251 PMCID: PMC8877532 DOI: 10.3390/toxics10020065] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 11/17/2022]
Abstract
Persistent organic pollutants (POPs) are considered as potential obesogens that may affect adipose tissue development and functioning, thus promoting obesity. However, various POPs may have different mechanisms of action. The objective of the present review is to discuss the key mechanisms linking exposure to POPs to adipose tissue dysfunction and obesity. Laboratory data clearly demonstrate that the mechanisms associated with the interference of exposure to POPs with obesity include: (a) dysregulation of adipogenesis regulators (PPARγ and C/EBPα); (b) affinity and binding to nuclear receptors; (c) epigenetic effects; and/or (d) proinflammatory activity. Although in vivo data are generally corroborative of the in vitro results, studies in living organisms have shown that the impact of POPs on adipogenesis is affected by biological factors such as sex, age, and period of exposure. Epidemiological data demonstrate a significant association between exposure to POPs and obesity and obesity-associated metabolic disturbances (e.g., type 2 diabetes mellitus and metabolic syndrome), although the existing data are considered insufficient. In conclusion, both laboratory and epidemiological data underline the significant role of POPs as environmental obesogens. However, further studies are required to better characterize both the mechanisms and the dose/concentration-response effects of exposure to POPs in the development of obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Jan Aaseth
- Research Department, Innlandet Hospital Trust, P.O. Box 104, 2381 Brumunddal, Norway
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, P.O. Box 400, 2418 Elverum, Norway
| | - Dragana Javorac
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia; (D.J.); (A.B.D.); (Z.B.)
| | - Aleksandra Buha Djordjevic
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia; (D.J.); (A.B.D.); (Z.B.)
| | - Zorica Bulat
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia; (D.J.); (A.B.D.); (Z.B.)
| | - Anatoly V. Skalny
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Department of Bioelementology, KG Razumovsky Moscow State University of Technologies and Management, 109004 Moscow, Russia
| | - Irina P. Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia;
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| |
Collapse
|
19
|
Epigenetic toxicity and cytotoxicity of perfluorooctanoic acid and its effects on gene expression in embryonic mouse hypothalamus cells. ACTA ACUST UNITED AC 2021; 72:182-190. [PMID: 34587672 PMCID: PMC8576751 DOI: 10.2478/aiht-2021-72-3555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 09/01/2021] [Indexed: 11/21/2022]
Abstract
Even though the endocrine-disrupting potential of perfluorooctanoic acid (PFOA) is well known, the mechanisms underlying its cellular and epigenetic toxicity at the critical stage of hypothalamic development are poorly understood. This is why we studied its effects on the embryonic mouse hypothalamic cell line N46 (mHypoE-N46) with a hope to shed more light on the mechanisms through which PFOA causes embryonic hypothalamic cell damage. To do that, we studied cell viability, global DNA methylation, and gene expression in cells exposed to PFOA. As the PFOA dose increased, cell viability decreased, while global DNA methylation increased. PFOA also significantly altered the expression of genes related to the apoptosis and cell cycle, neurotrophic genes, and the Tet, Dnmt, and Mecp2 genes. Our findings suggest that exposure to PFOA affects cell survival through the reprogramming of embryonic hypothalamic DNA methylation patterns and altering cell homeostasis genes. DNA methylation and changes in the Mecp2 gene expression induced by PFOA also imply wider ramifications, as they alter genes of other major mechanisms of the embryonic hypothalamus. Our study may therefore serve as a good starting point for further research into the mechanisms of PFOA effect of hypothalamic development.
Collapse
|
20
|
Kim S, Thapar I, Brooks BW. Epigenetic changes by per- and polyfluoroalkyl substances (PFAS). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 279:116929. [PMID: 33751946 DOI: 10.1016/j.envpol.2021.116929] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 05/09/2023]
Abstract
Increasing studies are examining per- and polyfluoroalkyl substances (PFAS) induced toxicity and resulting health outcomes, including epigenetic modifications (e.g., DNA methylation, histone modification, microRNA expression). We critically reviewed current evidence from human epidemiological, in vitro, and animal studies, including mammalian and aquatic model organisms. Epidemiological studies identified the associations between perfluorooctane sulfonate (PFOS) or perfluorooctanoic acid (PFOA) exposure and epigenetic changes in both adult populations and birth cohorts. For in vitro studies, various cell types including neuroblasts, preadipocytes, and hepatocytes have been employed to understand epigenetic effects of PFAS. In studies with animal models, effects of early life exposure to PFAS have been examined using rodent models, and aquatic models (e.g., zebrafish) have been more frequently used in recent years. Several studies highlighted oxidative stress as a key mediator between epigenetic modification and health effects. Collectively, previous research clearly suggest involvement of epigenetic mechanisms in PFAS induced toxicity, though these efforts have primarily focused on specific PFASs (i.e. mainly PFOS and PFOA) or endpoints (i.e. cancer). Additional studies are necessary to define specific linkages among epigenetic mechanisms and related biomarkers or phenotypical changes. In addition, future research is also needed for understudied PFAS and complex mixtures. Studies of epigenetic effects elicited by individual PFAS and mixtures are needed within an adverse outcome pathways framework, which will advance an understanding of PFAS risks to public health and the environment, and support efforts to design less hazardous chemicals.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Environmental Science, Baylor University, Waco, TX, 76706, USA; Institute of Biomedical Studies, Baylor University, Waco, TX, 76706, USA.
| | - Isha Thapar
- Department of Environmental Science, Baylor University, Waco, TX, 76706, USA; Honors College, Baylor University, Waco, TX, 76706, USA
| | - Bryan W Brooks
- Department of Environmental Science, Baylor University, Waco, TX, 76706, USA; Institute of Biomedical Studies, Baylor University, Waco, TX, 76706, USA.
| |
Collapse
|
21
|
Averina M, Brox J, Huber S, Furberg AS. Exposure to perfluoroalkyl substances (PFAS) and dyslipidemia, hypertension and obesity in adolescents. The Fit Futures study. ENVIRONMENTAL RESEARCH 2021; 195:110740. [PMID: 33460636 DOI: 10.1016/j.envres.2021.110740] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 05/22/2023]
Abstract
BACKGROUND Prevalence of obesity, hypertension and dyslipidemia has been increasing in children and adolescents worldwide. Exposure to environmental pollutants may contribute to this development. Our aim was to study associations between perfluoroalkyl substances (PFAS) and dyslipidemia, hypertension and obesity in a population-based sample of adolescents. METHODS Serum PFAS concentrations were measured in 940 adolescents, mean age 16.4 (SD 1.3) years, from the cross-sectional Fit Futures study by the UHPLC-MS/MS method. The following endpoints were used: hypertension (systolic blood pressure over 130 mmHg and/or diastolic blood pressure over 80 mmHg); obesity (body mass index over 2 z-score, WHO charts for adolescents); dyslipidemia (total cholesterol ≥ 5.17 mmol/L, and/or LDL-cholesterol ≥ 3.36 mmol/l, and/or apolipoprotein B ≥ 1.10 g/L). RESULTS Perfluorooctane sulfonate (PFOS), perfluorononanoate (PFNA), perfluorodecanoate (PFDA) and perfluoroundecanoate (PFUnDA) serum concentrations were positively associated with apolipoprotein B, total- and LDL cholesterol. The highest vs. lowest quartiles of total PFAS (∑PFAS), PFNA and PFDA concentrations were positively associated with the risk of dyslipidemia: OR 2.24 (95% CI 1.10-4.54), OR 2.30 (95% CI 1.16-4.57) and 2.36 (95% CI 1.08-5.16), respectively. The highest vs. lowest quartiles of ∑PFAS, perfluorohexane sulfonate (PFHxS), PFOS, perfluorooctanoate (PFOA) concentrations were positively associated with the risk of hypertension: OR 1.91 (95% CI 1.12-3.26), OR 2.06 (95% CI 1.16-3.65), 1.86 (95% CI 1.08-3.19) and 2.08 (95% CI 1.17-3.69) respectively. PFHxS and perfluoroheptane sulfonate (PFHpS) concentrations were positively associated with obesity. CONCLUSIONS This cross-sectional study showed a possible link between several PFAS and dyslipidemia, hypertension and obesity in Norwegian adolescents.
Collapse
Affiliation(s)
- Maria Averina
- Department of Laboratory Medicine, University Hospital of North Norway, 9038, Tromsø, Norway; Department of Community Medicine, UiT the Arctic University of Norway, Tromsø, Norway.
| | - Jan Brox
- Department of Laboratory Medicine, University Hospital of North Norway, 9038, Tromsø, Norway
| | - Sandra Huber
- Department of Laboratory Medicine, University Hospital of North Norway, 9038, Tromsø, Norway
| | - Anne-Sofie Furberg
- Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway; Faculty of Health and Social Sciences, Molde University College, Molde, Norway
| |
Collapse
|
22
|
Ramskov Tetzlaff CN, Ramhøj L, Lardenois A, Axelstad M, Evrard B, Chalmel F, Taxvig C, Svingen T. Adult female rats perinatally exposed to perfluorohexane sulfonate (PFHxS) and a mixture of endocrine disruptors display increased body/fat weights without a transcriptional footprint in fat cells. Toxicol Lett 2021; 339:78-87. [DOI: 10.1016/j.toxlet.2020.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022]
|
23
|
Bokobza E, Hinault C, Tiroille V, Clavel S, Bost F, Chevalier N. The Adipose Tissue at the Crosstalk Between EDCs and Cancer Development. Front Endocrinol (Lausanne) 2021; 12:691658. [PMID: 34354670 PMCID: PMC8329539 DOI: 10.3389/fendo.2021.691658] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2021] [Indexed: 12/02/2022] Open
Abstract
Obesity is a major public health concern at the origin of many pathologies, including cancers. Among them, the incidence of gastro-intestinal tract cancers is significantly increased, as well as the one of hormone-dependent cancers. The metabolic changes caused by overweight mainly with the development of adipose tissue (AT), insulin resistance and chronic inflammation induce hormonal and/or growth factor imbalances, which impact cell proliferation and differentiation. AT is now considered as the main internal source of endocrine disrupting chemicals (EDCs) representing a low level systemic chronic exposure. Some EDCs are non-metabolizable and can accumulate in AT for a long time. We are chronically exposed to low doses of EDCs able to interfere with the endocrine metabolism of the body. Importantly, several EDCs have been involved in the genesis of obesity affecting profoundly the physiology of AT. In parallel, EDCs have been implicated in the development of cancers, in particular hormone-dependent cancers (prostate, testis, breast, endometrium, thyroid). While it is now well established that AT secretes adipocytokines that promote tumor progression, it is less clear whether they can initiate cancer. Therefore, it is important to better understand the effects of EDCs, and to investigate the buffering effect of AT in the context of progression but also initiation of cancer cells using adequate models recommended to uncover and validate these mechanisms for humans. We will review and argument here the potential role of AT as a crosstalk between EDCs and hormone-dependent cancer development, and how to assess it.
Collapse
Affiliation(s)
- Emma Bokobza
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
| | - Charlotte Hinault
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
| | | | | | - Frédéric Bost
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
| | - Nicolas Chevalier
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- *Correspondence: Nicolas Chevalier, ;
| |
Collapse
|
24
|
Starling AP, Liu C, Shen G, Yang IV, Kechris K, Borengasser SJ, Boyle KE, Zhang W, Smith HA, Calafat AM, Hamman RF, Adgate JL, Dabelea D. Prenatal Exposure to Per- and Polyfluoroalkyl Substances, Umbilical Cord Blood DNA Methylation, and Cardio-Metabolic Indicators in Newborns: The Healthy Start Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:127014. [PMID: 33356526 PMCID: PMC7759236 DOI: 10.1289/ehp6888] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 05/02/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are environmentally persistent chemicals widely detected in women of reproductive age. Prenatal PFAS exposure is associated with adverse health outcomes in children. We hypothesized that DNA methylation changes may result from prenatal PFAS exposure and may be linked to offspring cardio-metabolic phenotype. OBJECTIVES We estimated associations of prenatal PFAS with DNA methylation in umbilical cord blood. We evaluated associations of methylation at selected sites with neonatal cardio-metabolic indicators. METHODS Among 583 mother-infant pairs in a prospective cohort, five PFAS were quantified in maternal serum (median 27 wk of gestation). Umbilical cord blood DNA methylation was evaluated using the Illumina HumanMethylation450 array. Differentially methylated positions (DMPs) were evaluated at a false discovery rate ( FDR ) < 0.05 and differentially methylated regions (DMRs) were identified using comb-p (Šidák-adjusted p < 0.05 ). We estimated associations between methylation at candidate DMPs and DMR sites and the following outcomes: newborn weight, adiposity, and cord blood glucose, insulin, lipids, and leptin. RESULTS Maternal serum PFAS concentrations were below the median for females in the U.S. general population. Moderate to high pairwise correlations were observed between PFAS concentrations (ρ = 0.28 - 0.76 ). Methylation at one DMP (cg18587484), annotated to the gene TJAP1, was associated with perfluorooctanoate (PFOA) at FDR < 0.05 . Comb-p detected between 4 and 15 DMRs for each PFAS. Associated genes, some common across multiple PFAS, were implicated in growth (RPTOR), lipid homeostasis (PON1, PON3, CIDEB, NR1H2), inflammation and immune activity (RASL11B, RNF39), among other functions. There was suggestive evidence that two PFAS-associated loci (cg09093485, cg09637273) were associated with cord blood triglycerides and birth weight, respectively (FDR < 0.1 ). DISCUSSION DNA methylation in umbilical cord blood was associated with maternal serum PFAS concentrations during pregnancy, suggesting potential associations with offspring growth, metabolism, and immune function. Future research should explore whether DNA methylation changes mediate associations between prenatal PFAS exposures and child health outcomes. https://doi.org/10.1289/EHP6888.
Collapse
Affiliation(s)
- Anne P. Starling
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cuining Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Guannan Shen
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Ivana V. Yang
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Sarah J. Borengasser
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristen E. Boyle
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Weiming Zhang
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Harry A. Smith
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Antonia M. Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Richard F. Hamman
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John L. Adgate
- Department of Environmental and Occupational Health, Colorado School of Public Health, Aurora, Colorado, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
25
|
Li CH, Shi YL, Li M, Guo LH, Cai YQ. Receptor-Bound Perfluoroalkyl Carboxylic Acids Dictate Their Activity on Human and Mouse Peroxisome Proliferator-Activated Receptor γ. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:9529-9536. [PMID: 32639727 DOI: 10.1021/acs.est.0c02386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
In in vitro cell assays, nominal concentrations of a test chemical are most frequently used in the description of its dose-response curves. Although the biologically effective concentration (BEC) is considered as the most relevant dose metric, in practice, it is very difficult to measure. In this work, we attempted to determine the BEC of long-chain perfluoroalkyl carboxylic acids (PFCAs) in peroxisome proliferator-activated receptor γ (PPARγ) activity assays. In both adipogenesis and transcriptional activity assays with human and mouse cells, PPARγ activity of 7 PFCAs first increased and then decreased with their carbon chain length. The binding affinity of these PFCAs with the ligand-binding domain of PPARγ was measured by fluorescence competitive binding assay and showed very poor correlation with their receptor activity (r2 = 0.002-0.047). Internal concentrations of the PFCAs in the cells were measured by LC-MS/MS. Although their correlation with the receptor activity increased significantly, it is still low (r2 = 0.41-0.82). Using the binding affinity constant, internal concentration, and PPARγ concentration measured by immunoassays, concentrations of receptor-bound PFCAs in cells were calculated, which exhibited excellent correlation with PPARγ activity in both adipogenesis and transcriptional activity assays (r2 = 0.91-0.93). These results demonstrate that the concentration of receptor-bound PFCA is the BEC that dictates its activity on human and mouse PPARγ in cell assays. In the absence of any direct detection method, our approach can be used to calculate the target-site concentration of other ligands.
Collapse
Affiliation(s)
- Chuan-Hai Li
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
- School of Public Health, Qingdao University, 308 Ningxia Street, Qingdao, Shandong 266071, China
| | - Ya-Li Shi
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
| | - Minjie Li
- College of Quality & Safety Engineering, China Jiliang University, 258 Xueyuan Street, Hangzhou, Zhejiang 310018, China
| | - Liang-Hong Guo
- Institute of Environmental and Health Sciences, China Jiliang University, 168 Xueyuan Street, Hangzhou, Zhejiang 310018, China
- Institute of Environmental and Health, Jianghan University, Wuhan, Hubei 430056, China
| | - Ya-Qi Cai
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
| |
Collapse
|
26
|
Zhang L, Duan X, Sun W, Sun H. Perfluorooctane sulfonate acute exposure stimulates insulin secretion via GPR40 pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 726:138498. [PMID: 32305757 DOI: 10.1016/j.scitotenv.2020.138498] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 05/20/2023]
Abstract
Perfluoroalkyl substances (PFASs) are widely used synthetic chemicals, showing environmental/biological persistence and adverse effects on ecosystem and human health. Several epidemiological and animal studies have revealed that PFASs levels are associated with elevated serum insulin level; however, the effect of PFASs on insulin secretion and the underlying mechanism are not clear. In this study, the effect of a most concerned PFAS, perfluorooctane sulfonate (PFOS) on insulin secretion in Beta-TC-6 pancreatic cells was studied. The results showed that PFOS acute exposure stimulated insulin secretion and elevated intracellular calcium concentration ([Ca2+]i). The PFOS-stimulated [Ca2+]i elevation was resulted from both extra- and intra-cellular sources. PFOS acute exposure decreased ATP content and ATP/ADP ratio, indicating the mitochondrial function was damaged under PFOS acute exposure. The PFOS-stimulated insulin secretion was inhibited by GW1100, a G Protein-coupled Receptor 40 (GPR40) specific inhibitor, but not affected by GW9662, a specific antagonist to the peroxisome proliferator-activated receptor gamma (PPARγ). The observation of RNA silencing further demonstrated that the PFOS-stimulated insulin secretion is, at least partially, via GPR40. By using specific inhibitors, we found that the GPR40 downstream pathways, phospholipase C (PLC) and L-type Ca2+ channels (LTCC) were involved in PFOS-stimulated [Ca2+]i elevation and insulin secretion.
Collapse
Affiliation(s)
- Lianying Zhang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China; School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Xiaoyu Duan
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Weijie Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Hongwen Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| |
Collapse
|
27
|
Liu W, Irudayaraj J. Perfluorooctanoic acid (PFOA) exposure inhibits DNA methyltransferase activities and alters constitutive heterochromatin organization. Food Chem Toxicol 2020; 141:111358. [PMID: 32315686 DOI: 10.1016/j.fct.2020.111358] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 03/13/2020] [Accepted: 04/12/2020] [Indexed: 12/20/2022]
Abstract
Perfluorooctanoic acid (PFOA) is a persistent and widespread industry-made chemical. Emerging evidence indicates that PFOA exposure could be meditated through DNA methylation, yet, the molecular mechanisms governing the epigenetic states have not been well established. In this study, we investigated the epigenetic alterations and inhibitory mechanisms upon PFOA exposure by identifying changes related to DNA methyltransferase (DNMT) with fluorescence correlation spectroscopy and stimulated emission depletion nanoscopy in human breast epithelial cells (MCF7). PFOA exposure at 100 and 200 μM altered the mobility of DNMT3A and inhibited the enzymatic activity of DNMT, resulting in global DNA demethylation. Moreover, PFOA significantly altered the heterochromatin organization, as noted by the distribution profile of histone 3 lysine 9 tri-methylation (H3K9me3) at 200 and 400 μM exposure levels with super-resolution microscopy. An increased redistribution around the periphery of the nucleus was noted with a more diffused distribution beyond the 200 μM exposure. Overall, exposure of PFOA resulted in DNA demethylation accompanied by altered expression patterns of DNMT1 and DNMT3A. These findings provided new insights on the epigenetic alterations and revealed an altered heterochromatin packaging upon exposure to PFOA, implicating a mechanistic mode of action of DNA demethylation through direct impacts on DNMTs and increasing susceptibility to diseases such as cancer.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Biomedical Research Center in Mills Breast Cancer Institute, Carles Foundation Hospital, Urbana, IL, 61801, USA
| | - Joseph Irudayaraj
- Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Biomedical Research Center in Mills Breast Cancer Institute, Carles Foundation Hospital, Urbana, IL, 61801, USA.
| |
Collapse
|
28
|
Sun W, Duan X, Chen H, Zhang L, Sun H. Adipogenic activity of 2-ethylhexyl diphenyl phosphate via peroxisome proliferator-activated receptor γ pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 711:134810. [PMID: 31812418 DOI: 10.1016/j.scitotenv.2019.134810] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 06/10/2023]
Abstract
Recent studies have shown that exposure to some organophosphates, such as triphenyl phosphate (TPHP) and diphenyl phosphate (DPHP), can affect adipogenesis in preadipocytes. 2-Ethylhexyl diphenyl phosphate (EHDPP), an organophosphate, is frequently detected in various environmental media. However, there is less information about the toxicity effects and the mechanism by which EHDPP affects preadipocytes. In the present study, we investigated whether EHDPP could induce differentiation in 3T3-L1 preadipocytes through the peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway. The fluorescence competitive binding assay and the dual-luciferase reporter gene assay were used to assess the binding affinity and activation of PPARγ, and the results showed that EHDPP can bind to the ligand binding domain of PPARγ (PPARγ-LBD) and activate PPARγ in vitro. Exposure to EHDPP for 10 days extensively induced adipogenesis in 3T3-L1 preadipocytes as assessed by lipid accumulation and gene expression of adipogenic markers of fatty acid binding protein 4 (FABP4), lipoprotein lipase (Lpl), adiponectin (Adip), and fatty acid synthase (Fasn). Furthermore, the preadipocytes differentiation was blocked by the PPARγ-specific antagonist GW9662, indicating that the PPARγ signaling pathway plays an important part in 3T3-L1 cell differentiation induced by EHDPP. Taken together, EHDPP can bind to PPARγ-LBD, activate PPARγ receptor, and induce cell differentiation via the PPARγ signaling pathway in 3T3-L1 preadipocytes.
Collapse
Affiliation(s)
- Weijie Sun
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Xiaoyu Duan
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Hao Chen
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Lianying Zhang
- School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China.
| | - Hongwen Sun
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| |
Collapse
|
29
|
Temkin AM, Hocevar BA, Andrews DQ, Naidenko OV, Kamendulis LM. Application of the Key Characteristics of Carcinogens to Per and Polyfluoroalkyl Substances. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E1668. [PMID: 32143379 PMCID: PMC7084585 DOI: 10.3390/ijerph17051668] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/09/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) constitute a large class of environmentally persistent chemicals used in industrial and consumer products. Human exposure to PFAS is extensive, and PFAS contamination has been reported in drinking water and food supplies as well as in the serum of nearly all people. The most well-studied member of the PFAS class, perfluorooctanoic acid (PFOA), induces tumors in animal bioassays and has been associated with elevated risk of cancer in human populations. GenX, one of the PFOA replacement chemicals, induces tumors in animal bioassays as well. Using the Key Characteristics of Carcinogens framework for cancer hazard identification, we considered the existing epidemiological, toxicological and mechanistic data for 26 different PFAS. We found strong evidence that multiple PFAS induce oxidative stress, are immunosuppressive, and modulate receptor-mediated effects. We also found suggestive evidence indicating that some PFAS can induce epigenetic alterations and influence cell proliferation. Experimental data indicate that PFAS are not genotoxic and generally do not undergo metabolic activation. Data are currently insufficient to assess whether any PFAS promote chronic inflammation, cellular immortalization or alter DNA repair. While more research is needed to address data gaps, evidence exists that several PFAS exhibit one or more of the key characteristics of carcinogens.
Collapse
Affiliation(s)
- Alexis M. Temkin
- Environmental Working Group, Washington, DC 20009, USA; (D.Q.A.); (O.V.N.)
| | - Barbara A. Hocevar
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA; (B.A.H.); (L.M.K.)
| | - David Q. Andrews
- Environmental Working Group, Washington, DC 20009, USA; (D.Q.A.); (O.V.N.)
| | - Olga V. Naidenko
- Environmental Working Group, Washington, DC 20009, USA; (D.Q.A.); (O.V.N.)
| | - Lisa M. Kamendulis
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA; (B.A.H.); (L.M.K.)
| |
Collapse
|
30
|
Méndez-Tepepa M, Zepeda-Pérez D, Espindola-Lozano M, Rodríguez-Castelán J, Arroyo-Helguera O, Pacheco P, Nicolás-Toledo L, Cuevas-Romero E. Hypothyroidism modifies differentially the content of lipids and glycogen, lipid receptors, and intraepithelial lymphocytes among oviductal regions of rabbits. Reprod Biol 2020; 20:247-253. [PMID: 32089504 DOI: 10.1016/j.repbio.2020.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 10/25/2022]
Abstract
Hypothyroidism affects the content of triacylglycerol (TAG), total cholesterol (TC), oxidized lipids, glycogen, and infiltration of immune cells into the ovary and uterus. This study aimed to analyze the impact of hypothyroidism on the lipid content of different regions of the oviduct. Control (n = 6) and hypothyroid (n = 6; 10 mg/kg/day of methimazole in the drinking water for 30 days) adult rabbits were used. In the fimbriae/infundibulum (FIM/INF), ampulla, (AMP), isthmus (IST), and utero-tubal junction (UTJ), the TAG and TC concentrations, presence of oxidized lipid, relative expressions of perilipin A (PLIN A), peroxisome proliferator-activated receptor γ (PPARγ), CAAT/enhancer-binding protein α (C/EBPα), and farnesoid X receptor (FXRα) were analyzed. The content of glycogen and glycans, as well as the infiltration of lymphocytes, were also quantified. In the FIM/INF, hypothyroidism reduced the content of TC, expression of C/EBPα, and presence of glycans while increased the number of intraepithelial lymphocytes. In the AMP and IST-UTJ regions, hypothyroidism increased the content of TAG, oxidized lipids, expression of PPARγ, and glycogen content but decreased the expression of PLIN-A. The FXRα expression in secretory cells of IST-UTJ was higher in the hypothyroid rabbits compared to controls. Additionally, hypothyroidism reduced the C/EBPα expression and the number of intraepithelial lymphocytes in the AMP and IST-UTJ regions, respectively. We demonstrated that the effect of hypothyroidism depends on the oviductal region, possibly associated with different physiological functions specific to each region. These alterations may be related to infertility, tubal disturbances, and ectopic pregnancy observed in hypothyroid women.
Collapse
Affiliation(s)
- Maribel Méndez-Tepepa
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Dafne Zepeda-Pérez
- Maestría en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | - Julia Rodríguez-Castelán
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico; Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - Pablo Pacheco
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Leticia Nicolás-Toledo
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Estela Cuevas-Romero
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico.
| |
Collapse
|
31
|
Starling AP, Adgate JL, Hamman RF, Kechris K, Calafat AM, Dabelea D. Prenatal exposure to per- and polyfluoroalkyl substances and infant growth and adiposity: the Healthy Start Study. ENVIRONMENT INTERNATIONAL 2019; 131:104983. [PMID: 31284113 PMCID: PMC6728170 DOI: 10.1016/j.envint.2019.104983] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 05/20/2023]
Abstract
BACKGROUND Prenatal exposures to certain per- and polyfluoroalkyl substances (PFAS) have been linked to lower weight and adiposity at birth but greater weight and adiposity in childhood. We hypothesized that faster growth in early infancy may be associated with maternal PFAS concentrations. METHODS Among 415 mother-infant pairs in a longitudinal cohort study, we estimated associations between maternal pregnancy serum concentrations of six PFAS and offspring weight and adiposity at ~5 months of age, and growth in early infancy. Linear and logistic regression models were adjusted for potential confounders including maternal pre-pregnancy body mass index. Effect modification by infant sex was evaluated. We evaluated potential confounding by correlated exposures via multipollutant linear regression and elastic net penalized regression. RESULTS Associations between maternal PFAS concentrations and infant weight and adiposity differed by offspring sex. In male infants, maternal perfluorooctanoate and perfluorononanoate were positively associated with adiposity, with percent fat mass increases of 1.5-1.7% per ln-ng/mL increase in PFAS (median adiposity at ~5 months: 24.6%). Maternal perfluorooctane sulfonate (PFOS) and perfluorohexane sulfonate (PFHxS) were associated with lower weight-for-age z-score among female infants only (-0.26 SD per ln-ng/mL PFOS, 95% CI -0.43, -0.10; -0.17 SD per ln-ng/mL PFHxS, 95% CI -0.33, -0.01). In analyses pooled by sex, 2-(N-methyl-perfluorooctane sulfonamido) acetate above vs. below the limit of detection was associated with greater odds of rapid growth in weight-for-age (odds ratio [OR] 2.2, 95% CI 1.1, 4.3) and weight-for-length (OR 3.3, 95% CI 1.8, 6.2). Multipollutant models generally confirmed the results and strengthened some associations. DISCUSSION We observed sex- and chemical-specific associations between maternal serum PFAS concentrations and infant weight and adiposity. Multipollutant models suggested confounding by correlated PFAS with opposing effects. Although maternal PFAS concentrations are inversely associated with infant weight and adiposity at birth, rapid gain may occur in infancy, particularly in fat mass.
Collapse
Affiliation(s)
- Anne P Starling
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA; Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - John L Adgate
- Department of Environmental and Occupational Health, Colorado School of Public Health, Aurora, CO, USA
| | - Richard F Hamman
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA; Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA; Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
32
|
Lee MK, Blumberg B. Transgenerational effects of obesogens. Basic Clin Pharmacol Toxicol 2019; 125 Suppl 3:44-57. [PMID: 30801972 PMCID: PMC6708505 DOI: 10.1111/bcpt.13214] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/08/2019] [Indexed: 02/06/2023]
Abstract
Obesity and associated disorders are now a global pandemic. The prevailing clinical model for obesity is overconsumption of calorie-dense food and diminished physical activity (the calories in-calories out model). However, this explanation does not account for numerous recent research findings demonstrating that a variety of environmental factors can be superimposed on diet and exercise to influence the development of obesity. The environmental obesogen model proposes that exposure to chemical obesogens during in utero and/or early life can strongly influence later predisposition to obesity. Obesogens are chemicals that inappropriately stimulate adipogenesis and fat storage, in vivo either directly or indirectly. Numerous obesogens have been identified in recent years and some of these elicit transgenerational effects on obesity as well as a variety of health end-points after exposure of pregnant F0 females. Prenatal exposure to environmental obesogens can produce lasting effects on the exposed animals and their offspring to at least the F4 generation. Recent results show that some of these transgenerational effects of obesogen exposure can be carried across the generations via alterations in chromatin structure and accessibility. That some chemicals can have permanent effects on the offspring of exposed animals suggests increased caution in the debate about whether and to what extent exposure to endocrine-disrupting chemicals and obesogens should be regulated.
Collapse
Affiliation(s)
- Michelle Kira Lee
- Department of Developmental and Cell Biology, 2011 BioSci
3, University of California, Irvine, CA 926970-2300
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, 2011 BioSci
3, University of California, Irvine, CA 926970-2300
- Department of Pharmaceutical Sciences, University of
California, Irvine
- Dept of Biomedical Engineering, University of California,
Irvine
| |
Collapse
|
33
|
Wen Q, Xie X, Zhao C, Ren Q, Zhang X, Wei D, Emanuelli B, Du Y. The brominated flame retardant PBDE 99 promotes adipogenesis via regulating mitotic clonal expansion and PPARγ expression. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 670:67-77. [PMID: 30903904 DOI: 10.1016/j.scitotenv.2019.03.201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/09/2019] [Accepted: 03/14/2019] [Indexed: 06/09/2023]
Abstract
"Obesogens" have been widely accepted as chemicals that promote obesity, and there are many environmental pollutants that were functionally identified as obesogens. PBDE 99 is one of the most abundant PBDE congeners detected in human. However, its obesogenic effects are poorly understood. Here, we explore the in vitro effects of PBDE 99 on adipogenesis, which is a key process in obesogenesis. We observed an increase in adipogenesis when differentiating cells were exposed to PBDE 99. Further, the promoting effects of PBDE 99 on adipogenesis were most efficient during the first 4 days of 3T3-L1 differentiation. Consistent with this, early transcriptional factor CCAAT/enhancer-binding proteins β (C/EBPβ) was upregulated at Days 1 and 2 during differentiation, which is accompanied with the acceleration of mitotic clonal expansion (MCE) and the upregulation of terminal transcriptional factors C/EBPα and PPARγ2 from Day 2 or Day 4. Additionally, bisulfite genomic sequencing analysis revealed that PBDE 99 decreased methylation status of the CpG sites at PPARγ promoter region. Collectively, these findings demonstrate that PBDE 99 may be a potential environmental obesogen by promoting adipogenesis through facilitating MCE progression at early differentiation stage and upregulating key adipogenic factor PPARγ2 expression both in direct transcriptional and epigenetic regulation dependent manner.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinni Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Chuanfang Zhao
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qidong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyi Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongbin Wei
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yuguo Du
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
34
|
Li CH, Ren XM, Guo LH. Adipogenic Activity of Oligomeric Hexafluoropropylene Oxide (Perfluorooctanoic Acid Alternative) through Peroxisome Proliferator-Activated Receptor γ Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:3287-3295. [PMID: 30785727 DOI: 10.1021/acs.est.8b06978] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Hexafluoropropylene oxide trimer acid (HFPO-TA) and hexafluoropropylene oxide dimer acid (HFPO-DA) have been used as perfluorooctanoic acid (PFOA) alternatives in the fluoropolymer industry for years. Their widespread environmental distribution, high bioaccumulation capability, and human exposure have caused great concern. Nevertheless, their potential toxicity and health risk remain largely unknown. In the present study, we compared potential disruption effects of HFPO-TA, HFPO-DA, and PFOA on peroxisome proliferator-activated receptor γ (PPARγ) via the investigation of receptor binding, receptor activity, and cell adipogenesis effects. The receptor binding experiment showed HFPO-TA exhibited 4.8-7.5 folds higher binding affinity with PPARγ than PFOA, whereas HFPO-DA exhibited weaker binding affinity than PFOA. They also showed agonistic activity toward PPARγ signaling pathway in HEK 293 cells in the order of HFPO-TA > PFOA > HFPO-DA. Molecular docking simulation indicated HFPO-TA formed more hydrogen bonds than PFOA, whereas HFPO-DA formed fewer hydrogen bonds than PFOA. HFPO-TA promoted adipogenic differentiation and lipid accumulation in both mouse and human preadipocytes with potency higher than PFOA. Adipogenesis in human preadipocytes is a more sensitive end point than mouse preadipocytes. Collectively, HFPO-TA exerts higher binding affinity, agonistic activity, and adipogenesis activity than PFOA. The potential health risk of HFPO-TA should be of concern.
Collapse
Affiliation(s)
- Chuan-Hai Li
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences , Chinese Academy of Sciences , 18 Shuangqing Road , Beijing 100085 , P. R. China
- College of Resources and Environment , University of Chinese Academy of Sciences , Beijing 100039 , P. R. China
| | - Xiao-Min Ren
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences , Chinese Academy of Sciences , 18 Shuangqing Road , Beijing 100085 , P. R. China
| | - Liang-Hong Guo
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences , Chinese Academy of Sciences , 18 Shuangqing Road , Beijing 100085 , P. R. China
- College of Resources and Environment , University of Chinese Academy of Sciences , Beijing 100039 , P. R. China
- The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou 510150 , P. R. China
| |
Collapse
|
35
|
Liu S, Yang R, Yin N, Wang YL, Faiola F. Environmental and human relevant PFOS and PFOA doses alter human mesenchymal stem cell self-renewal, adipogenesis and osteogenesis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:564-572. [PMID: 30476818 DOI: 10.1016/j.ecoenv.2018.11.064] [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: 08/28/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 05/21/2023]
Abstract
PFOS and PFOA are two of the most abundant perfluorinated compounds (PFCs) in the environment. Previous studies have reported they have a long half-life (up to five years) once they enter into the human body. Moreover, they can potentially promote the adipogenic process by activating PPARγ. However, little is known about PFOS and PFOA chronic health impacts on humans. In this study, we employed primary human mesenchymal stem cells (hMSCs) and demonstrated that PFOS and PFOA exerted acute cytotoxicity and affected adipogenesis and osteogenesis at environmental and human relevant doses. In fact, PFOS and PFOA impaired the proper expression of CD90 (a surface antigen highly enriched in undifferentiated hMSCs) and promoted adipogenesis, presumably via their interaction with PPARγ. Moreover, PFOA partly disturbed osteogenesis. Thus, our findings not only validated the health risks of PFOS and PFOA, but also revealed new potential long-term PFOS/PFOA impacts on humans.
Collapse
Affiliation(s)
- Shuyu Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuan-Liang Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China; Section of Molecular Biology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|