1
|
Tian C, Cai H, Ao Z, Gu L, Li X, Niu VC, Bondesson M, Gu M, Mackie K, Guo F. Engineering human midbrain organoid microphysiological systems to model prenatal PFOS exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174478. [PMID: 38964381 PMCID: PMC11404128 DOI: 10.1016/j.scitotenv.2024.174478] [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: 04/25/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Perfluorooctane sulfonate (PFOS), a class of synthetic chemicals detected in various environmental compartments, has been associated with dysfunctions of the human central nervous system (CNS). However, the underlying neurotoxicology of PFOS exposure is largely understudied due to the lack of relevant human models. Here, we report bioengineered human midbrain organoid microphysiological systems (hMO-MPSs) to recapitulate the response of a fetal human brain to multiple concurrent PFOS exposure conditions. Each hMO-MPS consists of an hMO on a fully 3D printed holder device with a perfusable organoid adhesion layer for enhancing air-liquid interface culturing. Leveraging the unique, simply-fabricated holder devices, hMO-MPSs are scalable, easy to use, and compatible with conventional well-plates, and allow easy transfer onto a multiple-electrode array (MEA) system for plug-and-play measurement of neural activity. Interestingly, the neural activity of hMO-MPSs initially increased and subsequently decreased by exposure to a concentration range of 0, 30, 100, to 300 μM of PFOS. Furthermore, PFOS exposure impaired neural development and promoted neuroinflammation in the engineered hMO-MPSs. Along with PFOS, our platform is broadly applicable for studies toxicology of various other environmental pollutants.
Collapse
Affiliation(s)
- Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Xiang Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Vivian C Niu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States; Bloomington High School South, Bloomington, IN 47401, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH 45229, Cincinnati, United States; University of Cincinnati School of Medicine, OH 45229, Cincinnati, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States.
| |
Collapse
|
2
|
Huang Y, Jia Z, Lu X, Wang Y, Li R, Zhou A, Chen L, Wang Y, Zeng HC, Li P, Ghassabian A, Yuan N, Kong F, Xu S, Liu H. Prenatal Exposure to Per- and Polyfluoroalkyl Substances and ASD-Related Symptoms in Early Childhood: Mediation Role of Steroids. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:16291-16301. [PMID: 39226190 DOI: 10.1021/acs.est.4c04500] [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: 09/05/2024]
Abstract
Previous studies regarding the associations between perfluoroalkyl and polyfluoroalkyl substances (PFAS) and autism spectrum disorder (ASD) have yielded inconsistent results, with the underlying mechanisms remaining unknown. In this study, we quantified 13 PFAS in cord serum samples from 396 neonates and followed the children at age 4 to assess ASD-related symptoms. Our findings revealed associations between certain PFAS and ASD-related symptoms, with a doubling of perfluorononanoic acid (PFNA), perfluorodecanoic acid (PFDA), and perfluoroundecanoic acid (PFUnDA) concentrations associated with respective increases of 1.79, 1.62, and 1.45 units in language-related symptoms and PFDA exhibiting an association with higher score of sensory stimuli. Nonlinear associations were observed in the associations of 6:2 chlorinated polyfluorinated ether sulfonate (Cl-PFAES) and 8:2 Cl-PFAES with ASD-related symptoms. Employing weighted quantile sum (WQS) regression, we observed significant mixture effects of multiple PFAS on all domains of ASD-related symptoms, with PFNA emerging as the most substantial contributor. Assuming causality, we found that 39-40% of the estimated effect of long-chain PFAS (PFUnDA and PFDoDA) exposure on sensory stimuli was mediated by androstenedione. This study provides novel epidemiological data about prenatal PFAS mixture exposure and ASD-related symptoms.
Collapse
Affiliation(s)
- Yun Huang
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhenxian Jia
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xinhe Lu
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, Hainan, China
| | - Yin Wang
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ruizhen Li
- Women and Children Medical and Healthcare Center of Wuhan, Wuhan 430016, Hubei, China
| | - Aifen Zhou
- Women and Children Medical and Healthcare Center of Wuhan, Wuhan 430016, Hubei, China
| | - Lei Chen
- Women and Children Medical and Healthcare Center of Wuhan, Wuhan 430016, Hubei, China
| | - Yuyan Wang
- Department of Population Health, New York University Grossman School of Medicine, 10016 New York, New York, United States
| | - Huai-Cai Zeng
- School of Public Health, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Pei Li
- Department of Physiology and Biophysics, University of New York at Buffalo, 14260 New York, New York, United States
| | - Akhgar Ghassabian
- Department of Population Health, New York University Grossman School of Medicine, 10016 New York, New York, United States
| | - Ningxue Yuan
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Fanjuan Kong
- Medical Record Management Department, Maternal and Child Health Hospital of Hunan Province, Changsha 410008, Hunan, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, Hainan, China
| | - Hongxiu Liu
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
3
|
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
|
4
|
Yi W, Shi J, Wang L, Wang D, Wang Y, Song J, Xin L, Jiang F. Maternal PFOS exposure in mice induces hepatic lipid accumulation and inflammation in adult female offspring: Involvement of microbiome-gut-liver axis and autophagy. JOURNAL OF HAZARDOUS MATERIALS 2024; 470:134177. [PMID: 38565010 DOI: 10.1016/j.jhazmat.2024.134177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Perfluorooctane sulfonates (PFOS) are the persistent organic pollutants. In the present study, 0, 0.3, or 3-mg/kg PFOS were administered to pregnant mice from GD 11 to GD 18. The histopathology of liver and intestine, serum and hepatic lipid levels, lipid metabolism related genes, and gut microbiota were examined in adult female offspring. The results suggested that maternal PFOS exposure increased serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and induced F4/80+ macrophage infiltration in adult female offspring, in addition to the elevation of TNF-α and IL-1β mRNA levels in low-dose and high-dose groups, respectively. Furthermore, maternal exposure to PFOS increased serum triglyceride (TG) and hepatic total cholesterol (TC) levels, which was associated with the alteration of the process of fatty acid transport and β-oxidation, TG synthesis and transport, cholesterol synthesis and excretion in the liver. The AMPK/mTOR/autophagy signaling was also inhibited in the liver of adult female offspring. Moreover, changes in gut microbiota were also related to lipid metabolism, especially for the Desulfovibrio, Ligilactobacillus, Enterorhabdus, HT002 and Peptococcaceae_unclassified. Additionally, maternal exposure to PFOS decreased mRNA expressions of the tight junction protein and AB+ goblet cells in the colon, while increasing the overproduction of lipopolysaccharides (LPS) and F4/80+ macrophage infiltration. Collectively, maternal PFOS exposure induced liver lipid accumulation and inflammation, which strongly correlated with the disruption of the gut-liver axis and autophagy in adult female offspring, highlighting the persistent adverse effects in offspring exposed to PFOS.
Collapse
Affiliation(s)
- Wenjie Yi
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Junwen Shi
- Suzhou Industrial Park Center for Disease Control and Prevention, Suzhou, Jiangsu, China
| | - Liying Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Dongxuan Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Yiting Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Jingwen Song
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Lili Xin
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China.
| | - Fei Jiang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China; School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
5
|
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
|
6
|
Yang J, Wang Y, Xia Y, Ren Y, Wang Z, Meng X, Li S, Liu X, Shao J. PFOS Elicits Cytotoxicity in Neuron Through Astrocyte-Derived CaMKII-DLG1 Signaling In Vitro Rat Hippocampal Model. Neurochem Res 2024; 49:1226-1238. [PMID: 38393622 DOI: 10.1007/s11064-024-04109-9] [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: 08/15/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
Both epidemiological investigation and animal experiments demonstrated that pre-/postnatal exposure to perfluorooctane sulfonic acid (PFOS) could induce neurodevelopmental disorders. Previous studies showed that astrocyte was involved in PFOS-induced neurotoxicity, while little information is available. In the present study, the role of astrocyte-derived calmodulin-dependent protein kinase II (CaMKII)-phosphorylated discs large homolog 1 (DLG1) signaling in PFOS eliciting cytotoxicity in neuron was explored with primary cultured hippocampal astrocyte and neuron. The application of PFOS showed a decreased cell viability, synapse length and glutamate transporter 1 (GLT-1) expression, but an increased CaMKII, DLG1 and cyclic AMP response element binding protein (CREB) expression in primary cultured astrocyte. With 2-(2-hydroxyethylamino)-6-aminohexylcarbamic acid tert-butyl ester-9-isopropylpurine (CK59), the CaMKII inhibitor, the disturbed cell viability and molecules induced by PFOS could be alleviated (CREB expression was excluded) in astrocytes. The cytotoxic effect of neuron exposed to astrocyte conditional medium collected from PFOS (PFOS-ACM) pretreated with CK59 was also decreased. These results indicated that PFOS mediated GLT-1 expression through astrocyte-derived CaMKII-DLG signaling, which might be associated with injuries on neurons. The present study gave an insight in further exploration of mechanism in PFOS-induced neurotoxicity.
Collapse
Affiliation(s)
- Jiawei Yang
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Ying Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yuyan Xia
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Yajie Ren
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Zhi Wang
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xin Meng
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Shuangyue Li
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China.
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
7
|
Gao M, Shen H, Li Q, Gu X, Jia T, Wang Y. Perfluorooctane sulfonate (PFOS) induces apoptosis and autophagy by inhibition of PI3K/AKT/mTOR pathway in human granulosa cell line KGN. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123333. [PMID: 38211877 DOI: 10.1016/j.envpol.2024.123333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/22/2023] [Accepted: 01/07/2024] [Indexed: 01/13/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is recognized as an environmental endocrine disruptor with widespread use in industrial manufacturing and daily life, contributing to various public health concerns. However, the precise impacts of PFOS on the ovary and its regulatory mechanisms remain unclear. This study aims to delineate the ovarian toxicity of PFOS and scrutinize its effects on apoptosis and autophagy through modulation of the PI3K/AKT/mTOR pathway in the human granulosa cell line (KGN). Cell viability, assessed via the Cell Counting Kit-8 (CCK8), revealed a dose-dependent reduction in cell viability upon PFOS exposure. Flow cytometry analysis demonstrated an elevated proportion of apoptotic cells following PFOS treatment. Western blot analyses unveiled increased expression of Bax, Cyt c, cleaved caspase-9, and LC3-II/I, coupled with decreased expression of Bcl-2 and p62. Transmission electron microscopy (TEM) observations illustrated a heightened number of autophagosomes induced by PFOS. Molecular docking investigations, in conjunction with Western blot experiments, substantiated PFOS's significant inhibition of the PI3K/AKT/mTOR signaling pathway. These findings collectively underscore that PFOS induces apoptosis and autophagy in KGN cells through modulation of the PI3K/AKT/mTOR pathway, providing experimental evidence for PFOS-induced ovarian toxicity and elucidating the underlying regulatory mechanisms in KGN cells.
Collapse
Affiliation(s)
- Min Gao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Haofei Shen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Qiuyuan Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xuzhao Gu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tianyu Jia
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Yiqing Wang
- The First School of Clinical Medicine & Research Unit of Peptide Science, Chinese Academy of Medical Science, 2019RU066, Lanzhou University, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application, Key Laboratory for Reproductive Medicine and Embryo of Gansu Province & Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, China.
| |
Collapse
|
8
|
Gao XX, Zuo QL, Fu XH, Song LL, Cen MQ, Wu J. Association between prenatal exposure to per- and polyfluoroalkyl substances and neurodevelopment in children: Evidence based on birth cohort. ENVIRONMENTAL RESEARCH 2023; 236:116812. [PMID: 37536558 DOI: 10.1016/j.envres.2023.116812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/16/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Although numerous studies have examined the effect of prenatal per- and polyfluoroalkyl substances (PFAS) exposure on neurodevelopment in children, findings have been inconsistent. OBJECTIVE To better understand the effects of PFAS exposure during pregnancy on offspring neurodevelopment, we conducted a systematic review of prenatal exposure to different types of PFAS and neurodevelopment in children. METHODS A comprehensive search was conducted in the PubMed, Web of Science, and EMBASE electronic databases up to March 2023. Only birth cohort studies that report a specific association between PFAS exposure during pregnancy and neurodevelopment were included in this review. RESULTS 31 birth cohort studies that met the inclusion criteria were qualitatively integrated. Among these, 14 studies investigated the impact of PFAS exposure during pregnancy on cognition, 13 on neurobehavior, and 4 on both cognition and neurobehavior. Additionally, 4 studies explored the influence of PFAS on children's comprehensive development. CONCLUSION Prenatal PFAS exposure was associated with poor neurodevelopment in children, including psychomotor development, externalizing behavior, and comprehensive development. However, conclusive evidence regarding its effects on other neurological outcomes remains limited. In addition, sex-specific effects on social behavior and sleep problems were identified.
Collapse
Affiliation(s)
- Xin-Xin 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, China
| | - Qian-Lin 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, China
| | - Xi-Hang 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, China
| | - Ling-Ling 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, China
| | - Man-Qiu 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, China
| | - Jing 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, China.
| |
Collapse
|
9
|
Ren Z, Chen C, Zheng Y, Zhang S, Yan J, Cheng F, Wang X, Wang Q, Li C. Refined Qingkailing protects the in vitro neurovascular unit against oxygen-glucose deprivation and re-oxygenation-induced injury. Biomed Pharmacother 2023; 167:115580. [PMID: 37776640 DOI: 10.1016/j.biopha.2023.115580] [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: 07/25/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
Since the proposal of the neurovascular unit (NVU) theory, it has become almost mandatory for neuroprotective medicines against ischaemic stroke (IS) to focus on this unit. Refined Qingkailing (RQKL) is a compound composed of hyodeoxycholic acid, geniposide, baicalin and cholic acid, which has shown great potential in the treatment of IS, but its effect on NVU has not been fully studied. The purpose of this study was to investigate the potential biological pathways that underlie the protective effects of RQKL against NVU damage induced by oxygen-glucose deprivation and re-oxygenation (OGD/R). Using in vitro OGD/R models, we looked into whether RQKL protects the NVU. In order to create an in vitro NVU that resembles IS, we created an OGD/R injury model using primary cultures of brain microvascular endothelial cells, neurons, and astrocytes. Based on our results, we present evidence, for the first time, that RQKL treatment of the injury caused by OGD/R significantly (1) kept the blood brain barrier (BBB) functioning and maintained the architecture of the neurons, (2) mitigated the oxidative stress damage, inflammatory cytokine release, and neuronal death, and (3) upregulated the expression of neurotrophic factors generated from glial cells and the brain in the in vitro model. Therefore, RQKL has a variety of preventive effects against NVU damage caused by OGD/R. RQKL may be a suitable medication for treating IS in a clinical setting.
Collapse
Affiliation(s)
- Zilin Ren
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Congai Chen
- Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100700, China
| | - Yuxiao Zheng
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shuang Zhang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Juntang Yan
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xueqian Wang
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Qingguo Wang
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Changxiang Li
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
10
|
Kim JH, Moon N, Ji E, Moon HB. Effects of postnatal exposure to phthalate, bisphenol a, triclosan, parabens, and per- and poly-fluoroalkyl substances on maternal postpartum depression and infant neurodevelopment: a korean mother-infant pair cohort study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:96384-96399. [PMID: 37572253 DOI: 10.1007/s11356-023-29292-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/08/2023] [Indexed: 08/14/2023]
Abstract
Exposure to endocrine-disrupting chemicals (EDCs) can promote infant neurodevelopmental impairment and maternal postpartum depression (PPD). However, the associations between lactation exposure to EDCs, maternal PPD, and infant neurodevelopment are unclear. Hence, we investigated these relationships in infants aged 36-42 months. We recruited 221 Korean mothers and analyzed 29 EDCs. The Edinburgh Postnatal Depression Scale (EPDS) was used to assess maternal PPD. Bayley scales of infant development; the Swanson, Nolan, and Pelham rating scale (SNAP); and the Child Behavior Checklist (CBCL) were used to assess neurodevelopment in infants exposed to the top 30% of EDC over three years. Multiple regression analyses were adjusted for maternal age, pre-pregnancy body mass index, education, income, employment, residence, and infant age and sex. The rates of infants with clinically abnormal diagnoses on neurologic developmental tests (Balyey, SNAP, and CBCL scales) ranged from 7.7 to 38.5% in this study, with the motor and hyperactivity/impulsivity areas scoring the highest among 65 boys and girls. Mono-2-ethylhexyl phthalate (MEHP) and mono-isononyl phthalate (MiNP) levels in breast milk significantly correlated with infant inattention and hyperactivity. Perfluorononanoic acid (PFNA) and perfluorooctyl sulfonate (PFOS) levels correlated significantly with motor development of BSID-III and total CBCL score which mean infant might have lower developmental status. EDC concentrations in breast milk were not associated with maternal PPD. Overall, lactational exposure to EDCs during the postpartum period can exert a negative effect on maternal PPD and infant neurodevelopment.
Collapse
Affiliation(s)
- Ju Hee Kim
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Korea.
| | - Nalae Moon
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Korea
| | - Eunsun Ji
- Department of Nursing, Konkuk University Global Campus, Chungju, 27478, Korea
| | - Hyo-Bang Moon
- Department of Marine Science and Convergence Technology, College of Science and Convergence Technology, Hanyang University, Ansan, 15588, Korea
| |
Collapse
|
11
|
Zhang H, Zhang C, Xu D, Wang Q, Xu D. Effects of subchronic exposure of perfluorooctane sulfonate on cognitive function of mice and its mechanism. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121650. [PMID: 37062406 DOI: 10.1016/j.envpol.2023.121650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is an emerging persistent organic pollutant, and its potential impact on cognitive function remains unclear. We adopted the C57BL/6J mouse model to investigate the effect of PFOS on cognitive function, as well as the underlying mechanisms. Subchronic exposure was performed by administering PFOS via drinking water for 6 months (at doses of 0, 0.2, and 2.0 mg/kg/day), starting from 10.5 months old. The object recognition ability was tested at 2, 4, and 6 months of exposure, and spatial learning and memory were assessed at endpoint. The apoptosis of neurons and astrocytes in the cortex and hippocampus was analyzed, as well as the potential apoptotic signaling pathways. Our results showed that exposure to PFOS for 6 months caused a decrease in object recognition ability and a decline in learning and spatial memory. PFOS selectively increased apoptosis in neurons of the cerebral cortex and specifically activated the endoplasmic reticulum stress PERK/CHOP signaling pathway. In conclusion, our results confirmed that subchronic exposure to PFOS can lead to cognitive impairment in mice, which might be closely associated with the specific activation of an endoplasmic reticulum stress-induced pro-apoptosis pathway in the cerebral cortex neurons.
Collapse
Affiliation(s)
- Haijing Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Chao Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Donggang Xu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Qin Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Dongqun Xu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China.
| |
Collapse
|
12
|
Zhuchen HY, Wang JY, Liu XS, Shi YW. Research Progress on Neurodevelopmental Toxicity in Offspring after Indirect Exposure to PFASs in Early Life. TOXICS 2023; 11:571. [PMID: 37505537 PMCID: PMC10386615 DOI: 10.3390/toxics11070571] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 07/29/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are widespread environmental pollutants. There is increasing evidence that PFASs have various adverse health effects, including renal toxicity, metabolic dysfunction, endocrine disruption, and developmental toxicity. PFASs have been found to accumulate in the placenta, and some PFASs can cross the placental barrier and subsequently accumulate in the fetus via the maternal-fetal circulation. An increasing number of studies have shown that early life exposure to PFASs can affect fetal neurodevelopment. This paper reviews the characteristics of indirect exposure to PFASs in early life, the effects on neurodevelopment in offspring, and the possible mechanisms of toxic effects.
Collapse
Affiliation(s)
- Huai-Yu Zhuchen
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jie-Yu Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Shan Liu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Guangzhou 510000, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
13
|
Sun J, Shao X, Huang J, Gong M, Zhang J, Yuan Z. Multiple toxicity evaluations of perfluorooctane sulfonate on intact planarian Dugesia japonica. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:60932-60945. [PMID: 37042918 DOI: 10.1007/s11356-023-26842-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/03/2023] [Indexed: 05/10/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is gaining widespread attention as a persistent organic pollutant with multiple mechanisms of toxicity. In this study, PFOS at different concentrations and different exposure times was used to evaluate the multiple toxicities on intact planarian Dugesia japonica. The proliferation of neoblasts, apoptosis, DNA damage and the expression levels of neuronal genes and the major genes of the Wnt pathway were effectively studied. The results demonstrated that the balance between proliferation and apoptosis of intact planarian cells was disrupted after PFOS exposure, which in turn affected tissue homeostasis and differentiation. PFOS exposure led to increased DNA damage and altered neuronal gene expression. In addition, PFOS exposure could down-regulate the expression of Wnt pathway genes, but the inhibition of the Wnt pathway by PFOS was time- and concentration-dependent. These findings suggest that PFOS has multiple toxic effects on planarians and may interfere with cell proliferation and neurodevelopment by affecting the key gene expression in the Wnt pathway, providing estimable information on the neurodevelopmental toxicity and ecotoxicity of PFOS toxicity in aquatic animals and environments.
Collapse
Affiliation(s)
- Jingyi Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Xinxin Shao
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Jinying Huang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Mengxin Gong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Jianyong Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Zuoqing Yuan
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255000, Shandong, China.
| |
Collapse
|
14
|
Zhou A, Wang L, Pi X, Fan C, Chen W, Wang Z, Rong S, Wang T. Effects of perfluorooctane sulfonate (PFOS) on cognitive behavior and autophagy of male mice. J Toxicol Sci 2023; 48:513-526. [PMID: 37661368 DOI: 10.2131/jts.48.513] [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] [Indexed: 09/05/2023]
Abstract
Perfluorooctane sulfonate (PFOS), an emerging environmental pollutant, is reported to cause neurotoxicity in animals and humans, but its underlying mechanisms are still unclear. We used in vivo models to investigate the effects of PFOS on cognition-related behaviors and related mechanisms. After 45 days of intragastric administration of PFOS (2 mg/kg or 8 mg/kg) in 7-week-old C57BL/6 mice, muscle strength, cognitive function and anxiety-like behavior were evaluated by a series of behavioral tests. The underling mechanisms of PFOS on impaired behaviors were evaluated by HE/Nissl staining, electron microscopy observation and western blot analysis. The results indicated that PFOS-exposed mice exhibited significant cognitive impairment, anxiety, neuronal degeneration and the abnormities of synaptic ultrastructure in the cortex and hippocampus. Western blot analysis indicated that PFOS exposure increased microtubule-associated protein light chain 3 (LC3) and decreased p62 protein levels, which may be associated with activation of autophagy leading to neuron damage. In summary, our results suggest that chronic exposure to PFOS adversely affects cognitive-related behavior in mice. These findings provide new mechanistic insights into PFOS-induced neurotoxicity.
Collapse
Affiliation(s)
- Aojia Zhou
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Li Wang
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Xuejiao Pi
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Cheng Fan
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Wenwen Chen
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Ziping Wang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Shuang Rong
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Ting Wang
- Institute of Advanced Pharmaceutical Technology, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, China
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| |
Collapse
|
15
|
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
|
16
|
Starnes HM, Rock KD, Jackson TW, Belcher SM. A Critical Review and Meta-Analysis of Impacts of Per- and Polyfluorinated Substances on the Brain and Behavior. FRONTIERS IN TOXICOLOGY 2022; 4:881584. [PMID: 35480070 PMCID: PMC9035516 DOI: 10.3389/ftox.2022.881584] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/14/2022] [Indexed: 01/09/2023] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a class of structurally diverse synthetic organic chemicals that are chemically stable, resistant to degradation, and persistent in terrestrial and aquatic environments. Widespread use of PFAS in industrial processing and manufacturing over the last 70 years has led to global contamination of built and natural environments. The brain is a lipid rich and highly vascularized organ composed of long-lived neurons and glial cells that are especially vulnerable to the impacts of persistent and lipophilic toxicants. Generally, PFAS partition to protein-rich tissues of the body, primarily the liver and blood, but are also detected in the brains of humans, wildlife, and laboratory animals. Here we review factors impacting the absorption, distribution, and accumulation of PFAS in the brain, and currently available evidence for neurotoxic impacts defined by disruption of neurochemical, neurophysiological, and behavioral endpoints. Emphasis is placed on the neurotoxic potential of exposures during critical periods of development and in sensitive populations, and factors that may exacerbate neurotoxicity of PFAS. While limitations and inconsistencies across studies exist, the available body of evidence suggests that the neurobehavioral impacts of long-chain PFAS exposures during development are more pronounced than impacts resulting from exposure during adulthood. There is a paucity of experimental studies evaluating neurobehavioral and molecular mechanisms of short-chain PFAS, and even greater data gaps in the analysis of neurotoxicity for PFAS outside of the perfluoroalkyl acids. Whereas most experimental studies were focused on acute and subchronic impacts resulting from high dose exposures to a single PFAS congener, more realistic exposures for humans and wildlife are mixtures exposures that are relatively chronic and low dose in nature. Our evaluation of the available human epidemiological, experimental, and wildlife data also indicates heightened accumulation of perfluoroalkyl acids in the brain after environmental exposure, in comparison to the experimental studies. These findings highlight the need for additional experimental analysis of neurodevelopmental impacts of environmentally relevant concentrations and complex mixtures of PFAS.
Collapse
|
17
|
Chowdhury MI, Sana T, Panneerselvan L, Sivaram AK, Megharaj M. Perfluorooctane sulfonate (PFOS) induces several behavioural defects in Caenorhabditis elegans that can also be transferred to the next generations. CHEMOSPHERE 2022; 291:132896. [PMID: 34780740 DOI: 10.1016/j.chemosphere.2021.132896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/31/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a well-known global persistent organic pollutant of grave concern to ecological and human health. Toxicity of PFOS to animals and humans are well studied. Although few studies have reported the behavioral effect of PFOS on nematode Caenorhabditis elegans, it's transgenerational effects were seldom studied. Therefore, we investigated the toxicity of PFOS on several behavioral responses besides bioaccumulation and transgenerational effects in C. elegans. In contrast to the several published studies, we used lower concentrations (0.5-1000 μg/L or 0.001-2.0 μM) that are environmentally relevant and reported to occur close to the contaminated areas. The 48 h median lethal concentration of PFOS was found to be 3.15 μM (1575 μg/L). PFOS (≥0.01 μM) caused severe toxicity to locomotion, and this effect was even transferred to progeny. However, after a few generations, the defect was rectified in the progeny of single-time exposed parent nematodes. Whereas, continuous exposure at 0.001 μM PFOS, no visible defects were observed in the progeny. PFOS (≥0.01 μM) also significantly decreased the brood size in a concentration-dependent manner. Besides, lifespan was affected by the higher concentration of PFOS (≥1.0 μM). These two behavioral endpoints, lifespan and reproduction defects, became less severe in the progeny. Chemotaxis plasticity was also significantly retarded by ≥ 1.0 μM PFOS compared to the control group. Results indicate that PFOS can exert severe neurobehavioral defects that can be transferred from parents to their offspring. The findings of this study have significant implications for the risk assessment of perfluorinated substances in the environment.
Collapse
Affiliation(s)
- Manjurul Islam Chowdhury
- Global Centre for Environmental Remediation, College of Engineering, Science and the Environment, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Tanmoy Sana
- Global Centre for Environmental Remediation, College of Engineering, Science and the Environment, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Logeshwaran Panneerselvan
- Global Centre for Environmental Remediation, College of Engineering, Science and the Environment, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Anithadevi Kenday Sivaram
- Global Centre for Environmental Remediation, College of Engineering, Science and the Environment, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Mallavarapu Megharaj
- Global Centre for Environmental Remediation, College of Engineering, Science and the Environment, The University of Newcastle, Callaghan, NSW, 2308, Australia; Cooperative Research Centre for Contamination Assessment and Remediation of the Environment (CRC CARE), ATC Building, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
18
|
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
|
19
|
Di Nisio A, Pannella M, Vogiatzis S, Sut S, Dall'Acqua S, Rocca MS, Antonini A, Porzionato A, De Caro R, Bortolozzi M, Toni LD, Foresta C. Impairment of human dopaminergic neurons at different developmental stages by perfluoro-octanoic acid (PFOA) and differential human brain areas accumulation of perfluoroalkyl chemicals. ENVIRONMENT INTERNATIONAL 2022; 158:106982. [PMID: 34781208 DOI: 10.1016/j.envint.2021.106982] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
Perfluoroalkyl substances (PFASs) are synthetic chemicals widely used in industrial and consumer products. The environmental spreading of PFASs raises concerns for their impact on human health. In particular, the bioaccumulation in humans due to environmental exposure has been reported also in total brain samples and PFAS exposure has been associated with neurodevelopmental disorders. In this study we aimed to investigate the specific PFAS bioaccumulation in different brain areas. Our data reported major accumulation in the brainstem region, which is richly populated by dopaminergic neurons (DNs), in brain autopsy samples from people resident in a PFAS-polluted area of Italy. Since DNs are the main source of dopamine (DA) in the mammalian central nervous system (CNS), we evaluated the possible functional consequences of perfluoro-octanoic acid (PFOA) exposure in a human model of DNs obtained by differentiation of human induced pluripotent stem cells (hiPSCs). Particularly, we analyzed the specific effect of the exposure to PFOA for 24 h, at the concentration of 10 ng/ml, at 3 different steps of dopaminergic differentiation: the neuronal commitment phase (DP1), the neuronal precursor phase (DP2) and the mature dopaminergic differentiation phase (DP3). Interestingly, compared to untreated cells, exposure to PFOA was associated with a reduced expression of Tyrosine Hydroxylase (TH) and Neurofilament Heavy (NFH), both markers of dopaminergic maturation at DP2 phase. In addition, cells at DP3 phase exposed to PFOA showed a severe reduction in the expression of the Dopamine Transporter (DAT), functionally involved in pre-synaptic dopamine reuptake. In this proof-of-concept study we show a significant impact of PFOA exposure, mainly on the most sensitive stage of neural dopaminergic differentiation, prompting the way for further investigations more directly relevant to risk assessment of these chemicals.
Collapse
Affiliation(s)
| | | | - Stefania Vogiatzis
- Venetian Institute of Molecular Medicine - VIMM, Department of Physics and Astronomy, University of Padova, Italy
| | - Stefania Sut
- Department of Medicine, University of Padova, Padova, Italy
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Angelo Antonini
- Department of Neuroscience, University of Padua, Padova, Italy
| | | | | | - Mario Bortolozzi
- Venetian Institute of Molecular Medicine - VIMM, Department of Physics and Astronomy, University of Padova, Italy
| | - Luca De Toni
- Department of Medicine, University of Padova, Padova, Italy.
| | - Carlo Foresta
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
20
|
Gao Z, Zhang R, Jiang L, Zhou H, Wang Q, Ma Y, Zhang D, Qin Y, Tian P, Zhang N, Shi Z, Xu S. Administration of miR-195 Inhibitor Enhances Memory Function Through Improving Synaptic Degradation and Mitochondrial Dysfunction of the Hippocampal Neurons in SAMP8 Mice. J Alzheimers Dis 2021; 85:1495-1509. [PMID: 34924391 DOI: 10.3233/jad-215301] [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: 12/18/2022]
Abstract
BACKGROUND Mitochondrial dysfunction is an early feature of Alzheimer's disease (AD) and miR-195 is involved in mitochondrial disorder through targeting MFN-2 protein in hippocampal neurons of AD. OBJECTIVE To clarify if administration of miR-195 inhibitor could enhance the memory deficits through improving hippocampal neuron mitochondrial dysfunction in SAMP8 mice. METHODS The expression of miR-195 was detected by RT-qPCR in primary hippocampal neurons and HT-22 cells treated with Aβ 1-42. Morris water maze (MWM) was used to assess the learning and memory function in SAMP8 mice administrated with antagomir-195. Transmission electron microscopy was employed to determine the morphological changes of synapses and mitochondria of hippocampus in SAMP8 mice. Mitochondrial respiration was measured using a high-resolution oxygraph. RESULTS The expression of miR-195 were upregulated in the primary hippocampal neurons and HT-22 cells induced by Aβ 1-42. Inhibition of miR-195 ameliorated the mitochondrial dysfunction in HT-22 cells induced by Aβ 1-42, including mitochondrial morphologic damages, mitochondrial membrane potential, respiration function, and ATP production. Administration of antagomir-195 by the third ventricle injection markedly ameliorated the cognitive function, postsynaptic density thickness, length of synaptic active area, mitochondrial aspect ratio, and area in hippocampus of SAMP8 mice. Finally, antagomir-195 was able to promote an increase in the activity of respiratory chain complex CI and II in SAMP8 mice. CONCLUSION This study demonstrated that miR-195 inhibitor ameliorated the cognitive impairment of AD mice by improving mitochondrial structure damages and dysfunction in the hippocampal neurons, which provide an experimental basis for further exploring the treatment strategy of AD.
Collapse
Affiliation(s)
- Zhaoyu Gao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Rui Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Lei Jiang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Huimin Zhou
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China.,Department of Endocrinology, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qian Wang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Yingxin Ma
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Di Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Yushi Qin
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Pei Tian
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Nan Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Zhongli Shi
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China.,Hebei International Joint Research Center for Brain Science, Shijiazhuang, P. R. China.,Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| |
Collapse
|
21
|
Qu A, Cao T, Li Z, Wang W, Liu R, Wang X, Nie Y, Sun S, Liu X, Zhang X. The association between maternal perfluoroalkyl substances exposure and early attention deficit hyperactivity disorder in children: a systematic review and meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:67066-67081. [PMID: 34244930 DOI: 10.1007/s11356-021-15136-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/22/2021] [Indexed: 05/27/2023]
Abstract
Some studies have shown that maternal exposure to perfluoroalkyl substances (PFASs) may be associated with early attention deficit hyperactivity disorder (ADHD) in children. The purpose of this systematic review and meta-analysis is to verify this association by reviewing existing studies and to provide a strong basis for preventing ADHD. The researchers searched electronic databases such as PubMed, Science Direct, Scopus, Google Scholar, Web of Science, and Embase for all studies published before October 2020. Finally, we included nine articles for analysis. Our meta-analysis showed that maternal exposure to PFASs was not significantly associated with the prevalence rate of early childhood ADHD (perfluorooctanoic acid (PFOA), odds ratio (OR) = 1.00, 95% confidence interval (95% CI) = 0.75-1.25; perfluorooctane sulfonate (PFOS), OR = 1.01, 95% CI = 0.88-1.14; perfluorohexane sulfonate (PFHxS), OR = 1.08, 95% CI = 0.80-1.09; perfluorononanoic acid (PFNA), OR = 1.13, 95% CI = 0.99-1.28; perfluorodecanoic acid (PFDA), OR = 1.23, 95% CI = 0.15-2.32). Due to significant heterogeneity, we subsequently performed subgroup analysis and sensitivity analysis. Through subgroup analysis, we found that PFOS concentration of children's blood and the prevalence rate of early childhood ADHD were statistically positively correlated, and there was also a positive correlation between PFOS exposure and the prevalence rate of early childhood ADHD in the America. Moreover, there was also a statistically positive correlation between PFNA concentration in maternal blood and the prevalence rate of early childhood ADHD. Sensitivity analysis showed that the final results did not change much, the sensitivity was low, and the results were relatively stable. In conclusion, a causal relationship between maternal PFASs exposure and ADHD in children was unlikely. Among them, PFOS, PFNA, and ADHD might have positive associations worthy of further investigation.
Collapse
Affiliation(s)
- Aibin Qu
- Department of Preventive Medicine, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Tengrui Cao
- Department of Preventive Medicine, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Zixuan Li
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Wenjuan Wang
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Ran Liu
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Xue Wang
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Yaxiong Nie
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Suju Sun
- Department of Epidemiology and Hygienic Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Xuehui Liu
- Department of Epidemiology and Hygienic Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China.
| | - Xiaolin Zhang
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Zhongshan East Road 361, Shijiazhuang, 050017, Hebei, People's Republic of China.
| |
Collapse
|
22
|
Deepika D, Sharma RP, Schuhmacher M, Kumar V. Risk Assessment of Perfluorooctane Sulfonate (PFOS) using Dynamic Age Dependent Physiologically based Pharmacokinetic Model (PBPK) across Human Lifetime. ENVIRONMENTAL RESEARCH 2021; 199:111287. [PMID: 34000270 DOI: 10.1016/j.envres.2021.111287] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 06/12/2023]
Abstract
The widespread use of Perfluorooctane sulfonate (PFOS) in everyday life, its long half-life, and the lipophilicity that makes it easily accumulate in the body, raises the question of its safe exposure among different population groups. There are currently enough epidemiological studies showing evidence of PFOS exposure and its associated adverse effects on humans. Moreover, it is already known that physiological changes along with age e.g. organ volume, renal blood flow, cardiac output and albumin concentrations affect chemicals body burden. Human biomonitoring cohort studies have reported PFOS concentrations in blood and autopsy tissue data with PFOS present in sensitive organs across all human lifespan. However, to interpret such biomonitoring data in the context of chemical risk assessment, it is necessary to have a mechanistic framework that explains show the physiological changes across age affects the concentration of chemical inside different tissues of the human body. PBPK model is widely and successfully used in the field of risk assessment. The objective of this manuscript is to develop a dynamic age-dependent PBPK model as an extension of the previously published adult PFOS model and utilize this model to predict and compare the PFOS tissue distribution and plasma concentration across different age groups. Different cohort study data were used for exposure dose reconstruction and evaluation of time-dependent concentration in sensitive organs. Predicted plasma concentration followed trends observed in biomonitoring data and model predictions showed the increased disposition of PFOS in the geriatric population. PFOS model is sensitive to parameters governing renal resorption and elimination across all ages, which is related to PFOS half-life in humans. This model provides an effective framework for improving the quantitative risk assessment of PFOS throughout the human lifetime, particularly in susceptible age groups. The dynamic age-dependent PBPK model provides a step forward for developing such kind of dynamic model for other perfluoroalkyl substances.
Collapse
Affiliation(s)
- Deepika Deepika
- Environmental Engineering Laboratory, Departament d' Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007, Tarragona, Catalonia, Spain
| | - Raju Prasad Sharma
- Environmental Engineering Laboratory, Departament d' Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007, Tarragona, Catalonia, Spain
| | - Marta Schuhmacher
- Environmental Engineering Laboratory, Departament d' Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007, Tarragona, Catalonia, Spain
| | - Vikas Kumar
- Environmental Engineering Laboratory, Departament d' Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007, Tarragona, Catalonia, Spain; IISPV, Hospital Universitari Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Spain.
| |
Collapse
|
23
|
Chowdhury MI, Sana T, Panneerselvan L, Dharmarajan R, Megharaj M. Acute Toxicity and Transgenerational Effects of Perfluorobutane Sulfonate on Caenorhabditis elegans. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:1973-1982. [PMID: 33792982 DOI: 10.1002/etc.5055] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 03/29/2021] [Indexed: 06/12/2023]
Abstract
Perfluorobutane sulfonate (PFBS), due to its increasing use as an alternative to perfluooctane sulfonate (PFOS), is widely detected in humans and the environment, necessitating the evaluation of its potential ecotoxicological risk. We assessed the toxicity and bioaccumulation potential of PFBS in Caenorhabditis elegans, using lethality, locomotion, reproduction, life span, growth, and chemotactic behavior as the effect parameters. In addition, a total of 6 generations of exposed parent animals were monitored for locomotion, brood, and life span behaviors. Life span and brood size were significantly reduced in parent nematodes (P0) following exposure to ≥0.1 mM PFBS, but these negative effects did not transfer to the progeny. Although there was no remarkable effect on reproduction and life span in parent worms exposed to ≤0.01 mM PFBS, multigenerational exposure at 0.0005 mM significantly affected the F4 and F5 progeny. Furthermore, 0.01 to 2.0 mM of PFBS substantially retarded the locomotion behavior of P0 worms. At higher concentrations such as 1.0 mM, this negative effect on locomotion was transferred to the next generation (F1) but later recovered from F2 progeny onward. Our findings demonstrate for the first time that chronic exposure to PFBS at higher concentrations can cause behavioral toxicity and could be transferred to the progeny. These findings have significant implications for the environmental risk assessment of PFBS. Environ Toxicol Chem 2021;40:1973-1982. © 2021 SETAC.
Collapse
Affiliation(s)
- Manjurul Islam Chowdhury
- Global Centre for Environmental Remediation, College of Engineering, Science and Environment, University of Newcastle, Callaghan, New South Wales, Australia
| | - Tanmoy Sana
- Global Centre for Environmental Remediation, College of Engineering, Science and Environment, University of Newcastle, Callaghan, New South Wales, Australia
| | - Logeshwaran Panneerselvan
- Global Centre for Environmental Remediation, College of Engineering, Science and Environment, University of Newcastle, Callaghan, New South Wales, Australia
- Cooperative Research Centre for Contamination Assessment and Remediation of the Environment, University of Newcastle, Callaghan, New South Wales, Australia
| | - Rajarathnam Dharmarajan
- Global Centre for Environmental Remediation, College of Engineering, Science and Environment, University of Newcastle, Callaghan, New South Wales, Australia
- Cooperative Research Centre for Contamination Assessment and Remediation of the Environment, University of Newcastle, Callaghan, New South Wales, Australia
| | - Mallavarapu Megharaj
- Global Centre for Environmental Remediation, College of Engineering, Science and Environment, University of Newcastle, Callaghan, New South Wales, Australia
- Cooperative Research Centre for Contamination Assessment and Remediation of the Environment, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
24
|
Zhu Y, Yang D, Duan X, Zhang Y, Chen D, Gong Z, Liu C. Perfluorooctane sulfonate promotes doxycycline-induced liver tumor progression in male Kras v12 transgenic zebrafish. ENVIRONMENTAL RESEARCH 2021; 196:110962. [PMID: 33675800 DOI: 10.1016/j.envres.2021.110962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 06/12/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant that has been widely detected in the environment and has caused growing international concern. The liver is the main target organ of PFOS exposure. Animal experiments have shown that PFOS exposure can increase the risk of liver tumorigenesis. However, whether PFOS can accelerate liver tumor progression is still unclear. In this study, transgenic zebrafish Tg(fabp10:rtTA2s-M2; TRE2:EGFP-KRASG12V), a hepatocellular carcinoma (HCC) model that can cause liver tumorigenesis by doxycycline (DOX) induction, was used to investigate the effect of PFOS exposure in HCC progression. The male krasV12 transgenic zebrafish were exposed to 20 mg/L DOX, 500 μg/L PFOS or combined 20 mg/L DOX and 500 μg/L PFOS for 10 d. The results showed that co-treated with PFOS and DOX caused oncogenic Kras-induced liver enlargement, increased the percentages of zebrafish with HCC, and aggravated metabolic reprogramming of liver. To the best of our knowledge, this study for the first proved that PFOS could promote liver tumor progression. Decreased vitamin D level and increased fatty acid intake caused by PFOS might be responsible for the tumor-promoting effects. The results suggest that attention should be paid to the tumor-promoting effects of PFOS when assessing its environmental health risks, and these findings provide new insights into the toxicity of PFOS.
Collapse
Affiliation(s)
- Ya Zhu
- Zhejiang Provincial Key Laboratory of Watershed Science and Health, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, China
| | - Dandong Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xinbin Duan
- Yangtze River Fisheries Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China
| | - Yongkang Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Daqing Chen
- Yangtze River Fisheries Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore
| | - Chunsheng Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
25
|
ROS-Triggered Autophagy Is Involved in PFOS-Induced Apoptosis of Human Embryo Liver L-02 Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6625952. [PMID: 33880372 PMCID: PMC8046535 DOI: 10.1155/2021/6625952] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 01/09/2023]
Abstract
The liver is the primary target organ for perfluorooctane sulphonate (PFOS), a recently discovered persistent organic pollutant. However, the mechanisms mediating hepatotoxicity remain unclear. Herein, we explored the relationship between reactive oxygen species (ROS) and autophagy and apoptosis induced by PFOS in L-02 cells, which are incubated with different concentrations of PFOS (0, 50, 100, 150, 200, or 250 μmol/L) for 24 or 48 hrs at 37°C. The results indicated that PFOS exposure decreased cell activities, enhanced ROS levels in a concentration-dependent manner, decreased mitochondrial membrane potential (MMP), and induced autophagy and apoptosis. Compared with the control, 200 μmol/L PFOS increased ROS levels; enhanced the expression of Bax, cleaved-caspase-3, and LC3-II; induced autophagy; decreased MMP; and lowered Bcl-2, p62, and Bcl-2/Bax ratio. The antioxidant N-acetyl cysteine (NAC) protected MMP against PFOS-induced changes and diminished apoptosis and autophagy. Compared with 200 μmol/L PFOS treatment, NAC pretreatment reversed the increase in ROS, Bax, and cleaved-caspase-3 protein caused by PFOS, lowered the apoptosis rate increased by PFOS, and increased the levels of MMP and Bcl-2/Bax ratio decreased by PFOS. The autophagy inhibitor 3-methyladenine and chloroquine decreased apoptosis and cleaved-caspase-3 protein level and increased the Bcl-2/Bax ratio. In summary, our results suggest that ROS-triggered autophagy is involved in PFOS-induced apoptosis in L-02 cells.
Collapse
|
26
|
Zhang H, Lu H, Yu L, Yuan J, Qin S, Li C, Ge RS, Chen H, Ye L. Effects of gestational exposure to perfluorooctane sulfonate on the lung development of offspring rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 272:115535. [PMID: 33223333 DOI: 10.1016/j.envpol.2020.115535] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 06/11/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a man-made fluorosurfactant widely used in industry and consumer products. Previous studies with rats suggested that gestational exposure to PFOS may affect the lung development in the offspring. The mechanism, however, is still unknown. In the present study, we have exposed 24 pregnant SD rats from gestational day 12-18 to different doses of PFOS (0, 1 or 5 mg/kg BW/day). The lungs of the offspring were analyzed at postnatal days 1, 3, 7 and 14. PFOS treatment appeared to reduce the alveolar numbers, resulting in simplified alveolar structure and thickened alveolar septa. Also, PFOS treated animals had increased lung inflammation with up-regulated inflammasome associated proteins NLRP3, ASC, Caspase-1 and GSDMD and increased inflammatory cytokines IL-18 and IL-1β. At the same time, HIF-1α and VEGFA were significantly down-regulated. Since HIF-1α and VEGFA are critical factors promoting alveolar development and pulmonary angiogenesis, these results suggested that PFOS may also affect lung development by inhibiting HIF-1α and VEGFA expression. Our results here indicate that gestational exposure to PFOS may affect lung development in the offspring with pathological characteristics similar to bronchopulmonary dysplasia (BPD), a severe lung developmental defect. The results also suggest that environmental factors such as PFOS may contribute to the increasing incidence of developmental lung diseases, such as BPD, by elevating lung inflammation and inhibiting lung development.
Collapse
Affiliation(s)
- Huishan Zhang
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China; Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hemin Lu
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Lin Yu
- Department of Pediatrics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Jiexin Yuan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shan Qin
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Cong Li
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Ren-Shan Ge
- Department of Anesthesiology, Perioperative Medicine, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Haolin Chen
- Department of Anesthesiology, Perioperative Medicine, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Leping Ye
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China.
| |
Collapse
|
27
|
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
|
28
|
Mokra K. Endocrine Disruptor Potential of Short- and Long-Chain Perfluoroalkyl Substances (PFASs)-A Synthesis of Current Knowledge with Proposal of Molecular Mechanism. Int J Mol Sci 2021; 22:2148. [PMID: 33670069 PMCID: PMC7926449 DOI: 10.3390/ijms22042148] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 01/25/2023] Open
Abstract
Endocrine disruptors are a group of chemical compounds that, even in low concentrations, cause a hormonal imbalance in the body, contributing to the development of various harmful health disorders. Many industry compounds, due to their important commercial value and numerous applications, are produced on a global scale, while the mechanism of their endocrine action has not been fully understood. In recent years, per- and polyfluoroalkyl substances (PFASs) have gained the interest of major international health organizations, and thus more and more studies have been aimed to explain the toxicity of these compounds. PFASs were firstly synthesized in the 1950s and broadly used in the industry in the production of firefighting agents, cosmetics and herbicides. The numerous industrial applications of PFASs, combined with the exceptionally long half-life of these substances in the human body and extreme environmental persistence, result in a common and chronic exposure of the general population to their action. Available data have suggested that human exposure to PFASs can occur during different stages of development and may cause short- or/and long-term health effects. This paper synthetizes the current literature reports on the presence, bioaccumulation and, particularly, endocrine toxicity of selected long- and short-chain PFASs, with a special emphasis on the mechanisms underlying their endocrine actions.
Collapse
Affiliation(s)
- Katarzyna Mokra
- Department of Environmental Pollution Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 St., 90-236 Lodz, Poland
| |
Collapse
|
29
|
Perfluorooctane sulfonate induces autophagy-associated apoptosis through oxidative stress and the activation of extracellular signal-regulated kinases in renal tubular cells. PLoS One 2021; 16:e0245442. [PMID: 33471797 PMCID: PMC7817024 DOI: 10.1371/journal.pone.0245442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/31/2020] [Indexed: 01/09/2023] Open
Abstract
Perfluorooctane sulfonate (PFOS) is among the most abundant organic pollutants and is widely distributed in the environment, wildlife, and humans. Its toxic effects and biological hazards are associated with its long elimination half-life in humans. However, how it affects renal tubular cells (RTCs) remains unclear. In this study, PFOS was observed to mediate the increase in reactive oxygen species (ROS) generation, followed by the activation of the extracellular-signal-regulated kinase 1/2 (ERK1/2) pathway, which induced autophagy in RTCs. Although PFOS treatment induced autophagy after 6 h, prolonged treatment (24 h) reduced the autophagic flux by increasing lysosomal membrane permeability (LMP), leading to increased p62 protein accumulation and subsequent apoptosis. The increase in LMP was visualized through increased green fluorescence with acridine orange staining, and this was attenuated by 3-methyladenine, an autophagy inhibitor. N-acetyl cysteine and an inhibitor of the mitogen-activated protein kinase kinases (U0126) attenuated autophagy and apoptosis. Taken together, these results indicate that ROS activation and ROS-mediated phosphorylated ERK1/2 activation are essential to activate autophagy, resulting in the apoptosis of PFOS-treated RTCs. Our findings provide insight into the mechanism of PFOS-mediated renal toxicity.
Collapse
|
30
|
Iqubal A, Ahmed M, Ahmad S, Sahoo CR, Iqubal MK, Haque SE. Environmental neurotoxic pollutants: review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:41175-41198. [PMID: 32820440 DOI: 10.1007/s11356-020-10539-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/16/2020] [Indexed: 05/23/2023]
Abstract
Environmental pollutants are recognized as one of the major concerns for public health and responsible for various forms of neurological disorders. Some of the common sources of environmental pollutants related to neurotoxic manifestations are industrial waste, pesticides, automobile exhaust, laboratory waste, and burning of terrestrial waste. Among various environmental pollutants, particulate matter, ultrafine particulate matter, nanoparticles, and lipophilic vaporized toxicant (acrolein) easily cross the blood-brain barrier, activate innate immune responses in the astrocytes, microglia, and neurons, and exert neurotoxicity. Growing shreds of evidence from human epidemiological studies have correlated the environmental pollutants with neuroinflammation, oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, myelin sheath disruption, and alterations in the blood-brain barrier anatomy leading to cognitive dysfunction and poor quality of life. These environmental pollutants also considerably cause developmental neurotoxicity, exhibit teratogenic effect and mental growth retardance, and reduce IQ level. Until now, the exact mechanism of pollutant-induced neurotoxicity is not known, but studies have shown interference of pollutants with the endogenous antioxidant defense system, inflammatory pathway (Nrf2/NF-kB, MAPKs/PI3K, and Akt/GSK3β), modulation of neurotransmitters, and reduction in long-term potentiation. In the current review, various sources of pollutants and exposure to the human population, developmental neurotoxicity, and molecular mechanism of different pollutants involved in the pathogenesis of different neurological disorders have been discussed.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Musheer Ahmed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shahnawaz Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Chita Ranjan Sahoo
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
31
|
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
|
32
|
Tu W, Li W, Zhu X, Xu L. Di-2-ethylhexyl phthalate (DEHP) induces apoptosis of mouse HT22 hippocampal neuronal cells via oxidative stress. Toxicol Ind Health 2020; 36:844-851. [PMID: 32909914 DOI: 10.1177/0748233720947205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Di-2-ethylhexyl phthalate (DEHP) has been widely used as a plasticizer in industry and can affect memory; however, the underlying mechanism remains unclear. In the present study, mouse HT22 cells, an immortalized hippocampal neuronal cell line, was utilized as an in vitro model. We showed that DEHP dramatically inhibited cell viability and increased lactate dehydrogenase (LDH) release from the cells in a dose-dependent manner, suggesting that DEHP could cause cytotoxicity of mouse HT22 cells. The protein levels of cleaved Caspase-8, cleaved Caspase-3, and Bax markedly increased in the DEHP-treated cells, whereas there was a significant decrease in the Bcl-2 protein level, implying that DEHP could induce apoptosis of mouse HT22 cells. DEHP exposure significantly increased the content of malondialdehyde, whereas it markedly decreased the level of glutathione and the activities of glutathione peroxidase and superoxide dismutase, suggesting that DEHP induced oxidative stress of the cells. Compared with the DEHP-treated group, the inhibition of cell viability and the release of LDH were rescued in the N-acetyl-l-cysteine plus DEHP group. Furthermore, inhibition of oxidative stress could rescue the induction of apoptosis by DEHP. Collectively, our results indicated that DEHP could induce apoptosis of mouse HT22 cells via oxidative stress.
Collapse
Affiliation(s)
- Wei Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Weifeng Li
- Department of Oncology, Feng Cheng People’s Hospital, Fengcheng, People’s Republic of China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Linlin Xu
- Medical Research Center, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
33
|
Mshaty A, Haijima A, Takatsuru Y, Ninomiya A, Yajima H, Kokubo M, Khairinisa MA, Miyazaki W, Amano I, Koibuchi N. Neurotoxic effects of lactational exposure to perfluorooctane sulfonate on learning and memory in adult male mouse. Food Chem Toxicol 2020; 145:111710. [PMID: 32861761 DOI: 10.1016/j.fct.2020.111710] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 01/09/2023]
Abstract
The present study aims to examine the effect of early lactational perfluorooctane sulfonate (PFOS) exposures on learning and memory in male mice and reveal the underlying mechanisms involved. PFOS solution was orally administered to dams from the postpartum days 1-14, so that pups would be exposed through breast milk. At 8-10 weeks of age, we performed object location test (OLT), object recognition test (ORT), and pairwise visual discrimination (VD) task. We also performed in vivo microdialysis, and mRNA and protein analysis of genes responsible for hippocampal development and function. In both OLT and ORT, the performance of mice in the PFOS-exposed group was significantly lower than those in the control group. In the VD task, the PFOS-exposed group learned significantly slower than the control group. Concentrations of glutamate and gamma-aminobutyric acid in the dorsal hippocampus were significantly higher in the PFOS-exposed group than in the control group. No notable differences were shown in our mRNA and protein studies. The present study showed that lactational PFOS exposure has a profound, long-lasting neurotoxic effect in the hippocampus and consequently leads to learning and memory deficits.
Collapse
Affiliation(s)
- Abdallah Mshaty
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Asahi Haijima
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan; Laboratory for Environmental Brain Science, Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama, 359-1192, Japan.
| | - Yusuke Takatsuru
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan; Department of Nutrition and Health Science, Toyo University, Itakura, Gunma, 374-0193, Japan
| | - Ayane Ninomiya
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroyuki Yajima
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Michifumi Kokubo
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Miski Aghnia Khairinisa
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan; Department of Pharmacology and Clinical Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Wataru Miyazaki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan; Department of Bioscience and Laboratory Medicine, Graduate School of Health Sciences, Hirosaki University, 1 Bunkyo-cho, Hirosaki, Aomori, 036-8560, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
34
|
Wang B, Li D, Yuan Z, Zhang Y, Ma X, Lv Z, Xiao Y, Zhang J. Evaluation of joint effects of perfluorooctane sulfonate and wood vinegar on planarians, Dugesia japonica. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:18089-18098. [PMID: 32170611 DOI: 10.1007/s11356-020-08342-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/05/2020] [Indexed: 05/15/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant and can cause oxidative stress in animals. Wood vinegar (WV) is the water condensate of smoke produced during wood carbonization. It was used for antibacterial application, pest control, and antioxidant. In the study, PFOS and WV were used to treat the planarian, and then the oxidative stress induced by PFOS on the planarian (Dugesia japonica) and the protective effects of WV on lipid peroxidation, related antioxidant enzyme activity, and mRNA expression in the planarian were studied. PFOS caused an increase in malondialdehyde (MDA) contents, a decrease in superoxide dismutase (SOD) and catalase (CAT) activities, and a change in glutathione peroxidase (GPx), glutathione S-transferase (GST), glutathione reductase (GR) activities. The mRNA levels of glutathione peroxidase gene (gpx), glutathione S-transferase enzyme gene (gst), and glutathione reductase gene (gr) are upregulated or downregulated to varying degrees. The WV and co-treatment planarians reduced MDA levels, increased the activities of oxidative stress biomarker enzymes, and restored gene expression levels. Our results show that low concentration of WV has protective effects on the oxidative damage caused by PFOS in the planarian.
Collapse
Affiliation(s)
- Bin Wang
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Danping Li
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Zuoqing Yuan
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Yuejie Zhang
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Xue Ma
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Ziheng Lv
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Yu Xiao
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China
| | - Jianyong Zhang
- School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China.
| |
Collapse
|
35
|
Zeeshan M, Yang Y, Zhou Y, Huang W, Wang Z, Zeng XY, Liu RQ, Yang BY, Hu LW, Zeng XW, Sun X, Yu Y, Dong GH. Incidence of ocular conditions associated with perfluoroalkyl substances exposure: Isomers of C8 Health Project in China. ENVIRONMENT INTERNATIONAL 2020; 137:105555. [PMID: 32059142 DOI: 10.1016/j.envint.2020.105555] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/10/2020] [Accepted: 02/04/2020] [Indexed: 06/10/2023]
Abstract
The detrimental effects of perfluoroalkyl substances (PFASs) on several physiological systems have been reported, but the association of PFASs with eye, one of the most sensitive and exposed organ, has never been explored. To investigate the association between eye diseases including visual impairment (VI) and PFASs isomers, a cross-sectional stratified study was conducted in 1202 Chinese population, aged 22-96 years, from Shenyang, China. A standard protocol including Snellen vision chart, slit-lamp microscopy and direct ophthalmoscopy was used to examine eye diseases/conditions relating to anterior and posterior segment of eyes. In addition, we measured the blood concentrations of 19 linear and branched PFASs at one-time point. Results indicated that blood levels of PFASs were significantly higher in eye disease group than normal group. PFASs exposure were positively associated with both combined eye diseases and individual eye diseases. Among other PFASs, linear perfluorooctane sulfonate (n-PFOS; odds ratio [OR] = 3.37, 95% confidence interval [CI]: 2.50, 4.56), branched perfluorooctane sulfonate (Br-PFOS; OR = 2.25, 95% CI: 1.72, 2.93) and linear perfluorooctanoic acid (n-PFOA; OR = 1.79, 95% CI: 1.36, 2.37) significantly increases the odds of VI. Vitreous disorder was adversely associated with long-chain PFASs exposure. For example, perfluorotridecanoic acid (PFTrDA; OR = 1.86, 95% CI: 1.51, 2.29) and perfluorodecanoic acid (PFDA; OR = 1.79, 95% CI: 1.36, 2.36) showed the most significant association. In conclusion, this study suggests higher serum PFASs levels were associated with increase odds of VI and vitreous disorder in Chinese adults.
Collapse
Affiliation(s)
- Mohammed Zeeshan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yunqing Yang
- Department of Preventive Medicine, Guangzhou Institute of Dermatology, Guangzhou 510095, China
| | - Yang Zhou
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenzhong Huang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhibin Wang
- Department of Environmental Health Sciences, Laboratory of Human Environmental Epigenomes, Bloomberg School of Public Health, Johns Hopkins University, Baltimore 21205, USA
| | - Xiao-Yun Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Ru-Qing Liu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Bo-Yi Yang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Li-Wen Hu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Wen Zeng
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao Sun
- Department of Internal Medicine, Shenyang Women's and Children's Hospital. No.87 Danan Street, Shenhe District, Shenyang 110011, China.
| | - Yunjiang Yu
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Environmental Protection, Guangzhou 510655, China.
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
36
|
Tukker AM, Bouwman LMS, van Kleef RGDM, Hendriks HS, Legler J, Westerink RHS. Perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) acutely affect human α 1β 2γ 2L GABA A receptor and spontaneous neuronal network function in vitro. Sci Rep 2020; 10:5311. [PMID: 32210279 PMCID: PMC7093421 DOI: 10.1038/s41598-020-62152-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/04/2020] [Indexed: 11/28/2022] Open
Abstract
Concerns about the neurotoxic potential of polyfluoroalkyl substances (PFAS) such as perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) increase, although their neurotoxic mechanisms of action remain debated. Considering the importance of the GABAA receptor in neuronal function, we investigated acute effects of PFAS on this receptor and on spontaneous neuronal network activity. PFOS (Lowest Observed Effect Concentration (LOEC) 0.1 µM) and PFOA (LOEC 1 µM) inhibited the GABA-evoked current and acted as non-competitive human GABAA receptor antagonists. Network activity of rat primary cortical cultures increased following exposure to PFOS (LOEC 100 µM). However, exposure of networks of human induced pluripotent stem cell (hiPSC)-derived neurons decreased neuronal activity. The higher sensitivity of the α1β2γ2L GABAA receptor for PFAS as compared to neuronal networks suggests that PFAS have additional mechanisms of action, or that compensatory mechanisms are at play. Differences between rodent and hiPSC-derived neuronal networks highlight the importance of proper model composition. LOECs for PFAS on GABAA receptor and neuronal activity reported here are within or below the range found in blood levels of occupationally exposed humans. For PFOS, LOECs are even within the range found in human serum and plasma of the general population, suggesting a clear neurotoxic risk.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Lianne M S Bouwman
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Hester S Hendriks
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Juliette Legler
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands.
| |
Collapse
|
37
|
Zhang J, Zhang X, Wen C, Duan Y, Zhang H. Lotus seedpod proanthocyanidins protect against neurotoxicity after methyl-mercuric chloride injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 183:109560. [PMID: 31421536 DOI: 10.1016/j.ecoenv.2019.109560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
In the present study, to investigate the prevention mechanism of proanthocyanidins from lotus seedpod (LSPCs) on methyl mercuric chloride (MMC) induced neurotoxicity, neuron/astrocyte cells were co-cultured to simulate the microenvironment in vivo to the greatest extent. The results showed that, compared with MMC group, pretreatment with LSPCs not only improved cell survival rate, decreased the release of lactate dehydrogenase (LDH), decreased the intracellular reactive oxygen species (ROS) level, and prevented the increase of intracellular [Ca2+]i, but also significantly increased the total anti-oxidation capacity (T-AOC) (p<0.05), the levels of glutathione peroxidase (GSH-Px) (p<0.05), glutathione (GSH) (p<0.05), and mitochondrial membrane potential (MMP) (p<0.01). Besides, LSPCs up-regulated the expression of transcriptional factor Nrf2/HO-1 in a concentration-dependent manner. Moreover, LSPCs reduced the expression of Bax protein, significantly increased the expression of Bcl-xl, Bcl-2, β-Ⅲ-Tubulin, SYN, and Arc proteins. The expression of these proteins is mainly regulated by genes and reflects the changes of genes functions. Taken together, these results suggested that LSPCs could enhance cellular antioxidant defense capacity through regulating the activation of Nrf2/HO-1, and involving the inhibition of mitochondria-mediated apoptotic signaling pathway.
Collapse
Affiliation(s)
- Jixian Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Xuxu Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Chaoting Wen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Yuqing Duan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China; Institute of Food Physical Processing, Jiangsu University, Zhenjiang, 212013, China.
| | - Haihui Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
38
|
Fang A, Li D, Hao Z, Wang L, Pan B, Gao L, Qu X, He J. Effects of astrocyte on neuronal outgrowth in a layered 3D structure. Biomed Eng Online 2019; 18:74. [PMID: 31215491 PMCID: PMC6582480 DOI: 10.1186/s12938-019-0694-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/10/2019] [Indexed: 12/15/2022] Open
Abstract
Background Human brain models and pharmacological models of brain diseases are in high demand for drug screening because animal models have been found to be less than ideal for fully representing the human brain and are likely to fail during drug screening and testing; therefore, the construction of brain-like tissues is necessary. Due to the complexity of cortical tissue, the in vitro construction of brain-like tissue models has been restricted to mostly two-dimensional (2D) models and, on a limited scale, three-dimensional (3D) models. Methods In this study, 3D tissue blocks encapsulating neurons and astrocytes were constructed and cultured in vitro to mimic the cortex of the brain and to investigate the effects of astrocytes on the growth of neurons in a 3D culture. Results The results indicated that such methodology can provide a 3D culture environment suitable for neurons and astrocytes to live and function. When both cells were evenly mixed and cultured in a 3D manner, the astrocytes, which showed better outgrowth and a higher proliferation rate, benefited more than the neurons. On the other hand, the neurons benefited, showing longer axons and a denser network of dendrites, when they were accompanied by astrocytes at a certain distance. Conclusion In conclusion, astrocytes stimulated the outgrowth of neurons in a 3D culture environment in vitro. Regardless, the spatial relationship between both types of cells should be controlled. Thus, culturing cells in a 3D manner is necessary to investigate the correlations between them. This study provides a foundation for biofabricating 3D neurons’ cultures to allow for a deeper insight into the relationship between astrocytes or other glial cells and neurons in a 3D culture that is similar to the natural environment of the brain.
Collapse
Affiliation(s)
- Ao Fang
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Dichen Li
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Zhiyan Hao
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Ling Wang
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China. .,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China.
| | - Binglei Pan
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lin Gao
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xiaoli Qu
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jiankang He
- School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.,State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, 710054, China
| |
Collapse
|
39
|
Wang Y, Wang L, Chang W, Zhang Y, Zhang Y, Liu W. Neurotoxic effects of perfluoroalkyl acids: Neurobehavioral deficit and its molecular mechanism. Toxicol Lett 2019; 305:65-72. [DOI: 10.1016/j.toxlet.2019.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/17/2018] [Accepted: 01/25/2019] [Indexed: 02/04/2023]
|
40
|
Role of astrocytes-derived d-serine in PFOS-induced neurotoxicity through NMDARs in the rat primary hippocampal neurons. Toxicology 2019; 422:14-24. [PMID: 31004706 DOI: 10.1016/j.tox.2019.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 03/05/2019] [Accepted: 04/16/2019] [Indexed: 12/30/2022]
Abstract
Perfluorooctane sulfonate (PFOS) is one of the perfluorinated compounds (PFCs), and has been used in industrial and consumer products. It has already been shown that PFOS could be detected in the environmental media and biological species including humans, due to its resistance to environmental degradation. PFOS is known to induce a series of adverse impacts on human health, e.g., as a potential neurotoxic substance. Recent studies suggest that astrocytes act as the mediator in PFOS-induced neurotoxicity; however, the underlying molecular mechanism needs further investigation. Under the physiological condition, astrocytes play an important role in maintaining brain functions through releasing and up-taking of neurotransmitters between astrocytes and neurons. In the present study, astrocytes-derived d-serine was shown to be involved in PFOS-induced apoptosis and death in the rat primary hippocampal neurons. Significant alterations in d-serine were found in astrocytes, mediated by the molecules in d-serine synthesis (serine racemase), metabolism (d-amino acid oxidase) and delivery (calcium, vacuolar type H+-ATPase, alanine-serine-cysteine transporter and connexin 43 hemichannels). Meanwhile, the N-methyl-d-aspartate receptor (NMDAR) subunits (NR1, NR2 A and NR2B) gene and protein expressions were significantly increased in the hippocampal neurons exposed to the PFOS-activated astrocytes-conditional medium (ACM). Further, the adverse effects of PFOS could be attenuated by the fluorocitrate (an inhibitor for d-serine up-taken by the glial cells) application. Our data indicated that astrocytes-derived d-serine was involved in PFOS-induced neurotoxicity through the NMDARs in the rat primary hippocampal neurons.
Collapse
|
41
|
The mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK. Brain Res Bull 2019; 147:1-13. [PMID: 30731111 DOI: 10.1016/j.brainresbull.2019.01.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/01/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022]
Abstract
The previous studies have shown that glial glutamate transporter-1 (GLT-1) participates in cerebral ischemic injury in rats. However, the mechanism involved remains to be elucidated. This study was undertaken to investigate whether p38 MAPK was involved in regulating GLT-1 in the process. At first, it was observed that global brain ischemia for 8 min led to obvious delayed neuronal death, GLT-1 down-regulation and p-p38 MAPK up-regulation in CA1 hippocampus in rats. Then, whether p-p38 MAPK was involved in regulating GLT-1 during cerebral ischemic injury was studied in vitro. Astrocyte-neuron co-cultures exposed to oxygen and glucose deprivation (OGD) were used to mimic brain ischemia. It was observed that lethal OGD (4-h OGD) decreased GLT-1 expression and increased p-p38 MAPK expression in astrocytes. The p-p38 MAPK protein rised from 0 min to 48 h that is the end time of the observation, and the peak value was at 12 h, which was 12.45 times of the control group. Moreover, pre-administration of p38 MAPK inhibitor SB203580 or its siRNA dose-dependently increased GLT-1 expression, and meanwhile alleviated the neuronal death induced by lethal OGD. The above results indicated that p38 MAPK signaling pathway participated in regulating GLT-1 during OGD injury in vitro. Finally, back to in vivo experiment, it was found that pre-administration of SB203580 by intracerebroventricular injection dose-dependently reversed the down-regulation of GLT-1 expression and attenuated the delayed neuronal death normally induced by global brain ischemia in CA1 hippocampus in rats. Taken together, it can be concluded that the mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK.
Collapse
|
42
|
Impairment of neuro-renal cells on exposure to cosmopolitan polluted river water followed by differential protection of Launea taraxacifolia in male rats. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s00580-019-02898-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
43
|
Nrf2 Signaling Elicits a Neuroprotective Role Against PFOS-mediated Oxidative Damage and Apoptosis. Neurochem Res 2018; 43:2446-2459. [DOI: 10.1007/s11064-018-2672-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/24/2018] [Accepted: 10/25/2018] [Indexed: 01/22/2023]
|
44
|
Salgado-Freiría R, López-Doval S, Lafuente A. Perfluorooctane sulfonate (PFOS) can alter the hypothalamic–pituitary–adrenal (HPA) axis activity by modifying CRF1 and glucocorticoid receptors. Toxicol Lett 2018; 295:1-9. [DOI: 10.1016/j.toxlet.2018.05.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/28/2018] [Accepted: 05/20/2018] [Indexed: 12/22/2022]
|
45
|
Yuan Z, Shao X, Miao Z, Zhao B, Zheng Z, Zhang J. Perfluorooctane sulfonate induced neurotoxicity responses associated with neural genes expression, neurotransmitter levels and acetylcholinesterase activity in planarians Dugesia japonica. CHEMOSPHERE 2018; 206:150-156. [PMID: 29738904 DOI: 10.1016/j.chemosphere.2018.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 06/08/2023]
Abstract
As a persistent and widespread toxic organic pollutant in the environment, perfluorooctane sulfonate (PFOS) has the potential to cause great harm to wildlife. In our study, the effects of PFOS on neurodevelopment gene expression, neurotransmitter content, neuronal morphology, acetylcholinesterase (AChE) activity were examined, and the potential neurotoxicity mechanisms of PFOS were also investigated in planarians, Dugesia japonica. Using quantitative real-time PCR analysis, five neurodevelopmental related genes were measured, among which, DjotxA, DjotxB, DjFoxD, and DjFoxG were found to be down-regulated, while Djnlg was found to be up-regulated, following exposure to PFOS for 10 days compared with control groups. In addition, the neurotransmitters including dopamine, serotonin, and γ-aminobutyricacid as well as the acitivity of AChE were altered by PFOS exposure. Furthermore, PFOS exposure altered brain morphology as well as smaller cephalic ganglia which displayed reduced nerve fiber density decreased brain branches compared to controls. Our results demonstrate that neurotransmission was disturbed after exposure to PFOS and that exposure to this pollutant can cause neurotoxic defects. Results from this study provide valuable information regarding the neuro- and ecological toxicity of PFOS in aquatic animals and aquatic environments.
Collapse
Affiliation(s)
- Zuoqing Yuan
- School of Life Sciences, Shandong University of Technology, No. 266 Xincun West Road, Zibo 255000, China
| | - Xinxin Shao
- School of Life Sciences, Shandong University of Technology, No. 266 Xincun West Road, Zibo 255000, China
| | - Zili Miao
- School of Life Sciences, Shandong University of Technology, No. 266 Xincun West Road, Zibo 255000, China
| | - Bosheng Zhao
- School of Life Sciences, Shandong University of Technology, No. 266 Xincun West Road, Zibo 255000, China
| | - Ziyang Zheng
- School of Life Sciences, Shandong University of Technology, No. 266 Xincun West Road, Zibo 255000, China
| | - Jianyong Zhang
- School of Life Sciences, Shandong University of Technology, No. 266 Xincun West Road, Zibo 255000, China.
| |
Collapse
|
46
|
Perfluorooctanesulfonate induces neuroinflammation through the secretion of TNF-α mediated by the JAK2/STAT3 pathway. Neurotoxicology 2018. [DOI: 10.1016/j.neuro.2018.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Liu QS, Hao F, Sun Z, Long Y, Zhou Q, Jiang G. Perfluorohexadecanoic acid increases paracellular permeability in endothelial cells through the activation of plasma kallikrein-kinin system. CHEMOSPHERE 2018; 190:191-200. [PMID: 28987408 DOI: 10.1016/j.chemosphere.2017.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/28/2017] [Accepted: 10/01/2017] [Indexed: 06/07/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are ubiquitous and high persistent in human blood, thus potentially inducing a myriad of deleterious consequences. Plasma kallikrein-kinin system (KKS), which physiologically regulates vascular permeability, is vulnerable to exogenous stimulators, like PFASs with long-chain alkyl backbone substituted by electronegative fluorine. The study on the interactions of PFASs with the KKS and the subsequent effects on vascular permeability would be helpful to illustrate how the chemicals penetrate the biological vascular barriers to reach different tissues. In present study, three representative PFASs, including perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA) and perfluorohexadecanoic acid (PFHxDA), were investigated for their effects on the activation of the KKS, paracellular permeability in human retina endothelial cells (HRECs) and integrity of the adherens junctions. In contrast to either PFOS or PFOA, PFHxDA efficiently triggered KKS activation in a concentration-dependent manner based on protease activity assays. The plasma activated by PFHxDA significantly increased paracellular permeability of HRECs through the degradation of adherens junctions. As evidenced by the antagonistic effect of aprotinin, PFHxDA-involved effects on vascular permeability were mediated by KKS activation. The results herein firstly revealed the mechanistic pathway for PFHxDA induced effects on vascular endothelial cells. Regarding the possible structure-related activities of the chemicals, this finding would be of great help in the risk assessment of PFASs.
Collapse
Affiliation(s)
- Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Fang Hao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yanmin Long
- Institute of Environment and Health, Jianghan University, Wuhan, 430000, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
48
|
Neurodevelopmental Disorders and Environmental Toxicants: Epigenetics as an Underlying Mechanism. Int J Genomics 2017; 2017:7526592. [PMID: 28567415 PMCID: PMC5439185 DOI: 10.1155/2017/7526592] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/02/2017] [Indexed: 01/07/2023] Open
Abstract
The increasing prevalence of neurodevelopmental disorders, especially autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), calls for more research into the identification of etiologic and risk factors. The Developmental Origin of Health and Disease (DOHaD) hypothesizes that the environment during fetal and childhood development affects the risk for many chronic diseases in later stages of life, including neurodevelopmental disorders. Epigenetics, a term describing mechanisms that cause changes in the chromosome state without affecting DNA sequences, is suggested to be the underlying mechanism, according to the DOHaD hypothesis. Moreover, many neurodevelopmental disorders are also related to epigenetic abnormalities. Experimental and epidemiological studies suggest that exposure to prenatal environmental toxicants is associated with neurodevelopmental disorders. In addition, there is also evidence that environmental toxicants can result in epigenetic alterations, notably DNA methylation. In this review, we first focus on the relationship between neurodevelopmental disorders and environmental toxicants, in particular maternal smoking, plastic-derived chemicals (bisphenol A and phthalates), persistent organic pollutants, and heavy metals. We then review studies showing the epigenetic effects of those environmental factors in humans that may affect normal neurodevelopment.
Collapse
|