1
|
Nielsen G, Gondim DD, Cave MC, Heiger-Bernays WJ, Webster TF, Schlezinger JJ. Perfluorooctanoic acid increases serum cholesterol in a PPARα-dependent manner in female mice. Arch Toxicol 2025; 99:2087-2105. [PMID: 40021516 DOI: 10.1007/s00204-025-03984-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a large group of persistent chemicals that are pervasive in the environment leading to widespread exposure for humans. Perfluorooctanoic acid (PFOA), one of the most commonly measured PFAS in people, disrupts liver and serum lipid homeostasis as shown in animal toxicity and human epidemiological studies. We tested the hypothesis that the effects of PFOA exposure in mice expressing mouse PPARα (mPPARα) are driven largely through PPARα-dependent mechanisms while non-PPARα dependent mechanisms will be more apparent in mice expressing human PPARα (hPPARα). Female and male mPPARα, hPPARα, and PPARα null mice were exposed to PFOA (0.5, 1.4 or 6.2 mg PFOA/L) via drinking water for 14 weeks. Concurrently, mice consumed an American diet containing human diet-relevant amounts of fat and cholesterol. Here, we focused on the effects in female mice, given the dearth of data reported on PFAS-induced effects in females. Increasing the duration of PFOA exposure reduced weight gain in all genotypes of female mice while end-of-study body fat was lower in PFOA exposed hPPARα and PPARα null mice. Serum cholesterol, but not triacylglyceride, concentrations were increased by PFOA exposure in a PPARα-dependent manner. Hepatic triacylglycerides were higher in vehicle-exposed mPPARα and PPARα null mice than hPPARα mice, and PFOA significantly reduced concentrations in mPPARα and PPARα null mice only. In contrast, PFOA increased hepatic cholesterol content in a PPARα-dependent manner. Changes in liver and serum cholesterol may be explained by a strong, PPARα-dependent downregulation of Cyp7a1 expression. PFOA significantly increased PPARα target gene expression in mPPARα mice. Other nuclear receptors were examined: CAR target gene expression was only induced by PFOA in hPPARα and PPARα null mice. PXR target gene expression was induced by PFOA in all genotypes. Results were similar in male mice with two exceptions: (1) vehicle-exposed male mice of all genotypes were equally susceptible to diet-induced hepatic steatosis; (2) male mice drank less water, resulting in lower serum PFOA levels, which may explain the less significant changes in lipid endpoints. Overall, our results show that PFOA modifies triacylglyceride and cholesterol homeostasis independently and that PPARα plays an important role in PFOA-induced increases in liver and serum cholesterol.
Collapse
Affiliation(s)
- G Nielsen
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA
| | - D D Gondim
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY, USA
| | - M C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY, USA
| | - W J Heiger-Bernays
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA
| | - T F Webster
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA
| | - J J Schlezinger
- Department of Environmental Health, School of Public Heath, Boston University, Boston, MA, USA.
| |
Collapse
|
2
|
Shin MW, Kang H, Kim SH. Concentrations of Serum Per- and Polyfluoroalkyl Substances and Lipid Health in Adolescents: A Cross-Sectional Study from the Korean National Environmental Health Survey 2018-2020. TOXICS 2025; 13:91. [PMID: 39997906 PMCID: PMC11860244 DOI: 10.3390/toxics13020091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/26/2025]
Abstract
Emerging evidence indicates that environmental exposure to per- and polyfluoroalkyl substances (PFASs) may influence lipid metabolism, though studies on adolescents remain scarce. This study aimed to investigate the association between PFAS mixture exposure and lipid profiles in Korean adolescents. Using data from the Korean National Environmental Health Survey (2018-2020), we analyzed 824 adolescents aged 12-17 years. Serum concentrations of PFAS, including perfluorooctanoic acid (PFOA), perfluorooctane sulfonic acid (PFOS), perfluorononanoic acid (PFNA), perfluorohexane sulfonic acid (PFHxS), and perfluorodecanoic acid (PFDeA), and lipid profiles were assessed. In multivariate regression models, PFDeA and PFNA were positively associated with elevated total cholesterol and low-density lipoprotein cholesterol levels, and PFDeA was associated with hypercholesterolemia risk in boys. In girls, PFDeA was associated with higher high-density lipoprotein cholesterol and lower triglycerides, though no significant association with hypercholesterolemia risk was observed. Bayesian kernel machine regression demonstrated positive associations between PFAS mixture exposure and hypercholesterolemia risk in boys but not in girls. The quantile g-computation model also demonstrated an odds ratio (OR) of 1.47 (95% CI: 0.99-2.19, p = 0.057) for PFAS mixture exposure in boys, suggesting borderline statistical significance. These findings suggest that PFAS exposure may disrupt lipid metabolism, elevating hypercholesterolemia risk in adolescents, particularly boys.
Collapse
Affiliation(s)
- Min-Won Shin
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul 01757, Republic of Korea;
| | - Habyeong Kang
- Department of Preventive Medicine, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea;
| | - Shin-Hye Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul 01757, Republic of Korea;
| |
Collapse
|
3
|
Ohguro H, Nishikiori N, Sato T, Watanabe M, Higashide M, Furuhashi M. Pemafibrate Induces a Low Level of PPARα Agonist-Stimulated mRNA Expression of ANGPTL4 in ARPE19 Cell. Bioengineering (Basel) 2024; 11:1247. [PMID: 39768065 PMCID: PMC11673482 DOI: 10.3390/bioengineering11121247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
To elucidate the unidentified roles of a selective peroxisome proliferator-activated receptor α (PPARα) agonist, pemafibrate (Pema), on the pathogenesis of retinal ischemic diseases (RID)s, the pharmacological effects of Pema on the retinal pigment epithelium (RPE), which is involved in the pathogenesis of RID, were compared with the pharmacological effects of the non-fibrate PPARα agonist GW7647 (GW). For this purpose, the human RPE cell line ARPE19 that was untreated (NT) or treated with Pema or GW was subjected to Seahorse cellular metabolic analysis and RNA sequencing analysis. Real-time cellular metabolic function analysis revealed that pharmacological effects of the PPARα agonist actions on essential metabolic functions in RPE cells were substantially different between Pema-treated cells and GW-treated cells. RNA sequencing analysis revealed the following differentially expressed genes (DEGs): (1) NT vs. Pema-treated cells, 37 substantially upregulated and 72 substantially downregulated DEGs; (2) NT vs. GW-treated cells, 32 substantially upregulated and 54 substantially downregulated DEGs; and (3) Pema vs. GW, 67 substantially upregulated and 51 markedly downregulated DEGs. Gene ontology (GO) analysis and ingenuity pathway analysis (IPA) showed several overlaps or differences in biological functions and pathways estimated by the DEGs between NT and Pema-treated cells and between NT and GW-treated cells, presumably due to common PPARα agonist actions or unspecific off-target effects to each. For further estimation, overlaps of DEGs among different pairs of comparisons (NT vs. Pema, NT vs. GW, and Pema vs. GW) were listed up. Angiopoietin-like 4 (ANGPTL4), which has been shown to cause deterioration of RID, was the only DEG identified as a common significantly upregulated DEG in all three pairs of comparisons, suggesting that ANGPTL4 was upregulated by the PPARα agonist action but that its levels were substantially lower in Pema-treated cells than in GW-treated cells. In qPCR analysis, such lower efficacy for upregulation of the mRNA expression of ANGPTL4 by Pema than by GW was confirmed, in addition to substantial upregulation of the mRNA expression of HIF1α by both agonists. However, different Pema and GW-induced effects on mRNA expression of HIF1α (Pema, no change; GW, significantly downregulated) and mRNA expression of ANGPTL4 (Pema, significantly upregulated; GW, significantly downregulated) were observed in HepG2 cells, a human hepatocyte cell line. The results of this study suggest that actions of the PPARα agonists Pema and GW are significantly organ-specific and that lower upregulation of mRNA expression of the DR-worsening factor ANGPTL4 by Pema than by GW in ARPE19 cells may minimize the risk for development of RID.
Collapse
Affiliation(s)
- Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Nami Nishikiori
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan;
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan;
| |
Collapse
|
4
|
Karakuş F, Kuzu B. Predicting the molecular mechanisms of cardiovascular toxicity induced by per- and polyfluoroalkyl substances: an In Silico network toxicology perspective. Toxicol Res (Camb) 2024; 13:tfae206. [PMID: 39677493 PMCID: PMC11645662 DOI: 10.1093/toxres/tfae206] [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: 04/16/2024] [Revised: 09/11/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are human-made chemicals that accumulate in the human body and the environment over time. Humans are primarily exposed to PFAS through drinking water, food, consumer products, and dust. These exposures can have many adverse health effects, including cardiovascular diseases (CVDs) and factors contributing to CVDs. This study identified the molecular mechanisms of CVDs caused by PFAS. METHODS For this purpose, various computational tools, such as the Comparative Toxicogenomic Database, ShinyGO, ChEA3, MIENTURNET, GeneMANIA, STRING, and Cytoscape, were used to conduct in silico analyses. RESULTS The results showed that 10 genes were common between PFAS and CVDs, and among these common genes, the PPAR signaling pathway, fatty acid metabolic processes, and lipid binding were the most significantly associated gene ontology terms. Among the top 10 transcription factors (TFs) related to these common genes, peroxisome proliferator-activated receptor gamma and androgen receptor were the most prominent. Additionally, hsa-miR-130b-3p, hsa-miR-130a-3p, and hsa-miR-129-5p were featured microRNAs involved in PFAS-induced CVDs. Finally, PPARA and PPARG were identified as core genes involved in PFAS-induced CVDs. CONCLUSION These findings may contribute to a better understanding of the molecular mechanisms and reveal new potential targets in PFAS-induced CVDs.
Collapse
Affiliation(s)
- Fuat Karakuş
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Van Yuzuncu Yil University, 65080 Tuşba-Van, Türkiye
| | - Burak Kuzu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Van Yuzuncu Yil University, 65080 Tuşba-Van, Türkiye
| |
Collapse
|
5
|
Maerten A, Callewaert E, Sanz-Serrano J, Devisscher L, Vinken M. Effects of per- and polyfluoroalkyl substances on the liver: Human-relevant mechanisms of toxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176717. [PMID: 39383969 DOI: 10.1016/j.scitotenv.2024.176717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are abundantly used in a plethora of products with applications in daily life. As a result, PFAS are widely distributed in the environment, thus providing a source of exposure to humans. The majority of human exposure to PFAS is attributed to the human diet, which encompasses drinking water. Their chemical nature grants persistent, accumulative and toxic properties, which are currently raising concerns. Over the past few years, adverse effects of PFAS on different organs have been repeatedly documented. Numerous epidemiological studies established a clear link between PFAS exposure and liver toxicity. Likewise, effects of PFAS on liver homeostasis, lipid metabolism, bile acid metabolism and hepatocarcinogenesis have been reported in various in vitro and in vivo studies. This review discusses the role of PFAS in liver toxicity with special attention paid to human relevance as well as to the mechanisms underlying the hepatotoxic effects of PFAS. Future perspectives and remaining knowledge gaps were identified to enhance future PFAS risk assessment.
Collapse
Affiliation(s)
- Amy Maerten
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ellen Callewaert
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Julen Sanz-Serrano
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Sciences, Universiteit Gent, Gent, Belgium; Liver Research Center Ghent, Universiteit Gent, University Hospital Ghent, Gent, Belgium
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
6
|
Mahoney H, Ankley P, Roberts C, Lamb A, Schultz M, Zhou Y, Giesy JP, Brinkmann M. Unveiling the Molecular Effects of Replacement and Legacy PFASs: Transcriptomic Analysis of Zebrafish Embryos Reveals Surprising Similarities and Potencies. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18554-18565. [PMID: 39392652 DOI: 10.1021/acs.est.4c04246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The prevalence of per- and poly fluoroalkyl substances (PFASs) in the environment has prompted restrictions on legacy PFASs due to their recognized toxic effects. Consequently, alternative "replacement" PFASs have been introduced and are prevalent in environmental matrices. Few studies have investigated the molecular effects of both legacy and replacement PFASs under short-term exposures. This study aimed to address this by utilizing transcriptomic sequencing to compare the molecular impacts of exposure to concentrations 0.001-5 mg/L of the legacy PFOS and two of its replacements, PFECHS and FBSA. Using zebrafish embryos, the research assessed apical effects (mortality, morphology, and growth), identified differentially expressed genes (DEGs) and enriched pathways, and determined transcriptomic points of departure (tPoDs) for each compound. Results indicated that PFOS exhibited the highest relative potency, followed by PFECHS and then FBSA. While similarities were observed among the ranked DEGs across all compounds, over-representation analysis revealed slight differences. Notably, PFOS demonstrated the lowest tPoD identified to date. These findings raise concerns regarding the safety of emerging replacement PFASs and challenge assumptions about PFAS toxicity solely resulting from their accumulative potential. As replacement PFASs proliferate in the environment, this study underscores the need for heightened scrutiny of their effects and questions current regulatory thresholds.
Collapse
Affiliation(s)
- Hannah Mahoney
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Phillip Ankley
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Catherine Roberts
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Alicia Lamb
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Matthew Schultz
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Yutong Zhou
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - John P Giesy
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B4, Canada
- Department of Integrative Biology and Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, Texas 76798-7266, United States
| | - Markus Brinkmann
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan S7N 5B3, Canada
- School of Environment and Sustainability, University of Saskatchewan, 117 Science Pl, Saskatoon, Saskatchewan S7N 5C8, Canada
- Global Institute for Water Security, University of Saskatchewan, 11 Innovation Blvd, Saskatoon, Saskatchewan S7N 3H5, Canada
- Centre for Hydrology, University of Saskatchewan, 121 Research Dr, Saskatoon, Saskatchewan S7N 1K2, Canada
| |
Collapse
|
7
|
Waye AA, Ticiani E, Sharmin Z, Perez Silos V, Perera T, Tu A, Buhimschi IA, Murga-Zamalloa CA, Hu YS, Veiga-Lopez A. Reduced bioenergetics and mitochondrial fragmentation in human primary cytotrophoblasts induced by an EGFR-targeting chemical mixture. CHEMOSPHERE 2024; 364:143301. [PMID: 39251161 PMCID: PMC11540307 DOI: 10.1016/j.chemosphere.2024.143301] [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: 05/17/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Exposures to complex environmental chemical mixtures during pregnancy reach and target the feto-placental unit. This study investigates the influence of environmental chemical mixtures on placental bioenergetics. Recognizing the essential role of the epidermal growth factor receptor (EGFR) in placental development and its role in stimulating glycolysis and mitochondrial respiration in trophoblast cells, we explored the effects of chemicals known to disrupt EGFR signaling on cellular energy production. Human primary cytotrophoblasts (hCTBs) and a first-trimester extravillous trophoblast cell line (HTR-8/SVneo) were exposed to a mixture of EGFR-interfering chemicals, including atrazine, bisphenol S, niclosamide, PCB-126, PCB-153, and trans-nonachlor. An RNA sequencing approach revealed that the mixture altered the transcriptional signature of genes involved in cellular energetics. Next, the impact of the mixture on cellular bioenergetics was evaluated using a combination of mitochondrial and glycolytic stress tests, ATP production, glucose consumption, lactate synthesis, and super-resolution imaging. The chemical mixture did not alter basal oxygen consumption but diminished the maximum respiratory capacity in a dose-dependent manner, indicating a disruption of mitochondrial function. The respiratory capacity and ATP production were increased by EGF, while the Chem-Mix reduced both EGF- and non-EGF-mediated oxygen consumption rate in hCTBs. A similar pattern was observed in the glycolytic medium acidification, with EGF increasing the acidification, and the Chem-Mix blocking EGF-induced glycolytic acidification. Furthermore, direct stochastic optical reconstruction microscopy (dSTORM) imaging demonstrated that the Chem-Mix led to a reduction of the mitochondrial network architecture, with findings supported by a decrease in the abundance of OPA1, a mitochondrial membrane GTPase involved in mitochondrial fusion. In conclusion, we demonstrated that a mixture of EGFR-disrupting chemicals alters mitochondrial remodeling, resulting in disturbed cellular bioenergetics, reducing the capacity of human cytotrophoblast cells to generate energy. Future studies should investigate the mechanism by which mitochondrial dynamics are disrupted and the pathological significance of these findings.
Collapse
Affiliation(s)
- Anita A Waye
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Elvis Ticiani
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Zinat Sharmin
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | | | - Thilini Perera
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Alex Tu
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Irina A Buhimschi
- Department of Obstetrics & Gynecology, University of Illinois Chicago, Chicago, IL, USA
| | | | - Ying S Hu
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA; The Chicago Center for Health and Environment, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
8
|
Mahoney H, da Silva F, Brinkmann M, Giesy JP. Mixtures of legacy and replacement perfluorosulphonic acids (PFSAs) demonstrate ratio-, concentration- and endpoint-dependent synergistic interactions in vitro. CHEMOSPHERE 2024; 361:142446. [PMID: 38801907 DOI: 10.1016/j.chemosphere.2024.142446] [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: 01/08/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
The extensive use of poly- and per-fluoroalkyl substances (PFASs) has les to their widespread presence in the environment, raising concerns about potential toxicity. While certain PFASs of concern have been phased-out or banned, new PFASs continue to be produced. Two such substances are perfluoroethylcyclohexane sulphonate (PFECHS) and perfluorobutane sulphamide (FBSA), replacements of perfluoroctanesulphonic acid (PFOS) that have recently been detected in multiple environmental media around the globe. Despite PFASs generally occurring in the environment as mixtures, few data are available outlining the effects of PFAS mixtures. Therefore, this research investigated the interaction potential of binary and ternary mixtures of emerging and legacy PFASs. The immortalized rainbow trout gill cell line (RTgill-W1) was chosen as the experimental model to investigate two apical endpoints: cytotoxicity and phospholipidosis. RTgill-W1 cells were exposed for 24 h to each compound to obtain endpoint-specific effect concentrations (LCx; ECx). These values were then applied to formulate mixture predictions following the Loewes Additivity and Steel and Peckham methods. Based on cytotoxicity, relative potencies of individual compounds were: PFOS > PFECHS > FSBA. PFOS and PFECHS had nearly identical effects on phospholipidosis, while FSBA did not have any effects. Most mixtures had a synergistic effect on cytotoxicity, but the effect was both dose- and ratio-dependent. PFOS and PFECHS were additive at lower concentrations (LC10) and synergistic at higher concentrations (LC50; 3:1, 1:1, and 1:3). PFECHS and FSBA mixtures were synergistic at all doses and ratios (3:1, 1:1, 1:3), while FBSA and PFOS were mainly synergistic at higher concentrations and at ratios favouring PFOS (1:1, 1:3). Tertiary combinations were mainly synergistic. For phospholipidosis, mixtures were strictly additive. These results are strongly suggestive of synergism between emerging PFAS replacements and highlight that independent apical mechanisms of different PFASs could combine to induce unexpected toxicity. Considering that emerging replacements are continuing to increase in concentration in the environment, such mixture scenarios are also likely to continue to increase in probability.
Collapse
Affiliation(s)
- Hannah Mahoney
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan, Canada
| | - Francisco da Silva
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan, Canada
| | - Markus Brinkmann
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan, Canada; School of Environment and Sustainability, University of Saskatchewan, 117 Science Pl, Saskatoon, Saskatchewan, Canada; Global Institute for Water Security, University of Saskatchewan, 11 Innovation Blvd, Saskatoon, Saskatchewan, Canada; Centre for Hydrology, University of Saskatchewan, 121 Research Dr, Saskatoon, Saskatchewan, Canada.
| | - John P Giesy
- Toxicology Center, University of Saskatchewan, 44 Campus Dr, Saskatoon, Saskatchewan, Canada; Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Department of Integrative Biology and Center for Integrative Toxicology, Michigan State University, MI, USA; Department of Environmental Science, Baylor University, One Bear Place #97266, TX, USA
| |
Collapse
|
9
|
Carlin DJ, Rider CV. Combined Exposures and Mixtures Research: An Enduring NIEHS Priority. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:75001. [PMID: 38968090 PMCID: PMC11225971 DOI: 10.1289/ehp14340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/25/2024] [Accepted: 06/12/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND The National Institute of Environmental Health Sciences (NIEHS) continues to prioritize research to better understand the health effects resulting from exposure to mixtures of chemical and nonchemical stressors. Mixtures research activities over the last decade were informed by expert input during the development and deliberations of the 2011 NIEHS Workshop "Advancing Research on Mixtures: New Perspectives and Approaches for Predicting Adverse Human Health Effects." NIEHS mixtures research efforts since then have focused on key themes including a) prioritizing mixtures for study, b) translating mixtures data from in vitro and in vivo studies, c) developing cross-disciplinary collaborations, d) informing component-based and whole-mixture assessment approaches, e) developing sufficient similarity methods to compare across complex mixtures, f) using systems-based approaches to evaluate mixtures, and g) focusing on management and integration of mixtures-related data. OBJECTIVES We aimed to describe NIEHS driven research on mixtures and combined exposures over the last decade and present areas for future attention. RESULTS Intramural and extramural mixtures research projects have incorporated a diverse array of chemicals (e.g., polycyclic aromatic hydrocarbons, botanicals, personal care products, wildfire emissions) and nonchemical stressors (e.g., socioeconomic factors, social adversity) and have focused on many diseases (e.g., breast cancer, atherosclerosis, immune disruption). We have made significant progress in certain areas, such as developing statistical methods for evaluating multiple chemical associations in epidemiology and building translational mixtures projects that include both in vitro and in vivo models. DISCUSSION Moving forward, additional work is needed to improve mixtures data integration, elucidate interactions between chemical and nonchemical stressors, and resolve the geospatial and temporal nature of mixture exposures. Continued mixtures research will be critical to informing cumulative impact assessments and addressing complex challenges, such as environmental justice and climate change. https://doi.org/10.1289/EHP14340.
Collapse
Affiliation(s)
- Danielle J. Carlin
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Cynthia V. Rider
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
10
|
Heintz MM, Klaren WD, East AW, Haws LC, McGreal SR, Campbell RR, Thompson CM. Comparison of transcriptomic profiles between HFPO-DA and prototypical PPARα, PPARγ, and cytotoxic agents in wild-type and PPARα knockout mouse hepatocytes. Toxicol Sci 2024; 200:183-198. [PMID: 38574385 PMCID: PMC11199908 DOI: 10.1093/toxsci/kfae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
Recent in vitro transcriptomic analyses for the short-chain polyfluoroalkyl substance, HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate), support conclusions from in vivo data that HFPO-DA-mediated liver effects in mice are part of the early key events of the peroxisome proliferator-activated receptor alpha (PPARα) activator-induced rodent hepatocarcinogenesis mode of action (MOA). Transcriptomic responses in HFPO-DA-treated rodent hepatocytes have high concordance with those treated with a PPARα agonist and lack concordance with those treated with PPARγ agonists or cytotoxic agents. To elucidate whether HFPO-DA-mediated transcriptomic responses in mouse liver are PPARα-dependent, additional transcriptomic analyses were conducted on samples from primary PPARα knockout (KO) and wild-type (WT) mouse hepatocytes exposed for 12, 24, or 72 h with various concentrations of HFPO-DA, or well-established agonists of PPARα (GW7647) and PPARγ (rosiglitazone), or cytotoxic agents (acetaminophen or d-galactosamine). Pathway and predicted upstream regulator-level responses were highly concordant between HFPO-DA and GW7647 in WT hepatocytes. A similar pattern was observed in PPARα KO hepatocytes, albeit with a distinct temporal and concentration-dependent delay potentially mediated by compensatory responses. This delay was not observed in PPARα KO hepatocytes exposed to rosiglitazone, acetaminophen, d-galactosamine. The similarity in transcriptomic signaling between HFPO-DA and GW7647 in both the presence and absence of PPARα in vitro indicates these compounds share a common MOA.
Collapse
|
11
|
Dauwe Y, Mary L, Oliviero F, Dubois L, Rousseau-Bacquie E, Gomez J, Gayrard V, Mselli-Lakhal L. Synergistic Steatosis Induction in Mice: Exploring the Interactions and Underlying Mechanisms between PFOA and Tributyltin. Cells 2024; 13:940. [PMID: 38891072 PMCID: PMC11171786 DOI: 10.3390/cells13110940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially characterized by steatosis and potentially progresses to steatohepatitis and fibrosis. Epidemiological studies have linked NAFLD occurrence to the exposure to environmental contaminants like PFAS. This study aims to assess the simultaneous activation of nuclear receptors via perfluorooctanoic acid (PFOA) and RXR coactivation via Tributyltin (TBT), examining their combined effects on steatogenic mechanisms. Mice were exposed to PFOA (10 mg/kg/day), TBT (5 mg/kg/day) or a combination of them for three days. Mechanisms underlying hepatic steatosis were explored by measuring nuclear receptor target gene and lipid metabolism key gene expressions, by quantifying plasma lipids and hepatic damage markers. This study elucidated the involvement of the Liver X Receptor (LXR) in the combined effect on steatosis and highlighted the permissive nature of the LXR/RXR heterodimer. Antagonistic effects of TBT on the PFOA-induced activation of the Pregnane X Receptor (PXR) and Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) were also observed. Overall, this study revealed complex interactions between PFOA and TBT, shedding light on their combined impact on liver health.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Laïla Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), Ecole Nationale Veterinaire de Toulouse (ENVT), INP-Purpan, Université Paul Sabatier (UPS), 31027 Toulouse, France; (Y.D.); (L.M.); (F.O.); (L.D.); (E.R.-B.); (J.G.); (V.G.)
| |
Collapse
|
12
|
Azhagiya Singam E, Durkin KA, La Merrill MA, Furlow JD, Wang JC, Smith MT. Prediction of the Interactions of a Large Number of Per- and Poly-Fluoroalkyl Substances with Ten Nuclear Receptors. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:4487-4499. [PMID: 38422483 PMCID: PMC10938639 DOI: 10.1021/acs.est.3c05974] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Abstract
Per- and poly-fluoroalkyl substances (PFASs) are persistent, toxic chemicals that pose significant hazards to human health and the environment. Screening large numbers of chemicals for their ability to act as endocrine disruptors by modulating the activity of nuclear receptors (NRs) is challenging because of the time and cost of in vitro and in vivo experiments. For this reason, we need computational approaches to screen these chemicals and quickly prioritize them for further testing. Here, we utilized molecular modeling and machine-learning predictions to identify potential interactions between 4545 PFASs with ten different NRs. The results show that some PFASs can bind strongly to several receptors. Further, PFASs that bind to different receptors can have very different structures spread throughout the chemical space. Biological validation of these in silico findings should be a high priority.
Collapse
Affiliation(s)
| | - Kathleen A. Durkin
- Molecular
Graphics and Computation Facility, College of Chemistry, University of California, Berkeley, California 94720, United States
| | - Michele A. La Merrill
- Department
of Environmental Toxicology, University
of California, Davis, California 95616, United States
| | - J. David Furlow
- Department
of Neurobiology, Physiology and Behavior, University of California, Davis California 95616, United States
| | - Jen-Chywan Wang
- Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, United States
| | - Martyn T. Smith
- Division
of Environmental Health Sciences, School of Public Health, University of California Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
13
|
Ren W, Wang Z, Guo H, Gou Y, Dai J, Zhou X, Sheng N. GenX analogs exposure induced greater hepatotoxicity than GenX mainly via activation of PPARα pathway while caused hepatomegaly in the absence of PPARα in female mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123314. [PMID: 38218542 DOI: 10.1016/j.envpol.2024.123314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Despite their use as substitutes for perfluorooctanoic acid, the potential toxicities of hexafluoropropylene oxide dimer acid (HFPO-DA, commercial name: GenX) and its analogs (PFDMOHxA, PFDMO2HpA, and PFDMO2OA) remain poorly understood. To assess the hepatotoxicity of these chemicals on females, each chemical was orally administered to female C57BL/6 mice at the dosage of 0.5 mg/kg/d for 28 d. The contribution of peroxisome proliferator-activated receptors (PPARα and γ) and other nuclear receptors involving in these toxic effects of GenX and its analogs were identified by employing two PPAR knockout mice (PPARα-/- and PPARγΔHep) in this study. Results showed that the hepatotoxicity of these chemicals increased in the order of GenX < PFDMOHxA < PFDMO2HpA < PFDMO2OA. The increases of relative liver weight and liver injury markers were significantly much lower in PPARα-/- mice than in PPARα+/+ mice after GenX analog exposure, while no significant differences were observed between PPARγΔHep and its corresponding wildtype groups (PPARγF/F mice), indicating that GenX analog induce hepatotoxicity mainly via PPARα instead of PPARγ. The PPARα-dependent complement pathways were inhibited in PFDMO2HpA and PFDMO2OA exposed PPARα+/+ mice, which might be responsible for the observed liver inflammation. In PPARα-/- mice, hepatomegaly and increased liver lipid content were observed in PFDMO2HpA and PFDMO2OA treated groups. The activated pregnane X receptor (PXR) and constitutive activated receptor (CAR) pathways in the liver of PPARα-/- mice, which were highlighted by bioinformatics analysis, provided a reasonable explanation for hepatomegaly in the absence of PPARα. Our results indicate that GenX analogs could induce more serious hepatotoxicity than GenX whether there is a PPARα receptor or not. These chemicals, especially PFDMO2HpA and PFDMO2OA, may not be appropriate PFOA alternatives.
Collapse
Affiliation(s)
- Wanlan Ren
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiru Wang
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Guo
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yong Gou
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xuming Zhou
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nan Sheng
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
14
|
Gao Y, Zhang Y, Luo J, Mao D, Lei X, Liu C, Zhang S, Yao Q, Li J, Zhang J, Yu X, Tian Y. Effect modification by maternal vitamin D status in the association between prenatal exposure to per- and polyfluoroalkyl substances and neurodevelopment in 2-year-old children. ENVIRONMENT INTERNATIONAL 2024; 185:108563. [PMID: 38461776 DOI: 10.1016/j.envint.2024.108563] [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: 12/17/2023] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Pregnant women in the Shanghai Birth Cohort (SBC) of China faced dual threats of per- and polyfluoroalkyl substances (PFAS) exposure and vitamin D (VD) insufficiency, potentially impacting offspring neurodevelopment. However, little is known about whether maternal VD status modifies PFAS-related neurodevelopment effect. OBJECTIVES To explore the modifying role of maternal VD status in the effect of prenatal PFAS exposure on childhood neurodevelopment. METHODS We included 746 mother-child pairs from the SBC. Ten PFAS congeners and VD levels were measured in maternal blood samples collected during the first and second trimester respectively. At 2 years of age, toddlers underwent neurodevelopment assessments using Bayley-III Scales. Multivariate linear, logistic regression, and weighted quantile sum approach were used to estimate associations of Bayley-III scores with individual and mixture PFAS. We stratified participants into VD sufficient and insufficient groups and further balanced PFAS differences between these groups by matching all PFAS levels. We fitted the same statistical models in each VD group before and after matching. RESULTS Nearly half (46.5 %) of pregnant women were VD insufficient (<30 ng/mL). In the overall population, PFAS exposure was associated with lower language scores and an increased risk for neurodevelopmental delay, but higher cognitive scores. However, adverse associations with PFAS were mainly observed in the VD sufficient group, while the VD insufficient group showed positive cognitive score associations. Higher PFAS concentrations were found in the VD sufficient group compared to the VD insufficient group. Post-matching, adverse associations in the VD sufficient group were nullified, whereas in the VD insufficient group, positive associations disappeared and adverse associations becoming more pronounced. CONCLUSION In this Chinese birth cohort, high prenatal PFAS exposure and low maternal VD levels collectively heighten the risk of adverse childhood neurodevelopment. However, disentangling PFAS and VD interrelationships is crucial to avoid paradoxical findings.
Collapse
Affiliation(s)
- Yu Gao
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, PR China; Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China
| | - Yan Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China
| | - Jiajun Luo
- Institute for Population and Precision Health, the University of Chicago, Chicago, IL, United States
| | - Dandan Mao
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China
| | - Xiaoning Lei
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China
| | - Chong Liu
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China
| | - Shanyu Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China
| | - Qian Yao
- Clinical Research Unit, Shanghai Pulmonary Hospital, 200433 Shanghai, PR China
| | - Jiong Li
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Epidemiology, School of Public Health, Nanjing Medical University, 211166 Nanjing, PR China
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, PR China
| | - Xiaodan Yu
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, PR China.
| | - Ying Tian
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, PR China; Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, PR China.
| |
Collapse
|
15
|
Waye AA, Ticiani E, Veiga-Lopez A. Chemical mixture that targets the epidermal growth factor pathway impairs human trophoblast cell functions. Toxicol Appl Pharmacol 2024; 483:116804. [PMID: 38185387 PMCID: PMC11212468 DOI: 10.1016/j.taap.2024.116804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
Pregnant women are exposed to complex chemical mixtures, many of which reach the placenta. Some of these chemicals interfere with epidermal growth factor receptor (EGFR) activation, a receptor tyrosine kinase that modulates several placenta cell functions. We hypothesized that a mixture of chemicals (Chem-Mix) known to reduce EGFR activation (polychlorinated biphenyl (PCB)-126, PCB-153, atrazine, trans-nonachlor, niclosamide, and bisphenol S) would interfere with EGFR-mediated trophoblast cell functions. To test this, we determined the chemicals' EGFR binding ability, EGFR and downstream effectors activation, and trophoblast functions (proliferation, invasion, and endovascular differentiation) known to be regulated by EGFR in extravillous trophoblasts (EVTs). The Chem-Mix competed with EGF for EGFR binding, however only PCB-153, niclosamide, trans-nonachlor, and BPS competed for binding as single chemicals. The effects of the Chem-Mix on EGFR phosphorylation were tested by exposing the placental EVT cell line, HTR-8/SVneo to control (0.1% DMSO), Chem-Mix (1, 10, or 100 ng/ml), EGF (30 ng/ml), or Chem-Mix + EGF. The Chem-Mix - but not the individual chemicals - reduced EGF-mediated EGFR phosphorylation in a dose dependent manner, while no effect was observed in its downstream effectors (AKT and STAT3). None of the individual chemicals affected EVT cell invasion, but the Chem-Mix reduced EVT cell invasion independent of EGF. In support of previous studies that have explored chemicals targeting a specific pathway (estrogen/androgen receptor), current findings indicate that exposure to a chemical mixture that targets the EGFR pathway can result in a greater impact compared to individual chemicals in the context of placental cell functions.
Collapse
Affiliation(s)
- Anita A Waye
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Elvis Ticiani
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA; The Chicago Center for Health and Environment, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Tanabe P, Key PB, Chung KW, Pisarski EC, Reiner JL, Rodowa AE, Magnuson JT, DeLorenzo ME. Mixture Effects of Per- and Polyfluoroalkyl Substances on Embryonic and Larval Sheepshead Minnows ( Cyprinodon variegatus). TOXICS 2024; 12:91. [PMID: 38276726 PMCID: PMC10819493 DOI: 10.3390/toxics12010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are ubiquitous and persistent environmental contaminants originating from many everyday products. Perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) are two PFAS that are commonly found at high concentrations in aquatic environments. Both chemicals have previously been shown to be toxic to fish, as well as having complex and largely uncharacterized mixture effects. However, limited information is available on marine and estuarine species. In this study, embryonic and larval sheepshead minnows (Cyprinodon variegatus) were exposed to several PFAS mixtures to assess lethal and sublethal effects. PFOS alone was acutely toxic to larvae, with a 96 h LC50 of 1.97 mg/L (1.64-2.16). PFOS + PFOA resulted in a larval LC50 of 3.10 (2.62-3.79) mg/L, suggesting an antagonistic effect. These observations were supported by significant reductions in malondialdehyde (105% ± 3.25) and increases in reduced glutathione concentrations (43.8% ± 1.78) in PFOS + PFOA exposures compared to PFOS-only treatments, indicating reduced oxidative stress. While PFOA reduced PFOS-induced mortality (97.0% ± 3.03), perfluorohexanoic acid (PFHxA) and perfluorobutanoic acid (PFBA) did not. PFOS alone did not affect expression of peroxisome proliferator-activated receptor alpha (pparα) but significantly upregulated apolipoprotein A4 (apoa4) (112.4% ± 17.8), a downstream product of pparα, while none of the other individually tested PFAS affected apoa4 expression. These findings suggest that there are antagonistic interactions between PFOA and PFOS that may reduce mixture toxicity in larval sheepshead minnows through reduced oxidative stress. Elucidating mechanisms of toxicity and interactions between PFAS will aid environmental regulation and management of these ubiquitous pollutants.
Collapse
Affiliation(s)
- Philip Tanabe
- National Oceanic and Atmospheric Administration, National Ocean Service, National Centers for Coastal Ocean Science, Charleston, SC 29412, USA; (P.B.K.); (K.W.C.); (E.C.P.); (M.E.D.)
| | - Peter B. Key
- National Oceanic and Atmospheric Administration, National Ocean Service, National Centers for Coastal Ocean Science, Charleston, SC 29412, USA; (P.B.K.); (K.W.C.); (E.C.P.); (M.E.D.)
| | - Katy W. Chung
- National Oceanic and Atmospheric Administration, National Ocean Service, National Centers for Coastal Ocean Science, Charleston, SC 29412, USA; (P.B.K.); (K.W.C.); (E.C.P.); (M.E.D.)
| | - Emily C. Pisarski
- National Oceanic and Atmospheric Administration, National Ocean Service, National Centers for Coastal Ocean Science, Charleston, SC 29412, USA; (P.B.K.); (K.W.C.); (E.C.P.); (M.E.D.)
| | - Jessica L. Reiner
- National Institute of Standards and Technology, Charleston, SC 29412, USA;
| | - Alix E. Rodowa
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA;
| | - Jason T. Magnuson
- U.S. Geological Survey, Columbia Environmental Research Center, Columbia, MO 65201, USA;
| | - Marie E. DeLorenzo
- National Oceanic and Atmospheric Administration, National Ocean Service, National Centers for Coastal Ocean Science, Charleston, SC 29412, USA; (P.B.K.); (K.W.C.); (E.C.P.); (M.E.D.)
| |
Collapse
|
17
|
Qin H, Lang Y, Wang Y, Cui W, Niu Y, Luan H, Li M, Zhang H, Li S, Wang C, Liu W. Adipogenic and osteogenic effects of OBS and synergistic action with PFOS via PPARγ-RXRα heterodimers. ENVIRONMENT INTERNATIONAL 2024; 183:108354. [PMID: 38043320 DOI: 10.1016/j.envint.2023.108354] [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: 09/13/2023] [Revised: 11/08/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Sodium p-perfluorous nonenoxybenzenesulfonate (OBS) is a novel alternative to perfluorooctane sulfonate (PFOS), with environmental health risks largely unknown. The present study aims to unravel the adipogenesis effects and underlying molecular initiating events of OBS, which are crucial for understanding and predicting its adverse outcome. In undifferentiated human mesenchymal stem cells (hMSCs), exposure to 1-100 nM of OBS for 7 days stimulated reactive oxygen species production. In the subsequent multipotent differentiation, hMSCs favored adipogenesis and repressed osteogenesis. The point of departure (PoD) for cellular responses of OBS was 38.85 nM, higher than PFOS (0.39 nM). Notably, OBS/PFOS co-exposure inhibited osteogenesis and synergistically promoted adipogenesis. Consistently, the expression of adipogenic marker genes was up-regulated, while that of osteogenic marker genes was down-regulated. The decreased adiponectin and elevated tumor necrosis factor α (TNFα) secretion were observed in differentiated cells exposed to the mixture of OBS and PFOS. The co-treatment of a peroxisome proliferator-activated receptor γ (PPARγ) antagonist alleviated the adipogenic effects of PFOS and its combination with OBS. Moreover, OBS/PFOS co-exposure induced peroxisome PPARγ activation in reporter gene assays, and increased formation of PPARγ - retinoid X receptor α (RXRα) heterodimers measured by co-immunoprecipitation assays. Molecular docking showed interaction energy of OBS (-20.7 kcal/mol) with intact PPARγ-RXRα complex was lower than that of PFOS (-25.9 kcal/mol). Overall, single OBS exhibited lower potency in inducing adipogenesis but is comparable to PFOS in repressing osteogenesis, whereas OBS/PFOS co-exposure increases interaction with PPARγ-RXRα heterodimers, resulting in the synergistic activation of PPARγ, ultimately enhancing adipogenesis at the expense of osteogenic differentiation. The results indicate the potential health risks of increased obesity and decreased bone density caused by OBS and its co-exposure with PFOS, as well as other perfluorinated alkylated substances mixtures.
Collapse
Affiliation(s)
- Hui Qin
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Yueming Lang
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Yiteng Wang
- Central Hospital of Dalian University of Technology, Sports Medicine Department, Dalian 116021, China
| | - Wei Cui
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Yuxin Niu
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Haiyang Luan
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Minghan Li
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Han Zhang
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian 116024, China
| | - Chenxi Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Wei Liu
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
18
|
Yun J, Kwon SC. The Association of Perfluoroalkyl Substance Exposure and a Serum Liver Function Marker in Korean Adults. TOXICS 2023; 11:965. [PMID: 38133366 PMCID: PMC10748130 DOI: 10.3390/toxics11120965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Perfluoroalkyl substances (PFAS), widely used throughout industry and daily life, are currently one of the environmental pollutants garnering the most attention worldwide. Recently, environmental pollutants have had a high profile as one of the main causes of chronic liver disease, such as non-alcoholic fatty liver disease. Research on PFAS is actively underway. Although Korea has a remarkably high prevalence of chronic liver disease, and it continues to increase, only a few studies have revealed the relationship between PFAS and liver disease. In addition, regulations on PFAS in Korea are delayed compared to developed countries, such as Europe and the United States, and public interest is insufficient compared to others. Therefore, we would like to investigate the exposure of Koreans to PFAS in the blood and examine the relationship between these substances and markers of liver function (AST, ALT, and GGT). This study was based on the results of the Korean National Environmental Health Survey (KoNEHS) 2018–2020 (Cycle 4), and a total of 2961 subjects were selected. The concentration of PFAS in the blood of Korean adults was measured to be significantly higher based on the geometric mean compared to the results of recently investigated American adults based on the National Health and Nutrition Examination Survey (NHANES, 2017-2018). A multivariable linear regression analysis adjusted for age, sex, body mass index (BMI), smoking status, alcohol intake, and regular exercise was performed to examine changes in three liver function markers as the serum PFAS concentration increased. We found that some of the five PFAS (PFOA, PFOS, PFHxS, PFNA, and PFDeA) were significantly associated with increased liver enzymes. It is necessary to recognize the threat of PFAS to the human body and to discuss regulations and alternatives in earnest. Continuous follow-up studies are required through a well-designed cohort.
Collapse
Affiliation(s)
| | - Soon-Chan Kwon
- Department of Occupational and Environmental Medicine, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea;
| |
Collapse
|
19
|
Murase W, Kubota A, Ikeda-Araki A, Terasaki M, Nakagawa K, Shizu R, Yoshinari K, Kojima H. Effects of perfluorooctanoic acid (PFOA) on gene expression profiles via nuclear receptors in HepaRG cells: Comparative study with in vitro transactivation assays. Toxicology 2023:153577. [PMID: 37302725 DOI: 10.1016/j.tox.2023.153577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
Perfluorooctanoic acid (PFOA), a synthetic perfluorinated eight-carbon organic chemical, has been reported to induce hepatotoxicity, including increased liver weight, hepatocellular hypertrophy, necrosis, and increased peroxisome proliferation in rodents. Epidemiological studies have demonstrated associations between serum PFOA levels and various adverse effects. In this study, we investigated the gene expression profiles of human HepaRG cells exposed to 10 and 100 μM PFOA for 24h. Treatment with 10 and 100 μM PFOA significantly modulated the expression of 190 genes and 996 genes, respectively. In particular, genes upregulated or downregulated by 100µM PFOA included peroxisome proliferator-activated receptor (PPAR) signaling genes related to lipid metabolism, adipocyte differentiation, and gluconeogenesis. In addition, we identified the "Nuclear receptors-meta pathways" following the activation of other nuclear receptors: constitutive androstane receptor (CAR), pregnane X receptor (PXR) and farnesoid X receptor (FXR), and the transcription factor, nuclear factor E2-related factor 2 (Nrf2). The expression levels of some target genes (CYP4A11, CYP2B6, CYP3A4, CYP7A1, and GPX2) of these nuclear receptors and Nrf2 were confirmed using quantitative reverse transcription polymerase chain reaction. Next, we performed transactivation assays using COS-7 or HEK293 cells to investigate whether these signaling-pathways were activated by the direct effects of PFOA on human PPARα, CAR, PXR, FXR and Nrf2. PFOA activated PPARα in a concentration-dependent manner, but did not activate CAR, PXR, FXR, or Nrf2. Taken together, these results suggest that PFOA affects the hepatic transcriptomic responses of HepaRG cells through direct activation of PPARα and indirect activation of CAR, PXR FXR and Nrf2. Our finding indicates that PPARα activation found in the "Nuclear receptors-meta pathways" functions as a molecular initiating event for PFOA, and indirect activation of alternative nuclear receptors and Nrf2 also provide important molecular mechanisms in PFOA-induced human hepatotoxicity.
Collapse
Affiliation(s)
- Wataru Murase
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Atsuhito Kubota
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Atsuko Ikeda-Araki
- Faculty of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-ku, Sapporo 060-0812, Japan; Center for Environmental and Health Sciences, Hokkaido University, Kita-12, Nishi-7, Kita-ku, Sapporo 060-0812, Japan
| | - Masaru Terasaki
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan; Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Koji Nakagawa
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan; Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Ryota Shizu
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kouichi Yoshinari
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiroyuki Kojima
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan; Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan.
| |
Collapse
|
20
|
Wen ZJ, Wei YJ, Zhang YF, Zhang YF. A review of cardiovascular effects and underlying mechanisms of legacy and emerging per- and polyfluoroalkyl substances (PFAS). Arch Toxicol 2023; 97:1195-1245. [PMID: 36947184 DOI: 10.1007/s00204-023-03477-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Cardiovascular disease (CVD) poses the leading threats to human health and life, and their occurrence and severity are associated with exposure to environmental pollutants. Per- and polyfluoroalkyl substances (PFAS), a group of widely used industrial chemicals, are characterized by persistence, long-distance migration, bioaccumulation, and toxicity. Some PFAS, particularly perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA) and perfluorohexanesulfonic acid (PFHxS), have been banned, leaving only legacy exposure to the environment and human body, while a number of novel PFAS alternatives have emerged and raised concerns, such as polyfluoroalkyl ether sulfonic and carboxylic acid (PFESA and PFECA) and sodium p-perfluorous nonenoxybenzene sulfonate (OBS). Overall, this review systematically elucidated the adverse cardiovascular (CV) effects of legacy and emerging PFAS, emphasized the dose/concentration-dependent, time-dependent, carbon chain length-dependent, sex-specific, and coexposure effects, and discussed the underlying mechanisms and possible prevention and treatment. Extensive epidemiological and laboratory evidence suggests that accumulated serum levels of legacy PFAS possibly contribute to an increased risk of CVD and its subclinical course, such as cardiac toxicity, vascular disorder, hypertension, and dyslipidemia. The underlying biological mechanisms may include oxidative stress, signaling pathway disturbance, lipid metabolism disturbance, and so on. Various emerging alternatives to PFAS also play increasingly prominent toxic roles in CV outcomes that are milder, similar to, or more severe than legacy PFAS. Future research is recommended to conduct more in-depth CV toxicity assessments of legacy and emerging PFAS and explore more effective surveillance, prevention, and treatment strategies, accordingly.
Collapse
Affiliation(s)
- Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi-Jing Wei
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
21
|
Heintz MM, Haws LC, Klaunig JE, Cullen JM, Thompson CM. Assessment of the mode of action underlying development of liver lesions in mice following oral exposure to HFPO-DA and relevance to humans. Toxicol Sci 2023; 192:15-29. [PMID: 36629480 PMCID: PMC10025879 DOI: 10.1093/toxsci/kfad004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)propanoate) is a short-chain polyfluorinated alkyl substance (PFAS) used in the manufacture of some types of fluorinated polymers. Like many PFAS, toxicity studies with HFPO-DA indicate the liver is the primary target of toxicity in rodents following oral exposure. Due to the structural diversity of PFAS, the mode of action (MOA) can differ between PFAS for the same target tissue. There is significant evidence for involvement of peroxisome proliferator-activated receptor alpha (PPARα) activation based on molecular and histopathological responses in the liver following HFPO-DA exposure, but other MOAs have also been hypothesized based on limited evidence. The MOA underlying the liver effects in mice exposed to HFPO-DA was assessed in the context of the Key Events (KEs) outlined in the MOA framework for PPARα activator-induced rodent hepatocarcinogenesis. The first 3 KEs (ie, PPARα activation, alteration of cell growth pathways, and perturbation of cell growth/survival) are supported by several lines of evidence from both in vitro and in vivo data available for HFPO-DA. In contrast, alternate MOAs, including cytotoxicity, PPARγ and mitochondrial dysfunction are generally not supported by the scientific literature. HFPO-DA-mediated liver effects in mice are not expected in humans as only KE 1, PPARα activation, is shared across species. PPARα-mediated gene expression in humans produces only a subset (ie, lipid modulating effects) of the responses observed in rodents. As such, the adverse effects observed in rodent livers should not be used as the basis of toxicity values for HFPO-DA for purposes of human health risk assessment.
Collapse
Affiliation(s)
| | | | - James E Klaunig
- School of Public Health, Indiana University, Bloomington, Indiana 47405, USA
| | - John M Cullen
- North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina 27606, USA
| | | |
Collapse
|
22
|
Conley JM, Lambright CS, Evans N, Medlock-Kakaley E, Dixon A, Hill D, McCord J, Strynar MJ, Ford J, Gray LE. Cumulative maternal and neonatal effects of combined exposure to a mixture of perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) during pregnancy in the Sprague-Dawley rat. ENVIRONMENT INTERNATIONAL 2022; 170:107631. [PMID: 36402036 PMCID: PMC9944680 DOI: 10.1016/j.envint.2022.107631] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/29/2022] [Accepted: 11/10/2022] [Indexed: 05/10/2023]
Abstract
Globally, biomonitoring data demonstrate virtually all humans carry residues of multiple per- and polyfluoroalkyl substances (PFAS). Despite pervasive co-exposure, limited mixtures-based in vivo PFAS toxicity research has been conducted. Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) are commonly detected PFAS in human and environmental samples and both produce adverse effects in laboratory animal studies, including maternal and offspring effects when orally administered during pregnancy and lactation. To evaluate the effects of combined exposure to PFOA and PFOS, we orally exposed pregnant Sprague-Dawley rats from gestation day 8 (GD8) to postnatal day 2 (PND2) to PFOA (10-250 mg/kg/d) or PFOS (0.1-5 mg/kg/d) individually to characterize effects and dose response curve parameters, followed by a variable-ratio mixture experiment with a constant dose of PFOS (2 mg/kg/d) mixed with increasing doses of PFOA (3-80 mg/kg/d). The mixture study design was intended to: 1) shift the PFOA dose response curves for endpoints shared with PFOS, 2) allow comparison of dose addition (DA) and response addition (RA) model predictions, 3) conduct relative potency factor (RPF) analysis for multiple endpoints, and 4) avoid overt maternal toxicity. Maternal serum and liver concentrations of PFOA and PFOS were consistent between the individual chemical and mixture experiments. Combined exposure with PFOS significantly shifted the PFOA dose response curves towards effects at lower doses compared to PFOA-only exposure for multiple endpoints and these effects were well predicted by dose addition. For endpoints amenable to mixture model analyses, DA produced equivalent or better estimates of observed data than RA. All endpoints evaluated were accurately predicted by RPF and DA approaches except for maternal gestational weight gain, which produced less-than-additive results in the mixture. Data support the hypothesis of cumulative effects on shared endpoints from PFOA and PFOS co-exposure and dose additive approaches for predictive estimates of mixture effects.
Collapse
Affiliation(s)
- Justin M Conley
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Christy S Lambright
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Nicola Evans
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Elizabeth Medlock-Kakaley
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Aaron Dixon
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Donna Hill
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - James McCord
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Environmental Measurement and Modeling, Research Triangle Park, NC, USA.
| | - Mark J Strynar
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Environmental Measurement and Modeling, Research Triangle Park, NC, USA.
| | - Jermaine Ford
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Computational Toxicology and Exposure, Research Triangle Park, NC, USA.
| | - L Earl Gray
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| |
Collapse
|
23
|
Zhang S, Lei X, Zhang Y, Shi R, Zhang Q, Gao Y, Yuan T, Li J, Tian Y. Prenatal exposure to per- and polyfluoroalkyl substances and childhood adiposity at 7 years of age. CHEMOSPHERE 2022; 307:136077. [PMID: 36002061 DOI: 10.1016/j.chemosphere.2022.136077] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND An increasing number of studies have reported that prenatal per- and polyfluoroalkyl substances (PFAS) exposure may increase childhood adiposity. However, limited data is available in China, and the overall effects of PFAS mixture remain unclear. OBJECTIVE To examine the association of prenatal exposure to individual PFAS and their mixture with childhood adiposity at 7 years of age. METHODS A total of 206 mother-infant pairs were recruited from the Laizhou Wan (Bay) Birth Cohort in China between 2010 and 2013. Ten PFAS were measured in maternal serum. The measurements of fat mass, body fat percentage, body mass index, waist circumference, waist-to-height ratio and overweight/obesity were used to assess adiposity in children aged 7. We fitted logistic regression, linear regression and weighted quantile sum (WQS) regression models to estimate the association of prenatal exposure to individual PFAS and their mixture with childhood adiposity. RESULTS We found negative associations of perfluoroheptanoic acid (PFHpA) and perfluorooctane sulfonamide (PFOSA) exposure with adiposity measurements in all children. The result from the WQS model consistently revealed that the PFAS mixture was inversely related to adiposity measurements. Each quartile increase of the PFAS mixture was associated with a 1.14 kg decrease (95% CI: -2.27, -0.02) in fat mass and a 2.32% decrease (95% CI: -4.51, -0.14) in body fat. Moreover, significant sex differences were found. PFAS mixture was negatively associated with five adiposity measurements in boys, but positively associated with all adiposity measurements except body fat percentage in girls. PFOSA, PFHpA and perfluorobutanesulfonate (PFBS) with weights >0.300 were the main contributors to the overall effects observed among all children, boys and girls, respectively. CONCLUSION This study suggests potential sex-specific associations of prenatal exposure to individual PFAS and their mixture with childhood adiposity, with the observed relationship being negative for boys but positive for girls.
Collapse
Affiliation(s)
- Shanyu Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoning Lei
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Shi
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianlong Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gao
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Yuan
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jiong Li
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Clinical Epidemiology-Department of Clinical Medicine, Arhus University Hospital, Aarhus, Denmark
| | - Ying Tian
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
24
|
Heintz MM, Chappell GA, Thompson CM, Haws LC. Evaluation of Transcriptomic Responses in Livers of Mice Exposed to the Short-Chain PFAS Compound HFPO-DA. FRONTIERS IN TOXICOLOGY 2022; 4:937168. [PMID: 35832492 PMCID: PMC9271854 DOI: 10.3389/ftox.2022.937168] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
HFPO-DA (ammonium 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate; CASRN 62037-80-3) is a component of the GenX technology platform used as a polymerization aid in the manufacture of some types of fluoropolymers. The liver is the primary target of toxicity for HFPO-DA in rodents and previous examination of hepatic transcriptomic responses in mice following oral exposure to HFPO-DA for 90 days showed induction of peroxisome proliferator-activated receptor signaling pathways, predominantly by PPARα, as well as increased gene expression of both peroxisomal and mitochondrial fatty acid metabolism. To further investigate the mechanism of liver toxicity, transcriptomic analysis was conducted on liver tissue from mice orally exposed to 0, 0.1, 0.5 or 5 mg/kg-bw/day HFPO-DA in a reproduction/developmental toxicity study. Hepatic gene expression changes demonstrated activation of the PPARα signaling pathway. Peroxisomal and mitochondrial fatty acid β-oxidation gene sets were enriched at lower HFPO-DA concentrations, and complement cascade, cell cycle and apoptosis related gene sets were enriched at higher HFPO-DA concentrations. These results support the reported histopathological findings in livers of mice from this study and indicate that the effects of HFPO-DA are mediated through rodent-specific PPARα signaling mechanisms regardless of reproductive status in mice.
Collapse
Affiliation(s)
- Melissa M. Heintz
- ToxStrategies, Inc, Asheville, NC, United States
- *Correspondence: Melissa M. Heintz,
| | | | | | | |
Collapse
|
25
|
MRA Toolbox v. 1.0: a web-based toolbox for predicting mixture toxicity of chemical substances in chemical products. Sci Rep 2022; 12:8880. [PMID: 35614210 PMCID: PMC9132927 DOI: 10.1038/s41598-022-13028-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
The chemical risk assessment paradigm is shifting from “substance-based” to “product/mixture-based” and from “animal testing” to “alternative testing” under chemical regulations. Organisms and the environment may be exposed to mixtures rather than a single substance. Conducting toxicity tests for all possible combinations is impractical due to the enormous combinatorial complexity. This study highlights the development and application case studies of Mixture Risk Assessment Toolbox, a novel web-based platform that supports mixture risk assessment through the use of different prediction models and public databases. This integrated framework provides new functional values for assessors to easily screen and compare the toxicity of mixture products using different computational techniques and find strategic solutions to reduce the mixture toxicity in the product development process. The toolbox (https://www.mratoolbox.org) includes four additive toxicity models: two conventional (Concentration Addition; and Independent Action) and two advanced (Generalized Concentration Addition; and Quantitative Structure–Activity Relationship-based Two-Stage Prediction) models. We demonstrated the multiple functions of the toolbox using three cases: (i) how it can be used to calculate the mixture toxicity, (ii) those for which safety data sheet (SDS) only indicating representative toxicity values (EC50; and LC50), and (iii) those comprising chemicals with low toxic effects.
Collapse
|