1
|
Lu EH, Rusyn I, Chiu WA. Incorporating new approach methods (NAMs) data in dose-response assessments: The future is now! JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024:1-35. [PMID: 39390665 DOI: 10.1080/10937404.2024.2412571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Regulatory dose-response assessments traditionally rely on in vivo data and default assumptions. New Approach Methods (NAMs) present considerable opportunities to both augment traditional dose-response assessments and accelerate the evaluation of new/data-poor chemicals. This review aimed to determine the potential utilization of NAMs through a unified conceptual framework that compartmentalizes derivation of toxicity values into five sequential Key Dose-response Modules (KDMs): (1) point-of-departure (POD) determination, (2) test system-to-human (e.g. inter-species) toxicokinetics and (3) toxicodynamics, (4) human population (intra-species) variability in toxicodynamics, and (5) toxicokinetics. After using several "traditional" dose-response assessments to illustrate this framework, a review is presented where existing NAMs, including in silico, in vitro, and in vivo approaches, might be applied across KDMs. Further, the false dichotomy between "traditional" and NAMs-derived data sources is broken down by organizing dose-response assessments into a matrix where each KDM has Tiers of increasing precision and confidence: Tier 0: Default/generic values, Tier 1: Computational predictions, Tier 2: Surrogate measurements, and Tier 3: Direct measurements. These findings demonstrated that although many publications promote the use of NAMs in KDMs (1) for POD determination and (5) for human population toxicokinetics, the proposed matrix of KDMs and Tiers reveals additional immediate opportunities for NAMs to be integrated across other KDMs. Further, critical needs were identified for developing NAMs to improve in vitro dosimetry and quantify test system and human population toxicodynamics. Overall, broadening the integration of NAMs across the steps of dose-response assessment promises to yield higher throughput, less animal-dependent, and more science-based toxicity values for protecting human health.
Collapse
Affiliation(s)
- En-Hsuan Lu
- Interdisciplinary Faculty of Toxicology and Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology and Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Weihsueh A Chiu
- Interdisciplinary Faculty of Toxicology and Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
2
|
Wang Y, Sun X, Xiong B, Duan M, Sun Y. Genetic and Environmental Factors Co-Contributing to Behavioral Abnormalities in adnp/ adnp2 Mutant Zebrafish. Int J Mol Sci 2024; 25:9469. [PMID: 39273418 PMCID: PMC11395604 DOI: 10.3390/ijms25179469] [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: 08/08/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Human mutations of ADNP and ADNP2 are known to be associated with neural developmental disorders (NDDs), including autism spectrum disorders (ASDs) and schizophrenia (SZ). However, the underlying mechanisms remain elusive. In this study, using CRISPR/Cas9 gene editing technology, we generated adnp and adnp2 mutant zebrafish models, which exhibited developmental delays, brain deficits, and core behavioral features of NDDs. RNA sequencing analysis of adnpa-/-; adnpb-/- and adnp2a-/-; adnp2b-/- larval brains revealed altered gene expression profiles affecting synaptic transmission, autophagy, apoptosis, microtubule dynamics, hormone signaling, and circadian rhythm regulation. Validation using whole-mount in situ hybridization (WISH) and real-time quantitative PCR (qRT-PCR) corroborated these findings, supporting the RNA-seq results. Additionally, loss of adnp and adnp2 resulted in significant downregulation of pan-neuronal HuC and neuronal fiber network α-Tubulin signals. Importantly, prolonged low-dose exposure to environmental endocrine disruptors (EEDs) aggravated behavioral abnormalities in adnp and adnp2 mutants. This comprehensive approach enhances our understanding of the complex interplay between genetic mutations and environmental factors in NDDs. Our findings provide novel insights and experimental foundations into the roles of adnp and adnp2 in neurodevelopment and behavioral regulation, offering a framework for future preclinical drug screening aimed at elucidating the pathogenesis of NDDs and related conditions.
Collapse
Affiliation(s)
- Yongxin Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyun Sun
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Xiong
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming Duan
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yuhua Sun
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
| |
Collapse
|
3
|
Wu S, Xie J, Zhao H, Zhao X, Sánchez OF, Rochet JC, Freeman JL, Yuan C. Developmental neurotoxicity of PFOA exposure on hiPSC-derived cortical neurons. ENVIRONMENT INTERNATIONAL 2024; 190:108914. [PMID: 39079332 PMCID: PMC11406754 DOI: 10.1016/j.envint.2024.108914] [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: 04/15/2024] [Revised: 06/27/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024]
Abstract
PFOA is a legacy Per- and Polyfluorinated Substances (PFAS), a group of chemicals widely used in various industrial applications and consumer products. Although there has been a voluntary phase out of PFOA since 2005, it is still widely detected in various water supplies. A growing body of evidence suggests an association between PFOA exposure, particularly during developmental stages, with increased risks of neurodegenerative diseases (NDs). The neurotoxic mechanism of developmental PFOA exposure, however, remains poorly understood. Utilizing human induced-pluripotent stem cell (hiPSC)-derived cortical neurons, we investigated the effect of PFOA exposure prior to differentiation and assessed changes in neuronal characteristics, transcriptome, and neurodegeneration markers mimicking a Developmental Origin of Health and Disease (DoHAD) paradigm. Exposure to PFOA before neuron differentiation resulted in persistent alterations in nuclear morphology, neuronal network, and calcium activity. RNA sequencing analysis further revealed transcriptomic changes aligning with Alzheimer's Disease (AD) after PFOA exposure. These observations were further corroborated by alterations in tau phosphorylation markers, the presence of fibrillar tau, an increase in liquid droplets, and a decrease in RNA translational efficiency characterized using a battery of biochemical assays. Taken together, our results revealed persistent deficits of key neuronal characteristics induced by pre-differentiation PFOA exposure, suggesting impairments in several AD-related pathways that can together contribute to the elevation of AD risk after pre-differentiation PFOA exposure.
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
| | - Xihui Zhao
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Oscar F Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, IN, 47907; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN, 47907
| | | | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN, 47907.
| |
Collapse
|
4
|
Gutsfeld S, Wehmas L, Omoyeni I, Schweiger N, Leuthold D, Michaelis P, Howey XM, Gaballah S, Herold N, Vogs C, Wood C, Bertotto L, Wu GM, Klüver N, Busch W, Scholz S, Schor J, Tal T. Investigation of Peroxisome Proliferator-Activated Receptor Genes as Requirements for Visual Startle Response Hyperactivity in Larval Zebrafish Exposed to Structurally Similar Per- and Polyfluoroalkyl Substances (PFAS). ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:77007. [PMID: 39046251 PMCID: PMC11268134 DOI: 10.1289/ehp13667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl Substances (PFAS) are synthetic chemicals widely detected in humans and the environment. Exposure to perfluorooctanesulfonic acid (PFOS) or perfluorohexanesulfonic acid (PFHxS) was previously shown to cause dark-phase hyperactivity in larval zebrafish. OBJECTIVES The objective of this study was to elucidate the mechanism by which PFOS or PFHxS exposure caused hyperactivity in larval zebrafish. METHODS Swimming behavior was assessed in 5-d postfertilization (dpf) larvae following developmental (1-4 dpf) or acute (5 dpf) exposure to 0.43 - 7.86 μ M PFOS, 7.87 - 120 μ M PFHxS, or 0.4% dimethyl sulfoxide (DMSO). After developmental exposure and chemical washout at 4 dpf, behavior was also assessed at 5-8 dpf. RNA sequencing was used to identify differences in global gene expression to perform transcriptomic benchmark concentration-response (BMC T ) modeling, and predict upstream regulators in PFOS- or PFHxS-exposed larvae. CRISPR/Cas9-based gene editing was used to knockdown peroxisome proliferator-activated receptors (ppars) pparaa/ab, pparda/db, or pparg at day 0. Knockdown crispants were exposed to 7.86 μ M PFOS or 0.4% DMSO from 1-4 dpf and behavior was assessed at 5 dpf. Coexposure with the ppard antagonist GSK3787 and PFOS was also performed. RESULTS Transient dark-phase hyperactivity occurred following developmental or acute exposure to PFOS or PFHxS, relative to the DMSO control. In contrast, visual startle response (VSR) hyperactivity only occurred following developmental exposure and was irreversible up to 8 dpf. Similar global transcriptomic profiles, BMC T estimates, and enriched functions were observed in PFOS- and PFHxS-exposed larvae, and ppars were identified as putative upstream regulators. Knockdown of pparda/db, but not pparaa/ab or pparg, blunted PFOS-dependent VSR hyperactivity to control levels. This finding was confirmed via antagonism of ppard in PFOS-exposed larvae. DISCUSSION This work identifies a novel adverse outcome pathway for VSR hyperactivity in larval zebrafish. We demonstrate that developmental, but not acute, exposure to PFOS triggered persistent VSR hyperactivity that required ppard function. https://doi.org/10.1289/EHP13667.
Collapse
Affiliation(s)
- Sebastian Gutsfeld
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Leah Wehmas
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Ifeoluwa Omoyeni
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Nicole Schweiger
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - David Leuthold
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Paul Michaelis
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Xia Meng Howey
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Shaza Gaballah
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Nadia Herold
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Carolina Vogs
- Department of Biomedical Science and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carmen Wood
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Luísa Bertotto
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Gi-Mick Wu
- Research and Development Institute for the Agri-Environment, Quebec, Quebec, Canada
| | - Nils Klüver
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Wibke Busch
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Stefan Scholz
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Jana Schor
- Department of Computational Biology and Chemistry, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Tamara Tal
- Department of Bioanalytical Ecotoxicology, Chemicals in the Environment Research Section, Helmholtz-Centre for Environmental Research–UFZ, Leipzig, Germany
- Medical Faculty, University Leipzig, Leipzig, Germany
| |
Collapse
|
5
|
Schildroth S, Claus Henn B, Vines AI, Geller RJ, Lovett SM, Coleman CM, Bethea TN, Botelho JC, Calafat AM, Milando C, Baird DD, Wegienka G, Wise LA. Per- and polyfluoroalkyl substances (PFAS), perceived stress, and depressive symptoms in a prospective cohort study of black women. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172445. [PMID: 38642767 PMCID: PMC11109747 DOI: 10.1016/j.scitotenv.2024.172445] [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: 01/23/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are endocrine-disrupting chemicals with neurotoxic properties. PFAS have been associated with depressive symptoms among women in some studies, but little research has evaluated the effects of PFAS mixtures. Further, no study has investigated interactions of PFAS-depression associations by perceived stress, which has been shown to modify the effects of PFAS on other health outcomes. OBJECTIVE In a prospective cohort study of reproductive-aged Black women, we investigated associations between PFAS and depressive symptoms and the extent to which perceived stress modified these associations. METHODS We analyzed data from 1499 participants (23-35 years) in the Study of Environment, Lifestyle, and Fibroids. We quantified concentrations of nine PFAS in baseline plasma samples using online solid-phase extraction-liquid chromatography-isotope dilution tandem mass spectrometry. Participants reported perceived stress via the Perceived Stress Scale (PSS-4; range = 0-16) at baseline and depressive symptoms via the Center for Epidemiologic Studies Depression Scale (CESD; range = 0-44) at the 20-month follow-up visit. We used Bayesian Kernel Machine Regression to estimate associations between PFAS concentrations, individually and as a mixture, and depressive symptoms, and to assess effect modification by PSS-4 scores, adjusting for confounders. RESULTS Baseline perfluorodecanoic acid concentrations were associated with greater depressive symptoms at the 20-month follow-up, but associations for other PFAS were null. The PFAS were not associated with depressive symptoms when evaluated as a mixture. The association between the 90th percentile (vs. 50th percentile) of the PFAS mixture with CES-D scores was null at the 10th (β = 0.03; 95 % CrI = 0.20, 0.25), 50th (β = 0.02; 95 % CrI = -0.16, 0.19), and 90th (β = 0.01; 95 % CrI = 0.18, 0.20) percentiles of PSS-4 scores, suggesting perceived stress did not modify the PFAS mixture. CONCLUSION In this prospective cohort study, PFAS concentrations-assessed individually or as a mixture-were not appreciably associated with depressive symptoms, and there was no evidence of effect modification by perceived stress.
Collapse
Affiliation(s)
- Samantha Schildroth
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.
| | - Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Anissa I Vines
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Ruth J Geller
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Sharonda M Lovett
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Chad M Coleman
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Traci N Bethea
- Office of Minority Health & Health Disparities Research, Georgetown Lombardi Comprehensive Cancer Institute, Washington, DC, USA
| | - Julianne Cook Botelho
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Chad Milando
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Donna D Baird
- National Institute of Environmental Health Sciences, Durham, NC, USA
| | | | - Lauren A Wise
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
6
|
Huang SN, Hu YH, Xu TT, Luan YL, Zeng LX, Zhang ZF, Guo Y. Exposure to per- and polyfluoroalkyl substances in lung cancer patients and their associations with clinical health indicators. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 350:123995. [PMID: 38636840 DOI: 10.1016/j.envpol.2024.123995] [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: 02/22/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Per- and polyfluoroalkyl substances (PFASs) have potential carcinogenicity, immunotoxicity, and hepatotoxicity. Research has been conducted on PFAS exposure in people to discuss their potential health effects, excluding lung cancer. In this study, we recruited participants (n = 282) with lung cancer from Heilongjiang Province, northeast China. The PFAS concentrations were measured in their serum to fill the data gap of exposure, and relationships were explored in levels between PFASs and clinical indicators of tumor, immune and liver function. Ten PFASs were found in over 80 % of samples and their total concentrations were 5.27-152 ng/mL, with the highest level for perfluorooctanesulfonate (median: 12.4 ng/mL). Long-chain PFASs were the main congeners and their median concentration (20.5 ng/mL) was nearly three times to that of short-chain PFASs (7.61 ng/mL). Significantly higher concentrations of perfluorobutanoic acid, perfluorononanoic acid and perfluorohexanesulfonate were found in males than in females (p < 0.05). Serum levels of neuro-specific enolase were positively associated with perfluoropentanoic acid in all participants and were negatively associated with perfluorononanesulfonate in females (p < 0.05, multiple linear regression models). Exposure to PFAS mixture was significantly positively associated with the lymphocytic absolute value (difference: 0.224, 95% CI: 0.018, 0.470; p < 0.05, quantile g-computation models) and serum total bilirubin (difference: 2.177, 95% CI: 0.0335, 4.33; p < 0.05). Moreover, PFAS exposure can affect γ-glutamyl transpeptidase through several immune markers (p < 0.05, mediating test). Our results suggest that exposure to certain PFASs could interfere with clinical indicators in lung cancer patients. To our knowledge, this is the first study to detect serum PFAS occurrence and check their associations with clinical indicators in lung cancer patients.
Collapse
Affiliation(s)
- Si-Nan Huang
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511436, China
| | - Ying-Hua Hu
- International Joint Research Center for Persistent Toxic Substances, Heilongjiang Institute of Labor Hygiene and Occupational Diseases, The Second Hospital of Heilongjiang Province, Harbin, 100028, China
| | - Ting-Ting Xu
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511436, China
| | - Yu-Ling Luan
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511436, China
| | - Li-Xi Zeng
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511436, China
| | - Zi-Feng Zhang
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, China
| | - Ying Guo
- Guangdong Provincial Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511436, China.
| |
Collapse
|
7
|
Wiklund L, Pípal M, Weiss J, Beronius A. Exploring a mechanism-based approach for the identification of endocrine disruptors using Adverse Outcome Pathways (AOPs) and New Approach Methodologies (NAMs) : A perfluorooctane sulfonic acid case study. Toxicology 2024; 504:153794. [PMID: 38580097 DOI: 10.1016/j.tox.2024.153794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
Endocrine disruptors (EDs) pose a serious threat to human health and the environment and require a comprehensive evaluation to be identified. The identification of EDs require a substantial amount of data, both in vitro and in vivo, due to the current scientific criteria in the EU. At the same time, the EU strives to reduce animal testing due to concerns regarding animal welfare and sensitivity of animal studies to adequately detect adverse effects relevant for human health. Perfluorooctane sulfonic acid (PFOS) is a persistent organic pollutant that is suspected to be an ED based on academic research, however it is not identified as such from a regulatory perspective. It has previously been shown that PFOS has the potential to cause neurotoxicity as well as affect the thyroid system, and it is known that specific thyroid hormone levels are critical in the development of the brain during. In this work, the aim was to evaluate a mechanism-based approach to identify ED properties of PFOS based on the Adverse Outcome Pathway (AOP) framework and using New Approach Methods (NAMs), by comparing this approach to an ED assessment based on the currently available guidance document. An AOP network (AOPN) was generated for the thyroid modality, and AOPs leading to developmental neurotoxicity (DNT) were identified. A literature search and screening process based on the AOPN, and systematic review methodology, was performed, followed by a rigorous Weight-of-Evidence (WoE) assessment. Evidence was mapped back onto the AOPN used for the literature search, to identify possible endocrine Modes-of-Action (MoAs) for PFOS and data gaps in the two assessments. It could be concluded that PFOS fulfils the criteria for ED classification in the standard ED assessment, but not in the mechanism-based assessment. The need for quantitative information, such as quantitative AOPs, for the mechanism-based approach is discussed. The possibility of a directly neurotoxic alternative MoA was also highlighted based on available in vitro data. Opportunities and challenges with implementing AOPs and NAMs into the regulatory assessment of EDs, and assessing hazard in the Next Generation Risk Assessment, is discussed. This case study exploring the mechanism-based approach to ED identification represents an important step toward more accurate and predictive assessment of EDs based on AOPs and NAMs, and to the Next Generation Risk Assessment (NGRA) concept.
Collapse
Affiliation(s)
- Linus Wiklund
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Marek Pípal
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jana Weiss
- Department of Environmental Science, Stockholm University, Stockholm, Sweden
| | - Anna Beronius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Reimann B, Remy S, Koppen G, Schoeters G, Den Hond E, Nelen V, Franken C, Covaci A, Bruckers L, Baeyens W, Loots I, van Larebeke N, Voorspoels S, De Henauw S, Nawrot TS, Plusquin M. Prenatal exposure to mixtures of per- and polyfluoroalkyl substances and organochlorines affects cognition in adolescence independent of postnatal exposure. Int J Hyg Environ Health 2024; 257:114346. [PMID: 38447259 DOI: 10.1016/j.ijheh.2024.114346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Studies on cognitive and neurodevelopmental outcomes have shown inconsistent results regarding the association with prenatal exposure to perfluoroalkyl substance (PFAS) and organochlorines. Assessment of mixture effects of correlated chemical exposures that persist in later life may contribute to the unbiased evaluation and understanding of dose-response associations in real-life exposures. METHODS For a subset of the 4th Flemish Environment and Health Study (FLEHS), concentrations of four PFAS and six organochlorines were measured in respectively 99 and 153-160 cord plasma samples and 15 years later in adolescents' peripheral serum by Ultra Performance Liquid Chromatography-Tandem Mass Spectrometry (UPLC-MS/MS). Sustained and selective attention were measured at 14-15 years with the Continuous Performance Test (CPT) and Stroop Test as indicators of potential neurodevelopmental deficits. Quantile g-computation was applied to assess the joint associations between prenatal exposure to separate and combined groups of PFAS and organochlorines and performance in the CPT and Stroop Test at adolescence. Subsequently, individual effects of each chemical compound were analyzed in mixed effects models with two sets of covariates. Analytical data at birth and at the time of cognitive assessment allowed for off-setting postnatal exposure. RESULTS In mixtures analysis, a simultaneous one-quantile increase in the natural log-transformed values of PFAS and organochlorines combined was associated with a decrease in the mean reaction time (RT) and the reaction time variability (RTV) in the CPT (β = -15.54, 95% CI:-29.64, -1.45, and β = -7.82, 95% CI: -14.97, -0.67 respectively) and for the mixture of PFAS alone with RT (β = -11.94, 95% CI: -23.29, -0.60). In the single pollutant models, these results were confirmed for the association between perfluorohexanesulfonate (PFHxS) with RT (β = -17.95, 95% CI = -33.35, -2.69) and hexachlorobenzene with RTV in the CPT (β = -5.78, 95% CI: -10.39, -0.76). Furthermore, the participants with prenatal exposure above the limit of quantification for perfluorononanoic acid (PFNA) had a significantly shorter RT and RTV in the CPT (β = -23.38, 95% CI: -41.55, -5.94, and β = -9.54, 95% CI: -19.75, -0.43, respectively). CONCLUSION Higher prenatal exposure to a PFAS mixture and a mixture of PFAS and organochlorines combined was associated with better sustained and selective attention during adolescence. The associations seemed to be driven by PFHxS and were not linked to exposure levels at the time of assessment.
Collapse
Affiliation(s)
- Brigitte Reimann
- Centre for Environmental Sciences, Hasselt University, 3590, Diepenbeek, Belgium
| | - Sylvie Remy
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Environmental Toxicology Unit, Mol, Belgium
| | - Gudrun Koppen
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Environmental Toxicology Unit, Mol, Belgium
| | - Greet Schoeters
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Elly Den Hond
- Provincial Institute of Hygiene, Kronenburgstraat 45, 2000, Antwerp, Belgium; Family Medicine and Population Health (FAMPOP), University of Antwerp, Gouverneur Kingsbergencentrum, Doornstraat 331, 2610, Wilrijk, Belgium
| | - Vera Nelen
- Provincial Institute of Hygiene, Kronenburgstraat 45, 2000, Antwerp, Belgium
| | - Carmen Franken
- Provincial Institute of Hygiene, Kronenburgstraat 45, 2000, Antwerp, Belgium
| | - Adrian Covaci
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Liesbeth Bruckers
- Data Science Institute, Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, 3590, Diepenbeek, Belgium
| | - Willy Baeyens
- Department of Analytical, Environmental and Geochemistry (AMGC), Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050, Elsene, Belgium
| | - Ilse Loots
- University of Antwerp, Department of Sociology (CRESC and IMDO), Sint-Jacobstraat 2, 2000, Antwerp, Belgium
| | - Nicolas van Larebeke
- Department of Analytical, Environmental and Geochemistry (AMGC), Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050, Elsene, Belgium; Study Centre for Carcinogenesis and Primary Prevention of Cancer, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Stefan Voorspoels
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Environmental Toxicology Unit, Mol, Belgium
| | - Stefaan De Henauw
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, 3590, Diepenbeek, Belgium; School of Public Health, Occupational & Environmental Medicine, Leuven University, 3000, Leuven, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, 3590, Diepenbeek, Belgium.
| |
Collapse
|
9
|
Hong X, Tao L, Guo L, Luo L, Lv J, Li R, Hu J, Gao C, Wang H, Xu DX, Cheng ZZ, Mai BX, Tang Q, Huang Y. PFASs in Cerebrospinal Fluids and Blood-CSF Barrier Permeability in Patients with Cognitive Impairment. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:5129-5138. [PMID: 38385684 DOI: 10.1021/acs.est.3c10511] [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: 02/23/2024]
Abstract
Attention has been drawn to the associations between PFASs and human cognitive decline. However, knowledge on the occurrence and permeability of PFASs in the brains of patients with cognitive impairment has not been reported. Here, we determined 30 PFASs in paired sera and cerebrospinal fluids (CSFs) from patients with cognitive impairment (n = 41) and controls without cognitive decline (n = 18). We revealed similar serum PFAS levels but different CSF PFAS levels, with lower CSF PFOA (median: 0.125 vs 0.303 ng/mL, p < 0.05), yet higher CSF PFOS (0.100 vs 0.052 ng/mL, p < 0.05) in patients than in controls. Blood-brain transfer rates also showed lower RCSF/Serum values for PFOA and higher RCSF/Serum values for PFOS in patients, implying potential heterogeneous associations with cognitive function. The RCSF/Serum values for C4-C14 perfluoroalkyl carboxylates exhibited a U-shape trend with increasing chain length. Logistic regression analyses demonstrated that CSF PFOS levels were linked to the heightened risk of cognitive impairment [odds ratio: 3.22 (1.18-11.8)] but not for serum PFOS. Toxicity inference results based on the Comparative Toxicogenomics Database suggested that PFOS in CSF may have a greater potential to impair human cognition than other PFASs. Our results contribute to a better understanding of brain PFAS exposure and its potential impact on cognitive function.
Collapse
Affiliation(s)
- Xiaowen Hong
- Department of Neurology, Anhui Provincial Hospital Affiliated to Anhui Medical University; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230032, Anhui, China
| | - Lin Tao
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Liyan Guo
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Lin Luo
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Jia Lv
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Ruonan Li
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Jiayue Hu
- Department of Hygiene Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Chang Gao
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Hua Wang
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| | - Zhao-Zhao Cheng
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230032, Anhui, China
| | - Bi-Xian Mai
- State Key Laboratory of Organic Geochemistry and Guangdong Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| | - Qiqiang Tang
- Department of Neurology, Anhui Provincial Hospital Affiliated to Anhui Medical University; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230032, Anhui, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health; Center for Big Data and Population Health of IHM; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
10
|
Zuo Q, Gao X, Fu X, Song L, Cen M, Qin S, Wu J. Association between mixed exposure to endocrine-disrupting chemicals and cognitive function in elderly Americans. Public Health 2024; 228:36-42. [PMID: 38262207 DOI: 10.1016/j.puhe.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024]
Abstract
OBJECTIVES Studies exploring the relationship between mixed exposure to endocrine-disrupting chemicals (EDCs) and cognition are limited, with even more scarce studies conducted in the elderly. The aim of this study was to investigate the association between mixed exposure to five categories of EDCs and cognition in elderly Americans. STUDY DESIGN Cross-sectional study. METHODS 727 participants from the 2011-2014 National Health and Nutrition Examination Survey were incorporated into this study, and the levels of 47 EDC metabolites were measured. Cognitive function was assessed using immediate recall test (IRT), delayed recall test (DRT), animal fluency test (AFT), and digit symbol substitution test (DSST), and all the cognitive test scores were standardized. The individual and combined effects of EDC metabolites on the cognitive function in older adults were assessed using three analytical methods. RESULTS The results showed that exposure to perfluorononanoic acid, polychlorinated biphenyl (PCB) 199, and PCB 206 was associated with the z-scores on the cognitive tests. Negative associations between mixed exposure to EDCs and the AFT and Global z-scores and a positive relationship with the DRT z-score were found in the WQS regression. The BKMR results revealed a positive trend between the mixture of EDCs and the DRT z-score. However, compared to the median, exposure to mixtures in the 45th percentile and below was associated with a decreased DRT z-score. CONCLUSIONS Mixed exposure to EDCs may adversely affect the global cognitive function in elderly individuals. Necessary measures are needed to restrict EDCs use to protect the cognitive health of older adults.
Collapse
Affiliation(s)
- Ql Zuo
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xx Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xh Fu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Ll Song
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Mq Cen
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Sf Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - J Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|
11
|
Ko MY, Park H, Chon SH, Kim YB, Cha SW, Lee BS, Hyun SA, Ka M. Differential regulations of neural activity and survival in primary cortical neurons by PFOA or PFHpA. CHEMOSPHERE 2024; 352:141379. [PMID: 38316277 DOI: 10.1016/j.chemosphere.2024.141379] [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: 09/20/2023] [Revised: 01/18/2024] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Perfluorinated compounds (PFCs), organofluoride compounds comprising carbon-fluorine and carbon-carbon bonds, are used as water and oil repellents in textiles and pharmaceutical tablets; however, they are associated with potential neurotoxic effects. Moreover, the impact of PFCs on neuronal survival, activity, and regulation within the brain remains unclear. Additionally, the mechanisms through which PFCs induce neuronal toxicity are not well-understood because of the paucity of data. This study elucidates that perfluorooctanoic acid (PFOA) and perfluoroheptanoic acid (PFHpA) exert differential effects on the survival and activity of primary cortical neurons. Although PFOA triggers apoptosis in cortical neurons, PFHpA does not exhibit this effect. Instead, PFHpA modifies dendritic spine morphogenesis and synapse formation in primary cortical neuronal cultures, additionally enhancing neural activity and synaptic transmission. This research uncovers a novel mechanism through which PFCs (PFHpA and PFOA) cause distinct alterations in dendritic spine morphogenesis and synaptic activity, shedding light on the molecular basis for the atypical behaviors noted following PFC exposure. Understanding the distinct effects of PFHpA and PFOA could guide regulatory policies on PFC usage and inform clinical approaches to mitigate their neurotoxic effects, especially in vulnerable population.
Collapse
Affiliation(s)
- Moon Yi Ko
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Heejin Park
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea; Collage of Veterinary of Medicine, Jeonbuk National University, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Sun-Hwa Chon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Yong-Bum Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Sin-Woo Cha
- Department of Nonclinical Studies, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Byoung-Seok Lee
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| | - Sung-Ae Hyun
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| | - Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| |
Collapse
|
12
|
Ford LC, Lin HC, Tsai HHD, Zhou YH, Wright FA, Sedykh A, Shah RR, Chiu WA, Rusyn I. Hazard and risk characterization of 56 structurally diverse PFAS using a targeted battery of broad coverage assays using six human cell types. Toxicology 2024; 503:153763. [PMID: 38423244 PMCID: PMC11214689 DOI: 10.1016/j.tox.2024.153763] [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: 01/18/2024] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
Per- and poly-fluoroalkyl substances (PFAS) are extensively used in commerce leading to their prevalence in the environment. Due to their chemical stability, PFAS are considered to be persistent and bioaccumulative; they are frequently detected in both the environment and humans. Because of this, PFAS as a class (composed of hundreds to thousands of chemicals) are contaminants of very high concern. Little information is available for the vast majority of PFAS, and regulatory agencies lack safety data to determine whether exposure limits or restrictions are needed. Cell-based assays are a pragmatic approach to inform decision-makers on potential health hazards; therefore, we hypothesized that a targeted battery of human in vitro assays can be used to determine whether there are structure-bioactivity relationships for PFAS, and to characterize potential risks by comparing bioactivity (points of departure) to exposure estimates. We tested 56 PFAS from 8 structure-based subclasses in concentration response (0.1-100 μM) using six human cell types selected from target organs with suggested adverse effects of PFAS - human induced pluripotent stem cell (iPSC)-derived hepatocytes, neurons, and cardiomyocytes, primary human hepatocytes, endothelial and HepG2 cells. While many compounds were without effect; certain PFAS demonstrated cell-specific activity highlighting the necessity of using a compendium of in vitro models to identify potential hazards. No class-specific groupings were evident except for some chain length- and structure-related trends. In addition, margins of exposure (MOE) were derived using empirical and predicted exposure data. Conservative MOE calculations showed that most tested PFAS had a MOE in the 1-100 range; ∼20% of PFAS had MOE<1, providing tiered priorities for further studies. Overall, we show that a compendium of human cell-based models can be used to derive bioactivity estimates for a range of PFAS, enabling comparisons with human biomonitoring data. Furthermore, we emphasize that establishing structure-bioactivity relationships may be challenging for the tested PFAS.
Collapse
Affiliation(s)
- Lucie C Ford
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Hsing-Chieh Lin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Han-Hsuan D Tsai
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Yi-Hui Zhou
- Department of Biological Sciences and Statistics, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Fred A Wright
- Department of Biological Sciences and Statistics, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | | | | | - Weihsueh A Chiu
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
13
|
Di Credico A, Weiss A, Corsini M, Gaggi G, Ghinassi B, Wilbertz JH, Di Baldassarre A. Machine learning identifies phenotypic profile alterations of human dopaminergic neurons exposed to bisphenols and perfluoroalkyls. Sci Rep 2023; 13:21907. [PMID: 38081991 PMCID: PMC10713827 DOI: 10.1038/s41598-023-49364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by the loss of midbrain dopaminergic neurons. Endocrine disrupting chemicals (EDCs) are active substances that interfere with hormonal signaling. Among EDCs, bisphenols (BPs) and perfluoroalkyls (PFs) are chemicals leached from plastics and other household products, and humans are unavoidably exposed to these xenobiotics. Data from animal studies suggest that EDCs exposure may play a role in PD, but data about the effect of BPs and PFs on human models of the nervous system are lacking. Previous studies demonstrated that machine learning (ML) applied to microscopy data can classify different cell phenotypes based on image features. In this study, the effect of BPs and PFs at different concentrations within the real-life exposure range (0.01, 0.1, 1, and 2 µM) on the phenotypic profile of human stem cell-derived midbrain dopaminergic neurons (mDANs) was analyzed. Cells exposed for 72 h to the xenobiotics were stained with neuronal markers and evaluated using high content microscopy yielding 126 different phenotypic features. Three different ML models (LDA, XGBoost and LightGBM) were trained to classify EDC-treated versus control mDANs. EDC treated mDANs were identified with high accuracies (0.88-0.96). Assessment of the phenotypic feature contribution to the classification showed that EDCs induced a significant increase of alpha-synuclein (αSyn) and tyrosine hydroxylase (TH) staining intensity within the neurons. Moreover, microtubule-associated protein 2 (MAP2) neurite length and branching were significantly diminished in treated neurons. Our study shows that human mDANs are adversely impacted by exposure to EDCs, causing their phenotype to shift and exhibit more characteristics of PD. Importantly, ML-supported high-content imaging can identify concrete but subtle subcellular phenotypic changes that can be easily overlooked by visual inspection alone and that define EDCs effects in mDANs, thus enabling further pathological characterization in the future.
Collapse
Affiliation(s)
- Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | | | - Massimo Corsini
- Dipartimento Di Neuroscienze Umane, "Sapienza" University of Rome, Chieti, Italy
| | - Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | | | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| |
Collapse
|
14
|
Paquette SE, Martin NR, Rodd A, Manz KE, Allen E, Camarillo M, Weller HI, Pennell K, Plavicki JS. Evaluation of Neural Regulation and Microglial Responses to Brain Injury in Larval Zebrafish Exposed to Perfluorooctane Sulfonate. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:117008. [PMID: 37966802 PMCID: PMC10650473 DOI: 10.1289/ehp12861] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are biopersistent, global pollutants. Although some in vitro and epidemiological studies have explored the neurotoxic potential of perfluorooctane sulfonate (PFOS), a prevalent PFAS congener, it is unknown how developmental PFOS exposure affects neuronal signaling, microglia development, and microglial-neuron communication. OBJECTIVES We sought to determine the extent to which PFOS exposure disrupts brain health, neuronal activity, and microglia-neuron communication during development. In addition, although PFOS impairs humoral immunity, its impact on innate immune cells, including resident microglia, is unclear. As such, we investigated whether microglia are cellular targets of PFOS, and, if so, whether disrupted microglial development or function could contribute to or is influenced by PFOS-induced neural dysfunction. METHODS Zebrafish were chronically exposed to either a control solution [0.1% dimethyl sulfoxide (DMSO)], 7 μ M PFOS, 14 μ M PFOS, 28 μ M PFOS, or 64 μ M perfluorooctanoic acid (PFOA). We used in vivo imaging and gene expression analysis to assess microglial populations in the developing brain and to determine shifts in the microglia state. We functionally challenged microglia state using a brain injury model and, to assess the neuronal signaling environment, performed functional neuroimaging experiments using the photoconvertible calcium indicator calcium-modulated photoactivatable ratiometric integrator (CaMPARI). These studies were paired with optogenetic manipulations of neurons and microglia, an untargeted metabolome-wide association study (MWAS), and behavioral assays. RESULTS Developmental PFOS exposure resulted in a shift away from the homeostatic microglia state, as determined by functional and morphological differences in exposed larvae, as well as up-regulation of the microglia activation gene p2ry12. PFOS-induced effects on microglia state exacerbated microglia responses to brain injury in the absence of increased cell death or inflammation. PFOS exposure also heightened neural activity, and optogenetic silencing of neurons or microglia independently was sufficient to normalize microglial responses to injury. An untargeted MWAS of larval brains revealed PFOS-exposed larvae had neurochemical signatures of excitatory-inhibitory imbalance. Behaviorally, PFOS-exposed larvae also exhibited anxiety-like thigmotaxis. To test whether the neuronal and microglial phenotypes were specific to PFOS, we exposed embryos to PFOA, a known immunotoxic PFAS. PFOA did not alter thigmotaxis, neuronal activity, or microglial responses, further supporting a role for neuronal activity as a critical modifier of microglial function following PFOS exposure. DISCUSSION Together, this study provides, to our knowledge, the first detailed account of the effects of PFOS exposure on neural cell types in the developing brain in vivo and adds neuronal hyperactivity as an important end point to assess when studying the impact of toxicant exposures on microglia function. https://doi.org/10.1289/EHP12861.
Collapse
Affiliation(s)
- Shannon E. Paquette
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Nathan R. Martin
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - April Rodd
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Katherine E. Manz
- School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Eden Allen
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Manuel Camarillo
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Hannah I. Weller
- Department of Ecology, Evolution, and Organismal Biology, Brown University, Providence, Rhode Island, USA
| | - Kurt Pennell
- School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Jessica S. Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
15
|
Neisiani AK, Mousavi MK, Soltani M, Aliomrani M. Perfluorooctanoic acid exposure and its neurodegenerative consequences in C57BL6/J mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2357-2367. [PMID: 36700988 DOI: 10.1007/s00210-023-02387-8] [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: 08/28/2022] [Accepted: 01/03/2023] [Indexed: 01/27/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a member of Per- and polyfluoroalkyl substances (PFASs), an industrial pollutant that has been produced for decades and widely used in various industries. Accumulation of this compound in the environment and body of organisms led to increased concerns about this compound. The toxic effects of PFOA on the nervous system are unknown yet. We aimed to assess the myelination and neurogenesis in brain tissue. In this study, PFOA at doses of 1, 5, 10, and 20 mg/kg were injected intraperitoneally into C57BL/6 J mice for 14 days, and the myelin content, CD4 + and CD8 + cell infiltration to brain regions were evaluated. Also, bromodeoxyuridine (BrdU) labeling was performed to compare neurogenesis among the groups. Luxol Fast Blue (LFB) staining revealed a significant decrease in myelin content in both sex at high concentrations (p < 0.001). The BrdU incorporation changes were observed in both sexes especially females which was highly related to the dose of PFOA and region of the brain. The infiltration rates of CD4 + and CD8 + cells to the brain were shown to be decreased; meanwhile the lymphocyte count was not significantly changed among groups over time and vice versa for the monocyte and neutrophils. Our results showed that PFOA had a negative impact on neurogenesis and the myelination process through the specific region of the brain depending on the dose and sex. Also, PFOA could disturb the number of CD4 + and CD8 + cells infiltrating the brain, which plays a crucial role in neurogenesis, leading to toxicity and neurological abnormalities. It seems that more research is needed to determine the exact mechanisms of PFOA neurotoxicity and its long-term behavioral consequences.
Collapse
Affiliation(s)
- Azadeh Khosravi Neisiani
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Mahboobeh Kafi Mousavi
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Masoud Soltani
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Mehdi Aliomrani
- Department of Toxicology and Pharmacology, Isfahan Pharmaceutical Science Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
16
|
Li C, Bundy JD, Tian L, Zhang R, Chen J, Kelly TN, He J. Examination of Serum Metabolome Altered by Dietary Carbohydrate, Milk Protein, and Soy Protein Interventions Identified Novel Metabolites Associated with Blood Pressure: The ProBP Trial. Mol Nutr Food Res 2023; 67:e2300044. [PMID: 37650262 PMCID: PMC10592004 DOI: 10.1002/mnfr.202300044] [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: 01/25/2023] [Revised: 05/08/2023] [Indexed: 09/01/2023]
Abstract
SCOPE This study aims to discover metabolites of dietary carbohydrate, soy and milk protein supplements and evaluate their roles in blood pressure (BP) regulation in the protein and blood pressure (ProBP), a cross-over trial. METHODS AND RESULTS Plasma metabolites are profiled at pre-trial baseline and after 8 weeks of supplementation with carbohydrate, soy protein, and milk protein, respectively, among 80 ProBP participants. After Bonferroni correction (α = 6.49 × 10-4 ), dietary interventions significantly changed 40 metabolites. Changes of erucate (22:1n9), an omega-9 fatty acid, are positively associated with systolic BP changes (Beta = 1.90, p = 6·27 × 10-4 ). This metabolite is also associated with higher odds of hypertension among 1261 participants of an independent cohort (odds ratio per unit increase = 1.34; 95% confidence interval: 1.07-1.68). High levels of acylcholines dihomo-linolenoyl-choline (p = 4.71E-04) and oleoylcholine (p = 3.48E-04) at baseline predicted larger BP lowering effects of soy protein. Increasing cheese intake during the trial, as reflected by isobutyrylglycine and isovalerylglycine, reduces the BP lowering effect of soy protein. CONCLUSIONS The study identifies molecular signatures of dietary interventions. Erucate (22:1n9) increases systolic BP. Acylcholine enhances and cheese intake reduces the BP lowering effect of soy protein supplement.
Collapse
Affiliation(s)
- Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
| | - Joshua D. Bundy
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
| | - Ling Tian
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
| | - Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
- Department of Medicine, Tulane University School of Medicine
| | - Tanika N. Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine
- Department of Medicine, Tulane University School of Medicine
| |
Collapse
|
17
|
An Z, Yang J, Xiao F, Lv J, Xing X, Liu H, Wang L, Liu Y, Zhang Z, Guo H. Hippocampal Proteomics Reveals the Role of Glutamatergic Synapse Activation in the Depression Induced by Perfluorooctane Sulfonate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7866-7877. [PMID: 37191230 DOI: 10.1021/acs.jafc.3c01344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS), a new type of persistent organic pollutant in the environment of water, has drawn significant attention in recent years due to its widespread prevalence and high toxicity. Neurotoxicity is regarded as one of the major toxic effects of PFOS, while research studies on PFOS-induced depression and the underlying mechanisms remain scarce. In this study, behavioral tests revealed the depressive-like behaviors in PFOS-exposed male mice. Neuron damages including pyknosis and staining deepening were identified through hematoxylin and eosin staining. Then, we noticed the elevation of glutamate and proline levels as well as the decline of glutamine and tryptophan levels. Proteomics analysis identified 105 differentially expressed proteins that change in a dose-dependent manner and revealed that PFOS exposure activated the glutamatergic synapse signaling pathway, which were further confirmed by Western blot, and the data were consistent with the findings of the proteomics analysis. Additionally, the downstream signaling cyclic AMP-responsive element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) and synaptic plasticity-related postsynaptic density protein 95, synaptophysin, were downregulated. Our results highlight that PFOS exposure may inhibit the synaptic plasticity of the hippocampus via glutamatergic synapse and the CREB/BDNF signaling pathway to cause depressive-like behaviors in male mice.
Collapse
Affiliation(s)
- Ziwen An
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jing Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Fang Xiao
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Junli Lv
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoqing Xing
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang 050017, China
| | - Heqiong Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Lei Wang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Yi Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhanchi Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Huicai Guo
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| |
Collapse
|
18
|
Hartmann J, Henschel N, Bartmann K, Dönmez A, Brockerhoff G, Koch K, Fritsche E. Molecular and Functional Characterization of Different BrainSphere Models for Use in Neurotoxicity Testing on Microelectrode Arrays. Cells 2023; 12:cells12091270. [PMID: 37174670 PMCID: PMC10177384 DOI: 10.3390/cells12091270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The currently accepted methods for neurotoxicity (NT) testing rely on animal studies. However, high costs and low testing throughput hinder their application for large numbers of chemicals. To overcome these limitations, in vitro methods are currently being developed based on human-induced pluripotent stem cells (hiPSC) that allow higher testing throughput at lower costs. We applied six different protocols to generate 3D BrainSphere models for acute NT evaluation. These include three different media for 2D neural induction and two media for subsequent 3D differentiation resulting in self-organized, organotypic neuron/astrocyte microtissues. All induction protocols yielded nearly 100% NESTIN-positive hiPSC-derived neural progenitor cells (hiNPCs), though with different gene expression profiles concerning regional patterning. Moreover, gene expression and immunocytochemistry analyses revealed that the choice of media determines neural differentiation patterns. On the functional level, BrainSpheres exhibited different levels of electrical activity on microelectrode arrays (MEA). Spike sorting allowed BrainSphere functional characterization with the mixed cultures consisting of GABAergic, glutamatergic, dopaminergic, serotonergic, and cholinergic neurons. A test method for acute NT testing, the human multi-neurotransmitter receptor (hMNR) assay, was proposed to apply such MEA-based spike sorting. These models are promising tools not only in toxicology but also for drug development and disease modeling.
Collapse
Affiliation(s)
- Julia Hartmann
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | - Noah Henschel
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | - Kristina Bartmann
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
| | - Arif Dönmez
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
| | - Gabriele Brockerhoff
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | - Katharina Koch
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
| | - Ellen Fritsche
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
- Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
19
|
Pini L, Salvalaggio A, Wennberg AM, Dimakou A, Matteoli M, Corbetta M. The pollutome-connectome axis: a putative mechanism to explain pollution effects on neurodegeneration. Ageing Res Rev 2023; 86:101867. [PMID: 36720351 DOI: 10.1016/j.arr.2023.101867] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy.
| | | | - Alexandra M Wennberg
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Dimakou
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy
| | - Michela Matteoli
- Neuro Center, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milano, Italy; CNR Institute of Neuroscience, Milano, Italy
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy
| |
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: 24] [Impact Index Per Article: 24.0] [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
|
Bose R, Spulber S, Ceccatelli S. The Threat Posed by Environmental Contaminants on Neurodevelopment: What Can We Learn from Neural Stem Cells? Int J Mol Sci 2023; 24:4338. [PMID: 36901772 PMCID: PMC10002364 DOI: 10.3390/ijms24054338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Exposure to chemicals may pose a greater risk to vulnerable groups, including pregnant women, fetuses, and children, that may lead to diseases linked to the toxicants' target organs. Among chemical contaminants, methylmercury (MeHg), present in aquatic food, is one of the most harmful to the developing nervous system depending on time and level of exposure. Moreover, certain man-made PFAS, such as PFOS and PFOA, used in commercial and industrial products including liquid repellants for paper, packaging, textile, leather, and carpets, are developmental neurotoxicants. There is vast knowledge about the detrimental neurotoxic effects induced by high levels of exposure to these chemicals. Less is known about the consequences that low-level exposures may have on neurodevelopment, although an increasing number of studies link neurotoxic chemical exposures to neurodevelopmental disorders. Still, the mechanisms of toxicity are not identified. Here we review in vitro mechanistic studies using neural stem cells (NSCs) from rodents and humans to dissect the cellular and molecular processes changed by exposure to environmentally relevant levels of MeHg or PFOS/PFOA. All studies show that even low concentrations dysregulate critical neurodevelopmental steps supporting the idea that neurotoxic chemicals may play a role in the onset of neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
22
|
Choo SS, Keever JY, Brown J, Strickland JD, Shafer TJ. Assaying Spontaneous Network Activity and Cellular Viability Using Multi-Well Microelectrode Arrays. Methods Mol Biol 2023; 2644:133-154. [PMID: 37142920 DOI: 10.1007/978-1-0716-3052-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Microelectrode array (MEA) technology is a neurophysiological method that allows for the measurement of spontaneous or evoked neural activity to determine chemical effects thereon. Following assessment of compound effects on multiple endpoints that evaluate network function, a cell viability endpoint in the same well is determined using a multiplexed approach. Recently, it has become possible to measure electrical impedance of cells attached to the electrodes, where greater impedance indicates greater number of cells attached. This would allow rapid and repeated assessments of cell health as the neural network develops in longer exposure assays without impacting cell health. Typically, the lactate dehydrogenase (LDH) assay for cytotoxity and CellTiter-Blue® (CTB) assay for cell viability are only performed at the end of the chemical exposure period because these assays involve lysing of the cells. Procedures describing the multiplexed methods in acute and network formation screening are included in this chapter.
Collapse
Affiliation(s)
- Seline S Choo
- Oak Ridge Institute for Science and Engineering, Oak Ridge, TN, USA
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jackson Y Keever
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
- Oak Ridge Associated Universities Student Contractor, Oak Ridge, TN, USA
| | - Jasmine Brown
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Jenna D Strickland
- Axion Biosystems, Atlanta, GA, USA
- LabCorp Drug Development, Madison, WI, USA
| | - Timothy J Shafer
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
23
|
Seizurogenic effect of perfluorooctane sulfonate in zebrafish larvae. Neurotoxicology 2022; 93:257-264. [DOI: 10.1016/j.neuro.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
|
24
|
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: 9] [Impact Index Per Article: 4.5] [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
|
25
|
Jane L Espartero L, Yamada M, Ford J, Owens G, Prow T, Juhasz A. Health-related toxicity of emerging per- and polyfluoroalkyl substances: Comparison to legacy PFOS and PFOA. ENVIRONMENTAL RESEARCH 2022; 212:113431. [PMID: 35569538 DOI: 10.1016/j.envres.2022.113431] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are highly persistent, manufactured chemicals used in various manufacturing processes and found in numerous commercial products. With over 9000 compounds belonging to this chemical class, there is increasing concern regarding human exposure to these compounds due to their persistent, bioaccumulative, and toxic nature. Human exposure to PFAS may occur from a variety of exposure sources, including, air, food, indoor dust, soil, water, from the transfer of PFAS from non-stick wrappers to food, use of cosmetics, and other personal care products. This critical review presents recent research on the health-related impacts of PFAS exposure, highlighting compounds other than Perfluorooctanoic acid (PFOA) and Perfluoroctane sulfonate (PFOS) that cause adverse health effects, updates the current state of knowledge on PFAS toxicity, and, where possible, elucidates cause-and-effect relationships. Recent reviews identified that exposure to PFAS was associated with adverse health impacts on female and male fertility, metabolism in pregnancy, endocrine function including pancreatic dysfunction and risk of developing Type 2 diabetes, lipid metabolism and risk of childhood adiposity, hepatic and renal function, immune function, cardiovascular health (atherosclerosis), bone health including risk for dental cavities, osteoporosis, and vitamin D deficiency, neurological function, and risk of developing breast cancer. However, while cause-and-effect relationships for many of these outcomes were not able to be clearly elucidated, it was identified that 1) the evidence derived from both animal models and humans suggested that PFAS may exert harmful impacts on both animals and humans, however extrapolating data from animal to human studies was complicated due to differences in exposure/elimination kinetics, 2) PFAS precursor kinetics and toxicity mechanism data are still limited despite ongoing exposures, and 3) studies in humans, which provide contrasting results require further investigation of the long-term-exposed population to better evaluate the biological toxicity of chronic exposure to PFAS.
Collapse
Affiliation(s)
- Lore Jane L Espartero
- Future Industries Institute (FII), University of South Australia, Mawson Lakes, South Australia, Australia
| | - Miko Yamada
- Future Industries Institute (FII), University of South Australia, Mawson Lakes, South Australia, Australia
| | - Judith Ford
- University of Sydney, New South Wales, United Kingdom
| | - Gary Owens
- Future Industries Institute (FII), University of South Australia, Mawson Lakes, South Australia, Australia
| | - Tarl Prow
- Future Industries Institute (FII), University of South Australia, Mawson Lakes, South Australia, Australia; Skin Research Centre, York Biomedical Research Institute, Hull York Medical School, University of York, United Kingdom
| | - Albert Juhasz
- Future Industries Institute (FII), University of South Australia, Mawson Lakes, South Australia, Australia.
| |
Collapse
|
26
|
Obeng-Gyasi E. Factors associated with elevated Per- and Polyfluoroalkyl substances serum levels in older adults. AGING AND HEALTH RESEARCH 2022; 2. [DOI: 10.1016/j.ahr.2022.100086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
27
|
Brown-Leung JM, Cannon JR. Neurotransmission Targets of Per- and Polyfluoroalkyl Substance Neurotoxicity: Mechanisms and Potential Implications for Adverse Neurological Outcomes. Chem Res Toxicol 2022; 35:1312-1333. [PMID: 35921496 PMCID: PMC10446502 DOI: 10.1021/acs.chemrestox.2c00072] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a group of persistent environmental pollutants that are ubiquitously found in the environment and virtually in all living organisms, including humans. PFAS cross the blood-brain barrier and accumulate in the brain. Thus, PFAS are a likely risk for neurotoxicity. Studies that measured PFAS levels in the brains of humans, polar bears, and rats have demonstrated that some areas of the brain accumulate greater amounts of PFAS. Moreover, in humans, there is evidence that PFAS exposure is associated with attention-deficit/hyperactivity disorder (ADHD) in children and an increased cause of death from Parkinson's disease and Alzheimer's disease in elderly populations. Given possible links to neurological disease, critical analyses of possible mechanisms of neurotoxic action are necessary to advance the field. This paper critically reviews studies that investigated potential mechanistic causes for neurotoxicity including (1) a change in neurotransmitter levels, (2) dysfunction of synaptic calcium homeostasis, and (3) alteration of synaptic and neuronal protein expression and function. We found growing evidence that PFAS exposure causes neurotoxicity through the disruption of neurotransmission, particularly the dopamine and glutamate systems, which are implicated in age-related psychiatric illnesses and neurodegenerative diseases. Evaluated research has shown there are highly reproduced increased glutamate levels in the hippocampus and catecholamine levels in the hypothalamus and decreased dopamine in the whole brain after PFAS exposure. There are significant gaps in the literature relative to the assessment of the nigrostriatal system (striatum and ventral midbrain) among other regions associated with PFAS-associated neurologic dysfunction observed in humans. In conclusion, evidence suggests that PFAS may be neurotoxic and associated with chronic and age-related psychiatric illnesses and neurodegenerative diseases. Thus, it is imperative that future mechanistic studies assess the impact of PFAS and PFAS mixtures on the mechanism of neurotransmission and the consequential functional effects.
Collapse
Affiliation(s)
- Josephine M Brown-Leung
- School of Health Sciences, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
28
|
Luo F, Chen Q, Yu G, Huo X, Wang H, Nian M, Tian Y, Xu J, Zhang J, Zhang J. Exposure to perfluoroalkyl substances and neurodevelopment in 2-year-old children: A prospective cohort study. ENVIRONMENT INTERNATIONAL 2022; 166:107384. [PMID: 35834943 DOI: 10.1016/j.envint.2022.107384] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/28/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Humans are widely exposed to per- and polyfluoroalkyl substances (PFAS). As fetal stage is a critical window for neurodevelopment, it is important to know if in utero exposure to PFAS affects fetal neurodevelopment. However, previous human studies are both limited and inconsistent. OBJECTIVES To investigate the relationship between PFAS exposure during early pregnancy and the neurodevelopmental status at 2 years of age in a prospective cohort study. METHODS We measured 10 PFAS in maternal plasma samples collected prior to 16 weeks of gestation in the Shanghai Birth Cohort Study between 2013 and 2016. Childhood neurodevelopment was assessed at 2 years of age using the Bayley Scales of Infant and Toddler Development-Third Edition (BSID-III). Associations with domain specific scores for individual PFAS were assessed by multiple linear regression and binary logistic regression when scores were dichotomized. Quantile-based g-computation was used to estimate the joint effects of PFAS mixture. RESULTS A total of 2257 mother-child pairs who had both PFAS and BSID measurements were included in our analyses. The means and standard deviations of comprehensive scores were 115 ± 22, 96 ± 15 and 108 ± 15 for cognition, language, and motor, respectively. In multiple linear regressions, we observed significant negative associations of perfluorooctane sulfonate (PFOS), perfluorononanoic acid (PFNA), perfluorodecanoic acid (PFDeA), perfluoroundecanoic acid (PFUnDA) with cognitive scores; PFNA, PFDeA, PFUnDA and perfluorohexanesulfonate (PFHxS) negatively with language scores; and PFNA and PFUnDA negatively with motor scores. Quantile-based g-computation showed that PFAS mixture was significantly associated with decreased cognitive and language scores, with an estimated β of -2.1 [95% confidence interval (CI): -3.5, -0.7)] and -2.0 (95% CI: -2.9, -1.0) per one quartile increase in PFAS mixture for cognitive and language domains, respectively. PFAS mixture was associated with increased odds of low cognition (adjusted odds ratio [OR] = 1.3, 95% CI:1.0, 1.6) and language scores (OR = 1.2, 95% CI: 1.1, 1.3). CONCLUSIONS PFAS exposure during early pregnancy was significantly associated with the adverse neurodevelopmental status at 2 years of age, which raises a serious public health concern.
Collapse
Affiliation(s)
- Fei Luo
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200052, China
| | - Qian Chen
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Guoqi Yu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Xiaona Huo
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China; Obstetrics and Gynecology Department, International Peace Maternity and Child Health Hospital of China, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Hui Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Min Nian
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200052, China
| | - Ying Tian
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200052, China
| | - Jian Xu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingsong Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China; Department of Medical Psychology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200052, China.
| |
Collapse
|
29
|
Crofton KM, Bassan A, Behl M, Chushak YG, Fritsche E, Gearhart JM, Marty MS, Mumtaz M, Pavan M, Ruiz P, Sachana M, Selvam R, Shafer TJ, Stavitskaya L, Szabo DT, Szabo ST, Tice RR, Wilson D, Woolley D, Myatt GJ. Current status and future directions for a neurotoxicity hazard assessment framework that integrates in silico approaches. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 22:100223. [PMID: 35844258 PMCID: PMC9281386 DOI: 10.1016/j.comtox.2022.100223] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Neurotoxicology is the study of adverse effects on the structure or function of the developing or mature adult nervous system following exposure to chemical, biological, or physical agents. The development of more informative alternative methods to assess developmental (DNT) and adult (NT) neurotoxicity induced by xenobiotics is critically needed. The use of such alternative methods including in silico approaches that predict DNT or NT from chemical structure (e.g., statistical-based and expert rule-based systems) is ideally based on a comprehensive understanding of the relevant biological mechanisms. This paper discusses known mechanisms alongside the current state of the art in DNT/NT testing. In silico approaches available today that support the assessment of neurotoxicity based on knowledge of chemical structure are reviewed, and a conceptual framework for the integration of in silico methods with experimental information is presented. Establishing this framework is essential for the development of protocols, namely standardized approaches, to ensure that assessments of NT and DNT based on chemical structures are generated in a transparent, consistent, and defendable manner.
Collapse
Affiliation(s)
| | - Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Mamta Behl
- Division of the National Toxicology Program, National
Institutes of Environmental Health Sciences, Durham, NC 27709, USA
| | - Yaroslav G. Chushak
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental
Medicine & Medical Faculty Heinrich-Heine-University, Düsseldorf,
Germany
| | - Jeffery M. Gearhart
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | | | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Patricia Ruiz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Magdalini Sachana
- Environment Health and Safety Division, Environment
Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775
Paris Cedex 16, France
| | - Rajamani Selvam
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | - Timothy J. Shafer
- Biomolecular and Computational Toxicology Division, Center
for Computational Toxicology and Exposure, US EPA, Research Triangle Park, NC,
USA
| | - Lidiya Stavitskaya
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | | | | | | | - Dan Wilson
- The Dow Chemical Company, Midland, MI 48667, USA
| | | | - Glenn J. Myatt
- Instem, Columbus, OH 43215, USA
- Corresponding author.
(G.J. Myatt)
| |
Collapse
|
30
|
PFAS Molecules: A Major Concern for the Human Health and the Environment. TOXICS 2022; 10:toxics10020044. [PMID: 35202231 PMCID: PMC8878656 DOI: 10.3390/toxics10020044] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 01/09/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a group of over 4700 heterogeneous compounds with amphipathic properties and exceptional stability to chemical and thermal degradation. The unique properties of PFAS compounds has been exploited for almost 60 years and has largely contributed to their wide applicability over a vast range of industrial, professional and non-professional uses. However, increasing evidence indicate that these compounds represent also a serious concern for both wildlife and human health as a result of their ubiquitous distribution, their extreme persistence and their bioaccumulative potential. In light of the adverse effects that have been already documented in biota and human populations or that might occur in absence of prompt interventions, the competent authorities in matter of health and environment protection, the industries as well as scientists are cooperating to identify the most appropriate regulatory measures, substitution plans and remediation technologies to mitigate PFAS impacts. In this review, starting from PFAS chemistry, uses and environmental fate, we summarize the current knowledge on PFAS occurrence in different environmental media and their effects on living organisms, with a particular emphasis on humans. Also, we describe present and provisional legislative measures in the European Union framework strategy to regulate PFAS manufacture, import and use as well as some of the most promising treatment technologies designed to remediate PFAS contamination in different environmental compartments.
Collapse
|
31
|
Chang CW, Evans MD, Yu X, Yu GQ, Mucke L. Tau reduction affects excitatory and inhibitory neurons differently, reduces excitation/inhibition ratios, and counteracts network hypersynchrony. Cell Rep 2021; 37:109855. [PMID: 34686344 PMCID: PMC8648275 DOI: 10.1016/j.celrep.2021.109855] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/04/2021] [Accepted: 09/27/2021] [Indexed: 11/27/2022] Open
Abstract
The protein tau has been implicated in many brain disorders. In animal models, tau reduction suppresses epileptogenesis of diverse causes and ameliorates synaptic and behavioral abnormalities in various conditions associated with excessive excitation-inhibition (E/I) ratios. However, the underlying mechanisms are unknown. Global genetic ablation of tau in mice reduces the action potential (AP) firing and E/I ratio of pyramidal cells in acute cortical slices without affecting the excitability of these cells. Tau ablation reduces the excitatory inputs to inhibitory neurons, increases the excitability of these cells, and structurally alters their axon initial segments (AISs). In primary neuronal cultures subjected to prolonged overstimulation, tau ablation diminishes the homeostatic response of AISs in inhibitory neurons, promotes inhibition, and suppresses hypersynchrony. Together, these differential alterations in excitatory and inhibitory neurons help explain how tau reduction prevents network hypersynchrony and counteracts brain disorders causing abnormally increased E/I ratios.
Collapse
Affiliation(s)
- Che-Wei Chang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mark D Evans
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Xinxing Yu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
32
|
Pierozan P, Karlsson O. Differential susceptibility of rat primary neurons and neural stem cells to PFOS and PFOA toxicity. Toxicol Lett 2021; 349:61-68. [PMID: 34126183 DOI: 10.1016/j.toxlet.2021.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/30/2022]
Abstract
Per and polifluorinated substances (PFAS) are ubiquitous and persistent contaminants. Studies have indicated that fetuses and infants can be exposed to these chemicals in utero and through breastfeeding. Despite this, limited data about their effects on brain development are available. Here, we compared the effects of perfluoroctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) exposure in rat primary neurons and neural stem cells (NSC). Treatment with 1-250 μM of either of these compounds caused no effects on cell viability or proliferation in primary neurons, while PFOS exposure increased the NSC proliferation already at the lowest concentration tested (1-100 μM). Further analysis showed that both PFOS and PFOA caused morphological alterations of NSC-derived neurons. The neurons derived from NSC treated with either of the PFAS demonstrated a decrease in cell body area. Exposure to 1 and 10 μM PFOA also affected the neurite network and caused an increase in the number of processes and branches per cell. None of the PFAS caused morphological alterations in primary neurons. These data suggest that NSC, mimicking the immature brain, is clearly more susceptible to PFOS and PFOA exposure than the primary neurons. The PFAS-induced alterations in NSC may be related to neurobehavioral alterations observed in rodents developmentally exposed to these compounds, and show the importance to consider the effects of these compounds on human brain development and disease.
Collapse
Affiliation(s)
- Paula Pierozan
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm, 114 18, Sweden.
| | - Oskar Karlsson
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm, 114 18, Sweden
| |
Collapse
|
33
|
O'Shaughnessy KL, Fischer F, Zenclussen AC. Perinatal exposure to endocrine disrupting chemicals and neurodevelopment: How articles of daily use influence the development of our children. Best Pract Res Clin Endocrinol Metab 2021; 35:101568. [PMID: 34565681 PMCID: PMC10111869 DOI: 10.1016/j.beem.2021.101568] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Substances that interfere with the body's hormonal balance or their function are called endocrine disrupting chemicals (EDCs). Many EDCs are ubiquitous in the environment and are an unavoidable aspect of daily life, including during early embryogenesis. Developmental exposure to these chemicals is of critical relevance, as EDCs can permanently alter developmental programs, including those that pattern and wire the brain. Of emerging interest is how these chemicals may also affect the immune response, given the cross-talk between the endocrine and immune systems. As brain development is strongly dependent on hormones including thyroid, androgens, and estrogens, and can also be affected by immunomodulation, this complicated interplay may have long-lasting neurodevelopmental consequences. This review focuses on data available from human cohorts, in vivo models, and in vitro assays regarding the impact of EDCs after a gestational and/or lactational exposure, and how they may impact the immune system and/or neurodevelopment.
Collapse
Affiliation(s)
- Katherine L O'Shaughnessy
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, NC, USA.
| | - Florence Fischer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany.
| |
Collapse
|
34
|
Yu T, Zhou G, Cai Z, Liang W, Du Y, Wang W. Behavioral effects of early-life exposure to perfluorooctanoic acid might synthetically link to multiple aspects of dopaminergic neuron development and dopamine functions in zebrafish larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 238:105926. [PMID: 34340000 DOI: 10.1016/j.aquatox.2021.105926] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 05/27/2023]
Abstract
Perfluorooctanoic acid (PFOA) is known as an environmental endocrine disruptor and has developmental neurotoxicity that could be associated with behavior changes in human and animal studies. Previous studies have shown that PFOA might affect the dopaminergic nervous system. However, the mode of action underlying the effects of PFOA remains poorly understood. Our study used zebrafish as an animal model to investigate the effects of early-life PFOA exposure on dopaminergic neuron development and dopamine functions in zebrafish larvae. Zebrafish fertilized eggs were exposed to different concentrations of PFOA (0, 10, 100, 1000 μg/L). After exposure to PFOA for 7 days, the locomotor activity of zebrafish was decreased; the mRNA levels of nuclear receptor subfamily 4 group a member 2b (nr4a2b), paired box 2 and 5 (pax2, pax5), tyrosine hydroxylase 1/2 (th1/th2) and dopamine transporter (dat) were increased; mRNA and protein level of mesencephalic astrocyte-derived neurotrophic factor (manf) were decreased. Neural cell proliferation in the preoptic area of hypothalamus was increased. In conclusion, dopaminergic neuron development might be one of the targets of early-life PFOA exposure. The neurobehavior changes induced by PFOA exposure might link to multiple aspects of dopaminergic neuron development and dopamine functions in zebrafish larvae.
Collapse
Affiliation(s)
- Tingting Yu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200292, China
| | - Guangdi Zhou
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200292, China
| | - Zhenzhen Cai
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200292, China
| | - Wei Liang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200292, China
| | - Yatao Du
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200292, China.
| | - Weiye Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200292, China.
| |
Collapse
|
35
|
Environmentally relevant developmental methylmercury exposures alter neuronal differentiation in a human-induced pluripotent stem cell model. Food Chem Toxicol 2021; 152:112178. [PMID: 33831500 DOI: 10.1016/j.fct.2021.112178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Developmental methylmercury (MeHg) exposure selectively targets the cerebral and cerebellar cortices, as seen by disruption of cytoarchitecture and glutamatergic (GLUergic) neuron hypoplasia. To begin to understand the mechanisms of this loss of GLUergic neurons, we aimed to develop a model of developmental MeHg neurotoxicity in human-induced pluripotent stem cells differentiating into cortical GLUergic neurons. Three dosing paradigms at 0.1 μM and 1.0 μM MeHg, which span different stages of neurodevelopment and reflect toxicologically relevant accumulation levels seen in human studies and mammalian models, were established. With these exposure paradigms, no changes were seen in commonly studied endpoints of MeHg toxicity, including viability, proliferation, and glutathione levels. However, MeHg exposure induced changes in mitochondrial respiration and glycolysis and in markers of neuronal differentiation. Our novel data suggests that GLUergic neuron hypoplasia seen with MeHg toxicity may be due to the partial inhibition of neuronal differentiation, given the increased expression of the early dorsal forebrain marker FOXG1 and corresponding decrease in expression on neuronal markers MAP2 and DCX and the deep layer cortical neuronal marker TBR1. Future studies should examine the persistent and latent functional effects of this MeHg-induced disruption of neuronal differentiation as well as transcriptomic and metabolomic alterations that may mediate MeHg toxicity.
Collapse
|
36
|
Ronchi S, Buccino AP, Prack G, Kumar SS, Schröter M, Fiscella M, Hierlemann A. Electrophysiological Phenotype Characterization of Human iPSC-Derived Neuronal Cell Lines by Means of High-Density Microelectrode Arrays. Adv Biol (Weinh) 2021; 5:e2000223. [PMID: 33729694 PMCID: PMC7610355 DOI: 10.1002/adbi.202000223] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/30/2020] [Indexed: 12/11/2022]
Abstract
Recent advances in the field of cellular reprogramming have opened a route to studying the fundamental mechanisms underlying common neurological disorders. High-density microelectrode-arrays (HD-MEAs) provide unprecedented means to study neuronal physiology at different scales, ranging from network through single-neuron to subcellular features. In this work, HD-MEAs are used in vitro to characterize and compare human induced-pluripotent-stem-cell-derived dopaminergic and motor neurons, including isogenic neuronal lines modeling Parkinson's disease and amyotrophic lateral sclerosis. Reproducible electrophysiological network, single-cell and subcellular metrics are used for phenotype characterization and drug testing. Metrics, such as burst shape and axonal velocity, enable the distinction of healthy and diseased neurons. The HD-MEA metrics can also be used to detect the effects of dosing the drug retigabine to human motor neurons. Finally, it is shown that the ability to detect drug effects and the observed culture-to-culture variability critically depend on the number of available recording electrodes.
Collapse
Affiliation(s)
- Silvia Ronchi
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Alessio Paolo Buccino
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Gustavo Prack
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Sreedhar Saseendran Kumar
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Manuel Schröter
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Michele Fiscella
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
- MaxWell Biosystems AG, Albisriederstrasse 253, Zürich, 8047, Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| |
Collapse
|
37
|
Davidsen N, Lauvås AJ, Myhre O, Ropstad E, Carpi D, Gyves EMD, Berntsen HF, Dirven H, Paulsen RE, Bal-Price A, Pistollato F. Exposure to human relevant mixtures of halogenated persistent organic pollutants (POPs) alters neurodevelopmental processes in human neural stem cells undergoing differentiation. Reprod Toxicol 2021; 100:17-34. [PMID: 33333158 PMCID: PMC7992035 DOI: 10.1016/j.reprotox.2020.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Halogenated persistent organic pollutants (POPs) like perfluorinated alkylated substances (PFASs), brominated flame retardants (BFRs), organochlorine pesticides and polychlorinated biphenyls (PCBs) are known to cause cancer, immunotoxicity, neurotoxicity and interfere with reproduction and development. Concerns have been raised about the impact of POPs upon brain development and possibly neurodevelopmental disorders. The developing brain is a particularly vulnerable organ due to dynamic and complex neurodevelopmental processes occurring early in life. However, very few studies have reported on the effects of POP mixtures at human relevant exposures, and their impact on key neurodevelopmental processes using human in vitro test systems. Aiming to reduce this knowledge gap, we exposed mixed neuronal/glial cultures differentiated from neural stem cells (NSCs) derived from human induced pluripotent stem cells (hiPSCs) to reconstructed mixtures of 29 different POPs using concentrations comparable to Scandinavian human blood levels. Effects of the POP mixtures on neuronal proliferation, differentiation and synaptogenesis were evaluated using in vitro assays anchored to common key events identified in the existing developmental neurotoxicity (DNT) adverse outcome pathways (AOPs). The present study showed that mixtures of POPs (in particular brominated and chlorinated compounds) at human relevant concentrations increased proliferation of NSCs and decreased synapse number. Based on a mathematical modelling, synaptogenesis and neurite outgrowth seem to be the most sensitive DNT in vitro endpoints. Our results indicate that prenatal exposure to POPs may affect human brain development, potentially contributing to recently observed learning and memory deficits in children.
Collapse
Affiliation(s)
- Nichlas Davidsen
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna Jacobsen Lauvås
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Oddvar Myhre
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Donatella Carpi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway; National Institute of Occupational Health, Oslo, Norway
| | - Hubert Dirven
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Ragnhild E Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | |
Collapse
|
38
|
Kasteel EEJ, Westerink RHS. Refining in vitro and in silico neurotoxicity approaches by accounting for interspecies and interindividual differences in toxicodynamics. Expert Opin Drug Metab Toxicol 2021; 17:1007-1017. [PMID: 33586568 DOI: 10.1080/17425255.2021.1885647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The process of chemical risk assessment traditionally relies on animal experiments and associated default uncertainty factors to account for interspecies and interindividual differences. To work toward a more precise and personalized risk assessment, these uncertainty factors should be refined and replaced by chemical-specific adjustment factors (CSAFs). AREAS COVERED This concise review discusses alternative (in vitro/in silico) approaches that can be used to assess interspecies and interindividual differences in toxicodynamics, ranging from targeted to more integrated approaches. Although data are available on interspecies differences, the increasing use of human-induced pluripotent stem cell (hiPSC)-derived neurons may provide opportunities to also assess interindividual variability in neurotoxicity. More integrated approaches, like adverse outcome pathways (AOPs) can provide a more quantitative understanding of the toxicodynamics of a chemical. EXPERT OPINION To improve chemical risk assessment, refinement of uncertainty factors is crucial. In vitro and in silico models can facilitate the development of CSAFs, but still these models cannot always capture the complexity of the in vivo situation, thereby potentially hampering regulatory acceptance. The combined use of more integrated approaches, like AOPs and physiologically based kinetic models, can aid in structuring data and increasing suitability of alternative approaches for regulatory purposes.
Collapse
Affiliation(s)
- Emma E J Kasteel
- Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Remco H S Westerink
- Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
39
|
Osemwegie O, Butler L, Subbiah S, Smith E. Effects of in vitro exposure of perfluorooctanoic acid and monocrotophos on astroglia SVG p12 cells. J Appl Toxicol 2021; 41:1380-1389. [PMID: 33569802 DOI: 10.1002/jat.4129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022]
Abstract
Glia cells provide supportive functions to the central nervous system and can be compromised by environmental contaminants. The primary objective of this study was to characterize the effects of in vitro exposure to perfluorooctanoic acid, a persistent environmental contaminant and/or monocrotophos (MCP), a neurotoxic organophosphate that is rapidly metabolized, to astroglia SVG p12 cells. The endpoints evaluated include cell viability, intracellular glutamate levels as a marker of astrocyte homeostasis function, differential gene expression for selected proteins, which include inflammatory markers (tachykinin), astrocytosis (nestin), S100B, and metabolism enzymes (CYP1A1). The results from cell viability revealed significant differences from the controls at some of the concentrations tested. Also, intracellular glutamate levels were elevated at the 10-μM concentration for perfluorooctanoic acid (PFOA) as well as the 10-μM PFOA/5-μM MCP concentration. Gene expression results at 80-μM PFOA concentration revealed a significant increase in the expression of S100B, tachykinin and CYP1A1. A combination of 10-μM PFOA/20-μM MCP caused a significant decrease in the expression of tachykinin. Gene expression for MCP exposures produced a decrease at the 20-μM MCP concentration. Immunofluorescence results indicated an increase in nestin protein expression for the 20-μM concentration of MCP, which contradicted the gene expression at the same concentration tested. The results indicate that toxicity to glia cells can compromise critical glia functions and could be implicated in neurodegenerative diseases.
Collapse
Affiliation(s)
- Odia Osemwegie
- Department of Environmental Toxicology, Texas Tech University, Lubbock, Texas, USA
| | - Landon Butler
- Department of Environmental Toxicology, Texas Tech University, Lubbock, Texas, USA
| | - Seenivasan Subbiah
- Department of Environmental Toxicology, Texas Tech University, Lubbock, Texas, USA
| | - Ernest Smith
- Department of Environmental Toxicology, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
40
|
Li D, Jiang L, Hong Y, Cai Z. Multilayered glycoproteomic analysis reveals the hepatotoxic mechanism in perfluorooctane sulfonate (PFOS) exposure mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115774. [PMID: 33143982 DOI: 10.1016/j.envpol.2020.115774] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is one of the most widely used and distributed perfluorinated compounds proven to cause adverse health outcomes. Datasets of ecotoxico-genomics and proteomics have given greater insights for PFOS toxicological effect. However, the molecular mechanisms of hepatotoxicity of PFOS on post-translational modifications (PTMs) regulation, which is most relevant for regulating the activity of proteins, are not well elucidated. Protein glycosylation is one of the most ubiquitous PTMs associated with diverse cellular functions, which are critical towards the understanding of the multiple biological processes and toxic mechanisms exposed to PFOS. Here, we exploit the multilayered glycoproteomics to quantify the global protein expression levels, glycosylation sites, and glycoproteins in PFOS exposure and wild-type mouse livers. The identified 2439 proteins, 1292 glycosites, and 799 glycoproteins were displayed complex heterogeneity in PFOS exposure mouse livers. Quantification results reveal that 241 dysregulated proteins (fold change ≥ 2, p < 0.05) in PFOS exposure mouse livers were involved in the lipid and xenobiotic metabolism. While, 16 overexpressed glycoproteins were exclusively related to neutrophil degranulation, cellular responses to stress, protein processing in endoplasmic reticulum (ER). Moreover, the interactome and functional network analysis identified HP and HSP90AA1 as the potential glycoprotein biomarkers. These results provide unique insights into a deep understanding of the mechanisms of PFOS induced hepatotoxicity and liver disease. Our platform of multilayered glycoproteomics can be adapted to diverse ecotoxicological research.
Collapse
Affiliation(s)
- Dapeng Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China
| | - Lilong Jiang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Yanjun Hong
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
41
|
Berntsen HF, Moldes-Anaya A, Bjørklund CG, Ragazzi L, Haug TM, Strandabø RAU, Verhaegen S, Paulsen RE, Ropstad E, Tasker RA. Perfluoroalkyl acids potentiate glutamate excitotoxicity in rat cerebellar granule neurons. Toxicology 2020; 445:152610. [PMID: 33027616 DOI: 10.1016/j.tox.2020.152610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022]
Abstract
Perfluoroalkyl acids (PFAAs) are persistent man-made chemicals, ubiquitous in nature and present in human samples. Although restrictions are being introduced, they are still used in industrial processes as well as in consumer products. PFAAs cross the blood-brain-barrier and have been observed to induce adverse neurobehavioural effects in humans and animals as well as adverse effects in neuronal in vitro studies. The sulfonated PFAA perfluorooctane sulfonic acid (PFOS), has been shown to induce excitotoxicity via the N-methyl-D-aspartate receptor (NMDA-R) in cultures of rat cerebellar granule neurons (CGNs). In the present study the aim was to further characterise PFOS-induced toxicity (1-60 μM) in rat CGNs, by examining interactions between PFOS and elements of glutamatergic signalling and excitotoxicity. Effects of the carboxylated PFAA, perfluorooctanoic acid (PFOA, 300-500 μM) on the same endpoints were also examined. During experiments in immature cultures at days in vitro (DIV) 8, PFOS increased both the potency and efficacy of glutamate, whereas in mature cultures at DIV 14 only increased potency was observed. PFOA also increased potency at DIV 14. PFOS-enhanced glutamate toxicity was further antagonised by the competitive NMDA-R antagonist 3-((R)-2-Carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP) at DIV 8. At DIV 8, PFOS also induced glutamate release (9-13 fold increase vs DMSO control) after 1-3 and 24 h exposure, whereas for PFOA a large (80 fold) increase was observed, but only after 24 h. PFOS and PFOA both also increased alanine and decreased serine levels after 24 h exposure. In conclusion, our results indicate that PFOS at concentrations relevant in an occupational setting, may be inducing excitotoxicity, and potentiation of glutamate signalling, via an allosteric action on the NMDA-R or by actions on other elements regulating glutamate release or NMDA-R function. Our results further support our previous findings that PFOS and PFOA at equipotent concentrations induce toxicity via different mechanisms of action.
Collapse
Affiliation(s)
- Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102, Oslo, Norway; National Institute of Occupational Health, P.O. Box 8149 Dep N-0033, Oslo, Norway.
| | - Angel Moldes-Anaya
- Research and Development (R&D) Section, PET Imaging Center, University Hospital of North Norway (UNN), Tromsø, Norway; Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Cesilie Granum Bjørklund
- Department of Production Animal Clinical Sciences NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102, Oslo, Norway
| | - Lorenzo Ragazzi
- Neurobiology Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | | | | | - Steven Verhaegen
- Department of Production Animal Clinical Sciences NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102, Oslo, Norway
| | - Ragnhild Elisabeth Paulsen
- Department of Pharmacy, Section for Pharmacology and Pharmaceutical Biosciences, University of Oslo, Oslo, Norway
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102, Oslo, Norway
| | - R Andrew Tasker
- Department of Biomedical Sciences, University of Prince Edward Island, Charlottetown, PEI, Canada; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|