1
|
Rachamalla M, da Silva Junior FC, Hecker M, Niyogi S. Transgenerational Inheritance of Cognitive Deficits Induced by Ancestral Arsenic Exposure in Zebrafish ( Danio rerio) via Maternal and Paternal Lineages. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:9930-9942. [PMID: 40358985 DOI: 10.1021/acs.est.5c00409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Exposure to arsenic is known to impair learning and memory functions in animal models and humans. However, the transgenerational inheritance of these cognitive deficits and the underlying epigenetic mechanisms remain poorly understood. The present study investigated the inter- and transgenerational effects of ancestral arsenic exposure on the cognitive performance (latent learning) of zebrafish via maternal and paternal lineages and the underlying biochemical and molecular alterations in the brain, including the DNA methylation patterns of cognition-related genes. Adult male zebrafish exposed to dietary arsenic [30, 60, or 100 μg/g as arsenite for 90 days; F0 generation] were crossed with unexposed (control) females and vice versa to generate F1 progeny of maternal and paternal arsenic exposure, respectively. Subsequently, F1 males and females of the same treatments were crossed to generate the F2 progeny of the respective maternal and paternal lineages of ancestral arsenic exposure. It was found that ancestral arsenic exposure induced cognitive dysfunction in F1 and F2 generations of both maternal and paternal lineages. However, the effects occurred at relatively lower levels of ancestral arsenic exposure (30 and 60 μg/g) in the former treatment relative to those (100 μg/g) in the latter. Inter (F1) and transgenerational (F2) cognitive effects of arsenic were associated with concomitant elevated oxidative stress and dopaminergic dysregulation, including repressed expression of cognition-related genes such as genes involved in dopamine signaling and metabolism (Drd1 and MAO) and the brain-derived neurotrophic factor (BDNF). Furthermore, DNA methylation analyses revealed that the downregulation of these genes across three generations (F0 to F2) resulted from the hypermethylation in their promotor regions (Drd1, MAO, BDNF). Collectively, these observations provide novel insights into the epigenetic mechanisms of the transgenerational inheritance of arsenic neurotoxicity.
Collapse
Affiliation(s)
- Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | | | - Markus Hecker
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
- School of the Environment and Sustainability, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C8, Canada
- Global Institute for Water Security, University of Saskatchewan, Saskatoon, Saskatchewan S7N 3H5, Canada
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
| |
Collapse
|
2
|
Wu X, Kreutz A, Dixon D, Tokar EJ. Engineering human cerebral organoids to explore mechanisms of arsenic-induced developmental neurotoxicity. Toxicol Appl Pharmacol 2025; 496:117230. [PMID: 39842615 PMCID: PMC11846691 DOI: 10.1016/j.taap.2025.117230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/26/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Modeling brain development and function is challenging due to complexity of the organ. Human pluripotent stem cell (PSC)-derived brain-like organoids provide new tools to study the human brain. Compared with traditional in vivo toxicological studies, these 3D models, together with 2D cellular assays, enhance our understanding of the mechanisms of developmental neurotoxicity (DNT) during the early stages of neurogenesis and offer numerous advantages including a rapid, cost-effective approach for understanding compound mechanisms and assessing chemical safety. Arsenic (As) exposure is associated with DNT, although the mechanisms involved are not well-defined. Here, we used 3D PSC-derived embryoid bodies (EBs) to recapitulate events involved in embryogenesis and neurogenesis before neural induction, and EB-derived cerebral organoids to mimic neural development in vivo. As (0.5 μM; 35 ppb) increased ectoderm differentiation within the EBs by upregulating genes (PAX6, SOX1) critical for embryonic development. Histological staining of EBs showed As disrupted neural rosette structures. qPCR and RNA-seq showed As inhibited expression of markers of mature neural cells (MAP2+/vGLUT2+) and astrocytes (GFAP+). In organoids, Ingenuity Pathway Analysis was used to identify the top 5 pathways affected by As exposure, and Gene Ontology enrichment analysis found several key signaling pathways to be inhibited by As exposure. These data provide insights into pathways contributing to As-induced inhibition of neurite outgrowth and disrupted neural rosette structures in the 2D neurite outgrowth assay and in organoids, respectively. Results herein show this multipronged 2D/3D approach can provide valuable insights into cellular events and molecular mechanisms of As-induced DNT.
Collapse
Affiliation(s)
- Xian Wu
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Anna Kreutz
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Darlene Dixon
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Erik J Tokar
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America.
| |
Collapse
|
3
|
Banaeeyeh S, Razavi BM, Hosseinzadeh H. Neuroprotective Effects of Morin Against Cadmium- and Arsenic-Induced Cell Damage in PC12 Neurons. Biol Trace Elem Res 2024:10.1007/s12011-024-04407-x. [PMID: 39436547 DOI: 10.1007/s12011-024-04407-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024]
Abstract
Arsenic and cadmium, both toxic metals and widespread environmental pollutants, can trigger apoptosis and oxidative stress in various tissues and cells. Morin, a natural flavonoid with diverse biological properties, has been found to protect neurons from oxidative stress and apoptosis-induced damage. This research aimed to examine the protective properties of morin against neurotoxicity caused by arsenic and cadmium, utilizing PC12 cells as a model for nerve cells. The cells were pre-treated with different concentrations of morin and then exposed to arsenic and cadmium, after which cell viability and reactive oxygen species (ROS) production were assessed. Additionally, western blotting was performed to evaluate the protein levels of the Bax/Bcl-2 ratio and cleaved-caspase-3. Following exposure to arsenic and cadmium, there were significant increases in ROS, Bax/Bcl-2 ratio, and cleaved-caspase-3. However, the results of the study demonstrated the beneficial effects of morin at various concentrations, as it increased cell viability and decreased ROS production. Furthermore, morin at a concentration of 10 µM was found to reduce the elevated levels of cleaved-caspase-3 induced by arsenic and diminish the increased Bax/Bcl-2 ratio after exposure to arsenic and cadmium. The findings of this study suggest that morin can effectively protect cells from arsenic and cadmium-induced neurotoxicity through its antioxidant and anti-apoptotic effects. Thus, morin should be considered a promising agent for treating arsenic and cadmium toxicity.
Collapse
Affiliation(s)
- Sara Banaeeyeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Park HR, Azzara D, Cohen ED, Boomhower SR, Diwadkar AR, Himes BE, O'Reilly MA, Lu Q. Identification of novel NRF2-dependent genes as regulators of lead and arsenic toxicity in neural progenitor cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 463:132906. [PMID: 37939567 PMCID: PMC10842917 DOI: 10.1016/j.jhazmat.2023.132906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Lead (Pb) and arsenic (As) are prevalent metal contaminants in the environment. Exposures to these metals are associated with impaired neuronal functions and adverse effects on neurodevelopment in children. However, the molecular mechanisms by which Pb and As impair neuronal functions remain poorly understood. Here, we identified F2RL2, TRIM16L, and PANX2 as novel targets of Nuclear factor erythroid 2-related factor 2 (NRF2)-the master transcriptional factor for the oxidative stress response-that are commonly upregulated with both Pb and As in human neural progenitor cells (NPCs). Using a ChIP (Chromatin immunoprecipitation)-qPCR assay, we showed that NRF2 directly binds to the promoter region of F2RL2, TRIM16L, and PANX2 to regulate expression of these genes. We demonstrated that F2RL2, PANX2, and TRIM16L have differential effects on cell death, proliferation, and differentiation of NPCs in both the presence and absence of metal exposures, highlighting their roles in regulating NPC function. Furthermore, the analyses of the transcriptomic data on NPCs derived from autism spectrum disorder (ASD) patients revealed that dysregulation of F2RL2, TRIM16L, and PANX2 was associated with ASD genetic backgrounds and ASD risk genes. Our findings revealed that Pb and As induce a shared NRF2-dependent transcriptional response in NPCs and identified novel genes regulating NPC function. While further in vivo studies are warranted, this study provides a novel mechanism linking metal exposures to NPC function and identifies potential genes of interest in the context of neurodevelopment.
Collapse
Affiliation(s)
- Hae-Ryung Park
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA.
| | - David Azzara
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Ethan D Cohen
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Steven R Boomhower
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Avantika R Diwadkar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
5
|
Perego MC, McMichael BD, McMurry NR, Ventrello SW, Bain LJ. Arsenic Impairs Differentiation of Human Induced Pluripotent Stem Cells into Cholinergic Motor Neurons. TOXICS 2023; 11:644. [PMID: 37624150 PMCID: PMC10458826 DOI: 10.3390/toxics11080644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/04/2023] [Accepted: 07/14/2023] [Indexed: 08/26/2023]
Abstract
Arsenic exposure during embryogenesis can lead to improper neurodevelopment and changes in locomotor activity. Additionally, in vitro studies have shown that arsenic inhibits the differentiation of sensory neurons and skeletal muscle. In the current study, human-induced pluripotent stem (iPS) cells were differentiated into motor neurons over 28 days, while being exposed to up to 0.5 μM arsenic. On day 6, neuroepithelial progenitor cells (NEPs) exposed to arsenic had reduced transcript levels of the neural progenitor/stem cell marker nestin (NES) and neuroepithelial progenitor marker SOX1, while levels of these transcripts were increased in motor neuron progenitors (MNPs) at day 12. In day 18 early motor neurons (MNs), choline acetyltransferase (CHAT) expression was reduced two-fold in cells exposed to 0.5 μM arsenic. RNA sequencing demonstrated that the cholinergic synapse pathway was impaired following exposure to 0.5 μM arsenic, and that transcript levels of genes involved in acetylcholine synthesis (CHAT), transport (solute carriers, SLC18A3 and SLC5A7) and degradation (acetylcholinesterase, ACHE) were all downregulated in day 18 early MNs. In day 28 mature motor neurons, arsenic significantly downregulated protein expression of microtubule-associated protein 2 (MAP2) and ChAT by 2.8- and 2.1-fold, respectively, concomitantly with a reduction in neurite length. These results show that exposure to environmentally relevant arsenic concentrations dysregulates the differentiation of human iPS cells into motor neurons and impairs the cholinergic synapse pathway, suggesting that exposure impairs cholinergic function in motor neurons.
Collapse
Affiliation(s)
- M. Chiara Perego
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | | | - Nicholas R. McMurry
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Scott W. Ventrello
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Lisa J. Bain
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
6
|
Wang S, Ning H, Wang X, Chen L, Hua L, Ren F, Hu D, Li R, Ma Z, Ge Y, Yin Z. Exposure to bisphenol A induces neurotoxicity associated with synaptic and cytoskeletal dysfunction in neuro-2a cells. Toxicol Ind Health 2023; 39:325-335. [PMID: 37122122 DOI: 10.1177/07482337231172827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Bisphenol A (BPA) has been reported to injure the developing and adult brain. However, the underlying mechanism still remains elusive. This study used neuro-2a cells as a cellular model to investigate the neurotoxic effects of BPA. Microtubule-associated protein 2 (MAP2) and tau protein maintain microtubule normal function and promote the normal development of the nervous system. Synaptophysin (SYP) and drebrin (Dbn) proteins are involved in regulating synaptic plasticity. Cells were exposed to the minimum essential medium (MEM), 0.01% (v/v) DMSO, and 150 μM BPA for 12, 24, or 36 h. Morphological analysis revealed that the cells in the BPA-treated groups shrank and collapsed compared with those in the control groups. CCK-8 and lactate dehydrogenase assay (LDH) assays showed that the mortality of neuro-2a cells increased as the BPA treatment time was prolonged. Ultrastructural analysis further revealed that cells demonstrated nucleolar swelling, dissolution of nuclear and mitochondrial membranes, and partial mitochondrial condensation following exposure to BPA. BPA also decreased the relative protein expression levels of MAP2, tau, and Dbn. Interestingly, the relative protein expression levels of SYP increased. These results indicated that BPA inhibited the proliferation and disrupted cytoskeleton and synaptic integrity of neuro-2a cells.
Collapse
Affiliation(s)
- Siting Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Hongmei Ning
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Xinrui Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Lingli Chen
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Liushuai Hua
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Fei Ren
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Dongfang Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Rongbo Li
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Zhisheng Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Yaming Ge
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Zhihong Yin
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| |
Collapse
|
7
|
Perego MC, McMichael BD, Bain LJ. Arsenic impairs stem cell differentiation via the Hippo signaling pathway. Toxicol Res (Camb) 2023; 12:296-309. [PMID: 37125325 PMCID: PMC10141767 DOI: 10.1093/toxres/tfad018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 04/03/2023] Open
Abstract
Arsenic is a ubiquitous toxic metalloid, with over 150 million people exposed to arsenic concentrations above the current 10 ppb drinking water standard through contaminated food and water. Arsenic is a known developmental toxicant as neuronal and muscle development are disrupted following arsenic exposure during embryogenesis. In this study, murine embryonic stem cells were chronically exposed to 0.1 μM (7.5 ppb) arsenic for 32 weeks. RNA sequencing showed that the Hippo signaling pathway, which is involved in embryonic development and pluripotency maintenance, is impaired following arsenic exposure. Thus, temporal changes in the Hippo pathway's core components and its downstream target genes Ctgf and c-Myc were investigated. Protein expression of the pathway's main effector YAP in its active form was significantly upregulated by 3.7-fold in arsenic-exposed cells at week 8, while protein expression of inactive phosphorylated YAP was significantly downregulated by 2.5- and 2-fold at weeks 8 and 16. Exposure to arsenic significantly increased the ratio between nuclear and cytoplasmic YAP by 1.9-fold at weeks 16 and 28. The ratio between nuclear and cytoplasmic transcriptional enhancer factor domain was similarly increased in arsenic-treated samples by 3.4- and 1.6-fold at weeks 16 and 28, respectively. Levels of Ctgf and c-Myc were also upregulated following arsenic exposure. These results suggest that chronic exposure to an environmentally relevant arsenic concentration might hinder cellular differentiation and maintain pluripotency through the impairment of the Hippo signaling pathway resulting in increased YAP activation.
Collapse
Affiliation(s)
- M Chiara Perego
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC, 29631, United States
| | - Benjamin D McMichael
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC, 29631, United States
- Department of Biology, University of North Carolina, 120 South Road, Chapel Hill, NC, 27599, United States
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC, 29631, United States
| |
Collapse
|
8
|
Lu Z, Wang F, Xia Y, Cheng S, Zhang J, Qin X, Tian X, Wang B, Qiu J, Zou Z, Jiang X, Chen C. Involvement of gut-brain communication in arsenite-induced neurobehavioral impairments in adult male mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114370. [PMID: 36508802 DOI: 10.1016/j.ecoenv.2022.114370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Arsenite is a well-documented neurotoxic metalloid that widely distributes in the natural environment. However, it remains largely unclear how arsenite affects neurological function. Therefore, in this study, the healthy adult male mice were exposed to 0.5 mg/L and 5 mg/L arsenite through drinking water for 30 and 90 days, respectively. Our results showed that there was no significant alteration in the intestine and brain for 30 days exposure, but exposure to arsenite for 90 days significantly induced a reduction of locomotor activity and anxiety-like behavior, caused pathological damage and inflammatory responses in the brain and intestine. We also found that arsenite remarkably disrupted intestinal barrier integrity, decreased the levels of lysozyme and digestive enzymes. Intriguingly, chronic exposure to arsenite significantly changed the levels of gut-brain peptides. Taken together, this study provides meaningful insights that gut-brain communication may involve in the neurobehavioral impairments of arsenite.
Collapse
Affiliation(s)
- Zhaohong Lu
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Fanghong Wang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Shuqun Cheng
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Bin Wang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Xuejun Jiang
- Research Center for Environment and Human Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
9
|
Thakur M, Rachamalla M, Niyogi S, Datusalia AK, Flora SJS. Molecular Mechanism of Arsenic-Induced Neurotoxicity including Neuronal Dysfunctions. Int J Mol Sci 2021; 22:10077. [PMID: 34576240 PMCID: PMC8471829 DOI: 10.3390/ijms221810077] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Arsenic is a key environmental toxicant having significant impacts on human health. Millions of people in developing countries such as Bangladesh, Mexico, Taiwan, and India are affected by arsenic contamination through groundwater. Environmental contamination of arsenic leads to leads to various types of cancers, coronary and neurological ailments in human. There are several sources of arsenic exposure such as drinking water, diet, wood preservatives, smoking, air and cosmetics, while, drinking water is the most explored route. Inorganic arsenic exhibits higher levels of toxicity compared its organic forms. Exposure to inorganic arsenic is known to cause major neurological effects such as cytotoxicity, chromosomal aberration, damage to cellular DNA and genotoxicity. On the other hand, long-term exposure to arsenic may cause neurobehavioral effects in the juvenile stage, which may have detrimental effects in the later stages of life. Thus, it is important to understand the toxicology and underlying molecular mechanism of arsenic which will help to mitigate its detrimental effects. The present review focuses on the epidemiology, and the toxic mechanisms responsible for arsenic induced neurobehavioral diseases, including strategies for its management from water, community and household premises. The review also provides a critical analysis of epigenetic and transgenerational modifications, mitochondrial oxidative stress, molecular mechanisms of arsenic-induced oxidative stress, and neuronal dysfunction.
Collapse
Affiliation(s)
- Manisha Thakur
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
- Toxicology Centre, Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Swaran Jeet Singh Flora
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| |
Collapse
|
10
|
Carmona A, Roudeau S, Ortega R. Molecular Mechanisms of Environmental Metal Neurotoxicity: A Focus on the Interactions of Metals with Synapse Structure and Function. TOXICS 2021; 9:toxics9090198. [PMID: 34564349 PMCID: PMC8471991 DOI: 10.3390/toxics9090198] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
Environmental exposure to neurotoxic metals and metalloids such as arsenic, cadmium, lead, mercury, or manganese is a global health concern affecting millions of people worldwide. Depending on the period of exposure over a lifetime, environmental metals can alter neurodevelopment, neurobehavior, and cognition and cause neurodegeneration. There is increasing evidence linking environmental exposure to metal contaminants to the etiology of neurological diseases in early life (e.g., autism spectrum disorder) or late life (e.g., Alzheimer’s disease). The known main molecular mechanisms of metal-induced toxicity in cells are the generation of reactive oxygen species, the interaction with sulfhydryl chemical groups in proteins (e.g., cysteine), and the competition of toxic metals with binding sites of essential metals (e.g., Fe, Cu, Zn). In neurons, these molecular interactions can alter the functions of neurotransmitter receptors, the cytoskeleton and scaffolding synaptic proteins, thereby disrupting synaptic structure and function. Loss of synaptic connectivity may precede more drastic alterations such as neurodegeneration. In this article, we will review the molecular mechanisms of metal-induced synaptic neurotoxicity.
Collapse
|
11
|
Ramalingam M, Jang S, Jeong HS. Therapeutic Effects of Conditioned Medium of Neural Differentiated Human Bone Marrow-Derived Stem Cells on Rotenone-Induced Alpha-Synuclein Aggregation and Apoptosis. Stem Cells Int 2021; 2021:6658271. [PMID: 33552161 PMCID: PMC7847328 DOI: 10.1155/2021/6658271] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been used against several diseases. Their potential mainly appears from its secreted biomolecules. Human bone marrow-derived stem cells (hBMSC) displayed neuronal functional characteristics after differentiation by basic fibroblast growth factor (bFGF) and forskolin. PD is a chronic age-related neurodegenerative disease (NDD) characterized by loss of dopaminergic neurons in the substantia nigra (SN) and abnormal accumulation of α-synuclein (α-syn) aggregations. In this present study, we evaluated the therapeutic effects of neural differentiated hBMSC (NI-hBMSC) conditioned medium (NI-hBMSC-CM) to a rotenone- (ROT-) induced Parkinson's disease (PD) model in SH-SY5Y cells. NI-hBMSC-CM treatment (50% diluted) in the last 24 h of 48 h ROT (0.5 μM) toxicity showed a significant increase in cell survival. The decreased tyrosine hydroxylase (TH) expression as a hallmark of PD was increased by NI-hBMSC-CM. The Triton X-100-soluble and Triton X-100-insoluble cell lysate fractions were used in Western blotting. The oligomeric, dimeric, and monomeric phosphorylated serine129 (p-S129) α-syn and total monomeric α-syn were decreased during ROT toxicity in the Triton X-100-soluble fraction. The Triton X-100-insoluble fraction revealed that ROT toxicity significantly increased the oligomeric but decreased the dimeric and monomeric p-S129 α-syn expressions while all forms of total α-syn were increased in SH-SY5Y cells. NI-hBMSC-CM stabilized the physiological α-syn monomers and reduced aggregated insoluble p-S129 α-syn against ROT. The cytoskeletal proteins, neurofilament-H (NF-H), β3-tubulin (Tuj1), neuronal nuclei (NeuN), and synaptophysin (SYP) were significantly decreased during ROT toxicity. In addition, proapoptotic Bax was increased by ROT with decreased antiapoptotic Bcl-2 and Mcl-1 as well as proforms of caspase-9, caspase-3, caspase-7, and PARP-1. NI-hBMSC-CM ameliorated the neurotrophic protein expressions, controlled the Bax/Bcl-2 ratio, upregulated procaspases, and inactivated PARP-1. From our results, we conclude that NI-hBMSC-CM containing released biomolecules during neural differentiation employs regenerative effects on the ROT model of PD in SH-SY5Y cells.
Collapse
Affiliation(s)
- Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| |
Collapse
|
12
|
Yin Z, Hua L, Chen L, Hu D, Li J, An Z, Tian T, Ning H, Ge Y. Bisphenol-A exposure induced neurotoxicity and associated with synapse and cytoskeleton in Neuro-2a cells. Toxicol In Vitro 2020; 67:104911. [PMID: 32512148 DOI: 10.1016/j.tiv.2020.104911] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 05/07/2020] [Accepted: 06/01/2020] [Indexed: 12/16/2022]
Abstract
Bisphenol A (BPA) is an environmental chemical that induces neurotoxic effects for human. Synaptophysin (SYP) and drebrin (Dbn) proteins are involved in regulating synaptic morphology. The stability of the cytoskeleton in nerve cells in the brain is regulated by Tau and MAP2. This study aimed to determine the toxicity of BPA to Neuro-2a cells by investigating the synaptic and cytoskeletal damage induced in these cells by 24 h of exposure to 0 (MEM), 50, 100, 150, or 200 μM BPA or DMSO. MTT and LDH assays showed that the death rates of Neuro-2a cells increased, as the BPA concentration increased. Ultrastructural assays revealed that cells underwent nucleolar swelling as well as nuclear membrane and partial mitochondrial dissolution or condensation, following BPA exposure. Morphological analysis further revealed that compared with the cells in the control group, the cells in the BPA-treated groups shrank, became rounded, and exhibited a reduced number of synapses. BPA also significantly decreased the relative protein and mRNA expression levels of Dbn, MAP2 and Tau (P < .01), but increased the relative protein and mRNA expression levels of SYP (P < .01). These results indicated that BPA suppressed the development and proliferation of Neuro-2a cells by disrupting cellular and synaptic integrity and inflicting cytoskeleton injury.
Collapse
Affiliation(s)
- Zhihong Yin
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Liushuai Hua
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Lingli Chen
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Dongfang Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Jinglong Li
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Zhixing An
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Tian Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Hongmei Ning
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China
| | - Yaming Ge
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China.
| |
Collapse
|
13
|
Niño SA, Chi-Ahumada E, Ortíz J, Zarazua S, Concha L, Jiménez-Capdeville ME. Demyelination associated with chronic arsenic exposure in Wistar rats. Toxicol Appl Pharmacol 2020; 393:114955. [PMID: 32171569 DOI: 10.1016/j.taap.2020.114955] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/18/2020] [Accepted: 03/10/2020] [Indexed: 02/01/2023]
Abstract
Inorganic arsenic is among the major contaminants of groundwater in the world. Worldwide population-based studies demonstrate that chronic arsenic exposure is associated with poor cognitive performance among children and adults, while research in animal models confirms learning and memory deficits after arsenic exposure. The aim of this study was to investigate the long-term effects of environmentally relevant arsenic exposure in the myelination process of the prefrontal cortex (PFC) and corpus callosum (CC). A longitudinal study with repeated follow-up assessments was performed in male Wistar rats exposed to 3 ppm sodium arsenite in drinking water. Animals received the treatment from gestation until 2, 4, 6, or 12 months of postnatal age. The levels of myelin basic protein (MBP) were evaluated by immunohistochemistry/histology and immunoblotting from the PFC and CC. As plausible alterations associated with demyelination, we considered mitochondrial mass (VDAC) and two axonal damage markers: amyloid precursor protein (APP) level and phosphorylated neurofilaments. To analyze the microstructure of the CC in vivo, we acquired diffusion-weighted images at the same ages, from which we derived metrics using the tensor model. Significantly decreased levels of MBP were found in both regions together with significant increases of mitochondrial mass and slight axonal damage at 12 months in the PFC. Ultrastructural imaging demonstrated arsenic-associated decreases of white matter volume, water diffusion anisotropy, and increases in radial diffusivity. This study indicates that arsenic exposure is associated with a significant and persistent negative impact on microstructural features of white matter tracts.
Collapse
Affiliation(s)
- Sandra A Niño
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, C.P 78210 San Luis Potosí, Mexico
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza 2405, C.P 78210 San Luis Potosí, Mexico
| | - Juan Ortíz
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro C.P 76230, Mexico
| | - Sergio Zarazua
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, C.P 78210 San Luis Potosí, Mexico
| | - Luis Concha
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro C.P 76230, Mexico
| | - Maria E Jiménez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza 2405, C.P 78210 San Luis Potosí, Mexico.
| |
Collapse
|
14
|
Bode K, Nolte L, Kamin H, Desens M, Ulmann A, Bergmann GA, Betker P, Reitmeier J, Ripken T, Stern M, Meyer H, Bicker G. Scanning laser optical tomography resolves developmental neurotoxic effects on pioneer neurons. Sci Rep 2020; 10:2641. [PMID: 32060340 PMCID: PMC7021824 DOI: 10.1038/s41598-020-59562-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/31/2020] [Indexed: 01/13/2023] Open
Abstract
Developmental neurotoxic compounds impair the developing human nervous system at lower doses than those affecting adults. Standardized test methods for assessing developmental neurotoxicity (DNT) require the use of high numbers of laboratory animals. Here, we use a novel assay that is based on the development of an intact insect embryo in serum-free culture. Neural pathways in the leg of embryonic locusts are established by a pair of afferent pioneer neurons, extending axons along a well-defined pathway to the central nervous system. After exposure to test chemicals, we analyze pioneer neuron shape with conventional fluorescence microscopy and compare it to 3D images, obtained by scanning laser optical tomography (SLOT) and processed by a segmentation algorithm. The segmented SLOT images resolve the 3D structure of the pioneers, recognize pathfinding defects and are thus advantageous for detecting DNT-positive compounds. The defects in axon elongation and pathfinding of pioneer axons caused by two DNT-positive reference compounds (methylmercury chloride; sodium(meta)arsenite) are compared to the biochemically measured general viability of the embryo. Using conventional fluorescence microscopy to establish concentration-response curves of axon elongation, we show that this assay identifies methylmercury chloride and the pro-apoptotic compound staurosporine as developmental neurotoxicants.
Collapse
Affiliation(s)
- Karsten Bode
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Lena Nolte
- Laser Zentrum Hannover e.V., Industrial and Biomedical Optics Department, D-30419, Hannover, Germany
| | - Hannes Kamin
- Laser Zentrum Hannover e.V., Industrial and Biomedical Optics Department, D-30419, Hannover, Germany
| | - Michael Desens
- Laser Zentrum Hannover e.V., Industrial and Biomedical Optics Department, D-30419, Hannover, Germany
| | - Arthur Ulmann
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Gregor A Bergmann
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Philine Betker
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Jennifer Reitmeier
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Tammo Ripken
- Laser Zentrum Hannover e.V., Industrial and Biomedical Optics Department, D-30419, Hannover, Germany
| | - Michael Stern
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Heiko Meyer
- Laser Zentrum Hannover e.V., Industrial and Biomedical Optics Department, D-30419, Hannover, Germany
| | - Gerd Bicker
- University of Veterinary Medicine Hannover, Institute of Physiology and Cell Biology, Bischofsholer Damm 15/102, 30173, Hannover, Germany.
| |
Collapse
|
15
|
Chen Y, Xie HQ, Sha R, Xu T, Zhang S, Fu H, Xia Y, Liu Y, Xu L, Zhao B. 2,3,7,8-Tetrachlorodibenzo-p-dioxin and up-regulation of neurofilament expression in neuronal cells: Evaluation of AhR and MAPK pathways. ENVIRONMENT INTERNATIONAL 2020; 134:105193. [PMID: 31775093 DOI: 10.1016/j.envint.2019.105193] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/25/2019] [Accepted: 09/16/2019] [Indexed: 06/10/2023]
Abstract
Dioxin exposure is reported to affect nervous system development and increase the risk of neurodegenerative diseases. Generally, dioxin exerts its neurotoxicity via aryl hydrocarbon receptor (AhR). Neurofilament (NF) light (NFL) protein is a biomarker for both neuronal differentiation and neurodegeneration and its expression is controlled by the mitogen-activated protein kinase (MAPK) pathway. However, the effects of dioxin on NFL expression and involved mechanisms are incompletely understood. We aimed to investigate the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on NFL expression and elucidate the underlining signaling pathways and their potential crosstalk, specifically between MAPK and AhR pathway. We employed primary cultured rat cortical neurons to evaluate the effect of TCDD exposure on NFL expression. We also used nerve growth factor (NGF)-treated PC12 cells with specific inhibitors to investigate the involvement of and potential crosstalk between the MAPK pathway and the AhR pathway in mediating the effects of TCDD on NFL expression. After TCDD exposure, NFL mRNA and protein levels were upregulated in cultured neurons. NFL protein was preferentially found in the cell body compared with neurites of the cultured neurons. In PC12 cells, TCDD enhanced both NGF-induced NFL expression and phosphorylation of ERK1/2 and p38. The addition of MAPK-pathway inhibitors (PD98059 and SB230580) partially blocked the TCDD-induced NFL upregulation. CH223191, an AhR antagonist, reversed the upregulation of NFL and phosphorylation of ERK1/2 and p38 induced by TCDD. This study demonstrated TCDD-induced upregulation of NFL in cultured neurons, with protein retained in the cell body. TCDD action was dependent on activation of AhR and MAPK, while crosstalk was found between these two signaling pathways.
Collapse
Affiliation(s)
- Yangsheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Rui Sha
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Tuan Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Songyan Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China; College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Hualing Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Yingjie Xia
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Yiyun Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100085, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Sciences, Beijing 100085, China.
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
16
|
Masjosthusmann S, Siebert C, Hübenthal U, Bendt F, Baumann J, Fritsche E. Arsenite interrupts neurodevelopmental processes of human and rat neural progenitor cells: The role of reactive oxygen species and species-specific antioxidative defense. CHEMOSPHERE 2019; 235:447-456. [PMID: 31272005 DOI: 10.1016/j.chemosphere.2019.06.123] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/15/2019] [Accepted: 06/16/2019] [Indexed: 05/10/2023]
Abstract
Arsenic exposure disturbs brain development in humans. Although developmental neurotoxicity (DNT) of arsenic has been studied in vivo and in vitro, its mode-of-action (MoA) is not completely understood. Here, we characterize the adverse neurodevelopmental effects of sodium arsenite on developing human and rat neural progenitor cells (hNPC, rNPC). Moreover, we analyze the involvement of reactive oxygen species (ROS) and the role of the glutathione (GSH)-dependent antioxidative defense for arsenite-induced DNT in a species-specific manner. We determined IC50 values for sodium arsenite-dependent (0.1-10 μM) inhibition of hNPC and rNPC migration (6.0 μM; >10 μM), neuronal (2.7 μM; 4.4 μM) and oligodendrocyte (1.1 μM; 2.0 μM) differentiation. ROS involvement was studied by quantifying the expression of ROS-regulated genes, measuring glutathione (GSH) levels, inhibiting GSH synthesis and co-exposing cells to the antioxidant N-acetylcysteine. Arsenite reduces NPC migration, neurogenesis and oligodendrogenesis of differentiating hNPC and rNPC at sub-cytotoxic concentrations. Species-specific arsenite cytotoxicity and induction of antioxidative gene expression is inversely related to GSH levels with rNPC possessing >3-fold the amount of GSH than hNPC. Inhibition of GSH synthesis increased the sensitivity towards arsenite in rNPC > hNPC. N-acetylcysteine antagonized arsenite-mediated induction of HMOX1 expression as well as reduction of neuronal and oligodendrocyte differentiation in hNPC suggesting involvement of oxidative stress in arsenite DNT. hNPC are more sensitive towards arsenite-induced neurodevelopmental toxicity than rNPC, probably due to their lower antioxidative defense capacities. This species-specific MoA data might be useful for adverse outcome pathway generation and future integrated risk assessment strategies concerning DNT.
Collapse
Affiliation(s)
- Stefan Masjosthusmann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Clara Siebert
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Ulrike Hübenthal
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Farina Bendt
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Jenny Baumann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Duesseldorf, Germany; Heinrich-Heine University, Universitätsstr. 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
17
|
Panneerselvam L, Raghunath A, Ravi MS, Vetrivel A, Subramaniam V, Sundarraj K, Perumal E. Ferulic acid attenuates arsenic‐induced cardiotoxicity in rats. Biotechnol Appl Biochem 2019; 67:186-195. [DOI: 10.1002/bab.1830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/30/2019] [Indexed: 12/19/2022]
Affiliation(s)
| | - Azhwar Raghunath
- Molecular Toxicology LaboratoryDepartment of BiotechnologyBharathiar University Coimbatore Tamil Nadu India
| | - Manoj Srinivas Ravi
- Molecular Toxicology LaboratoryDepartment of BiotechnologyBharathiar University Coimbatore Tamil Nadu India
| | - Amuthan Vetrivel
- Molecular Toxicology LaboratoryDepartment of BiotechnologyBharathiar University Coimbatore Tamil Nadu India
| | - Vinothini Subramaniam
- Molecular Toxicology LaboratoryDepartment of BiotechnologyBharathiar University Coimbatore Tamil Nadu India
| | - Kiruthika Sundarraj
- Molecular Toxicology LaboratoryDepartment of BiotechnologyBharathiar University Coimbatore Tamil Nadu India
| | - Ekambaram Perumal
- Molecular Toxicology LaboratoryDepartment of BiotechnologyBharathiar University Coimbatore Tamil Nadu India
| |
Collapse
|
18
|
Salmataj SA, Kamath SU, Murty VR, Pai SR. Amelioration of arsenic-induced oxidative stress in CHO cells by Ixora coccinea flower extract. 3 Biotech 2018; 8:446. [PMID: 30333948 PMCID: PMC6177355 DOI: 10.1007/s13205-018-1446-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 09/24/2018] [Indexed: 01/12/2023] Open
Abstract
Chronic exposure to inorganic arsenic creates various health problems. Ixora coccinea flower extract was investigated for its ability to protect against arsenic-induced cytotoxicity and genotoxicity in CHO cell line. MTT assay confirmed the efficacy of the extract in ameliorating arsenic-induced cytotoxicity. The value (48 mM) of 24 h inhibitory concentration (IC50) of sodium arsenate for CHO cells was obtained by MTT assay. Various free radical scavenging assays like DPPH, ABTS and nitric oxide scavenging assay confirmed antioxidant activity of the Ixora coccinea flower extract. Pretreatment of the extract significantly inhibited the arsenic-induced DNA damage (p < 0.01) in CHO cells. The extract administration significantly (p < 0.01) inhibited the intracellular ROS and depolarization of mitochondrial membrane induced by sodium arsenate. Ixora coccinea flower extract reduced oxidative stress in cells. Antioxidant enzymes like catalase and SOD activity was restored significantly (p < 0.01) in pretreated CHO cells. Ixora coccinea flower extract also exhibited the anti-apoptotic potential by decreasing the percentage apoptotic index (p < 0.01). These results may expand the applications of Ixora coccinea flowers as an alternative food with antioxidant properties and protective functions against arsenic (iAs) induced toxicological effects.
Collapse
Affiliation(s)
- S. A. Salmataj
- Department of Biotechnology, Manipal Institute of Technology, MAHE, Manipal, India
| | - Shobha U. Kamath
- Department of Biochemistry, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
| | - V. Ramachandra Murty
- Department of Biotechnology, Manipal Institute of Technology, MAHE, Manipal, India
| | - Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, MAHE, Manipal, India
| |
Collapse
|
19
|
Kaushal P, Kumar P, Mehra RD, Dhar P. Dendritic processes as targets for arsenic induced neurotoxicity: Protective role of curcumin. J ANAT SOC INDIA 2018. [DOI: 10.1016/j.jasi.2018.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Niyomchan A, Visitnonthachai D, Suntararuks S, Ngamsiri P, Watcharasit P, Satayavivad J. Arsenic impairs insulin signaling in differentiated neuroblastoma SH-SY5Y cells. Neurotoxicology 2018. [DOI: 10.1016/j.neuro.2018.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Niño SA, Martel-Gallegos G, Castro-Zavala A, Ortega-Berlanga B, Delgado JM, Hernández-Mendoza H, Romero-Guzmán E, Ríos-Lugo J, Rosales-Mendoza S, Jiménez-Capdeville ME, Zarazúa S. Chronic Arsenic Exposure Increases Aβ (1-42) Production and Receptor for Advanced Glycation End Products Expression in Rat Brain. Chem Res Toxicol 2017; 31:13-21. [PMID: 29155576 DOI: 10.1021/acs.chemrestox.7b00215] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic arsenic exposure during development is associated with alterations of chemical transmission and demyelination, which result in cognitive deficits and peripheral neuropathies. At the cellular level, arsenic toxicity involves increased generation of reactive species that induce severe cellular alterations such as DNA fragmentation, apoptosis, and lipid peroxidation. It has been proposed that arsenic-associated neurodegeneration could evolve to Alzheimer disease in later life.1,2 In this study, the effects of chronic exposure to inorganic arsenic (3 ppm by drinking water) in Wistar rats on the production and elimination of Amyloid-β (Aβ) were evaluated. Male Wistar rats were exposed to 3 ppm of arsenic in drinking water from fetal development until 4 months of age. After behavioral deficits induced by arsenic exposure through contextual fear conditioning were verified, the brains were collected for the determination of total arsenic by inductively coupled plasma-mass spectrometry, the levels of amyloid precursor protein and receptor for advanced glycation end products (RAGE) by Western blot analysis as well as their transcript levels by RT-qPCR, Aβ(1-42) estimation by ELISA assay and the enzymatic activity of β-secretase (BACE1). Our results demonstrate that chronic arsenic exposure induces behavioral deficits accompanied of higher levels of soluble and membranal RAGE and the increase of Aβ(1-42) cleaved. In addition, BACE1 enzymatic activity was increased, while immunoblot assays showed no differences in the low-density lipoprotein receptor-related protein 1 (LRP1) receptor among groups. These results provide evidence of the effects of arsenic exposure on the production of Aβ(1-42) and cerebral amyloid clearance through RAGE in an in vivo model that displays behavioral alterations. This work supports the hypothesis that early exposure to metals may contribute to neurodegeneration associated with amyloid accumulation.
Collapse
Affiliation(s)
| | | | | | | | | | - Héctor Hernández-Mendoza
- Laboratorio Nacional Forense Nuclear, Instituto Nacional de Investigaciones Nucleares , Carretera México-Toluca s/n, CP 52750 La Marquesa Ocoyoacac, México.,Centro de Biociencias, Universidad Autónoma de San Luis Potosí , Km. 14.5 carretera San Luis Potosí - Matehuala, Ejido "Palma de la Cruz", CP 78321 Soledad de Graciano Sánchez, San Luis Potosí, México
| | - Elizabeth Romero-Guzmán
- Laboratorio Nacional Forense Nuclear, Instituto Nacional de Investigaciones Nucleares , Carretera México-Toluca s/n, CP 52750 La Marquesa Ocoyoacac, México
| | | | | | | | | |
Collapse
|
22
|
Witt B, Ebert F, Meyer S, Francesconi KA, Schwerdtle T. Assessing neurodevelopmental effects of arsenolipids in pre-differentiated human neurons. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201700199] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Barbara Witt
- Institute of Nutritional Science; University of Potsdam; Nuthetal Germany
| | - Franziska Ebert
- Institute of Nutritional Science; University of Potsdam; Nuthetal Germany
| | - Sören Meyer
- Institute of Nutritional Science; University of Potsdam; Nuthetal Germany
| | | | - Tanja Schwerdtle
- Institute of Nutritional Science; University of Potsdam; Nuthetal Germany
| |
Collapse
|
23
|
Pandey R, Rai V, Mishra J, Mandrah K, Kumar Roy S, Bandyopadhyay S. From the Cover: Arsenic Induces Hippocampal Neuronal Apoptosis and Cognitive Impairments via an Up-Regulated BMP2/Smad-Dependent Reduced BDNF/TrkB Signaling in Rats. Toxicol Sci 2017; 159:137-158. [DOI: 10.1093/toxsci/kfx124] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
24
|
Witt B, Meyer S, Ebert F, Francesconi KA, Schwerdtle T. Toxicity of two classes of arsenolipids and their water-soluble metabolites in human differentiated neurons. Arch Toxicol 2017; 91:3121-3134. [PMID: 28180949 DOI: 10.1007/s00204-017-1933-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/12/2017] [Indexed: 01/17/2023]
Abstract
Arsenolipids are lipid-soluble organoarsenic compounds, mainly occurring in marine organisms, with arsenic-containing hydrocarbons (AsHCs) and arsenic-containing fatty acids (AsFAs) representing two major subgroups. Recently, toxicity studies of several arsenolipids showed a high cytotoxic potential of those arsenolipids in human liver and bladder cells. Furthermore, feeding studies with Drosophila melanogaster indicated an accumulation of arsenolipids in the fruit fly's brain. In this study, the neurotoxic potential of three AsHCs, two AsFAs and three metabolites (dimethylarsinic acid, thio/oxo-dimethylarsenopropanoic acid) was investigated in comparison to the toxic reference arsenite (iAsIII) in fully differentiated human brain cells (LUHMES cells). Thereby, in the case of AsHCs both the cell number and cell viability were reduced in a low micromolar concentration range comparable to iAsIII, while AsFAs and the applied metabolites were less toxic. Mechanistic studies revealed that AsHCs reduced the mitochondrial membrane potential, whereas neither iAsIII nor AsFAs had an impact. Furthermore, neurotoxic mechanisms were investigated by examining the neuronal network. Here, AsHCs massively disturbed the neuronal network and induced apoptotic effects, while iAsIII and AsFAs showed comparatively lesser effects. Taking into account the substantial in vitro neurotoxic potential of the AsHCs and the fact that they could transfer across the physiological barriers of the brain, a neurotoxic potential in vivo for the AsHCs cannot be excluded and needs to be urgently characterized.
Collapse
Affiliation(s)
- Barbara Witt
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Sören Meyer
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Franziska Ebert
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Kevin A Francesconi
- Institute of Chemistry-Analytical Chemistry, University of Graz, Universitaetsplatz 1, 8010, Graz, Austria
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| |
Collapse
|
25
|
Htike NTT, Maekawa F, Soutome H, Sano K, Maejima S, Aung KH, Tokuda M, Tsukahara S. Arsenic Exposure Induces Unscheduled Mitotic S Phase Entry Coupled with Cell Death in Mouse Cortical Astrocytes. Front Neurosci 2016; 10:297. [PMID: 27445668 PMCID: PMC4926759 DOI: 10.3389/fnins.2016.00297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/13/2016] [Indexed: 11/16/2022] Open
Abstract
There is serious concern about arsenic in the natural environment, which exhibits neurotoxicity and increases the risk of neurodevelopmental disorders. Adverse effects of arsenic have been demonstrated in neurons, but it is not fully understood how arsenic affects other cell types in the brain. In the current study, we examined whether sodium arsenite (NaAsO2) affects the cell cycle, viability, and apoptosis of in vitro-cultured astrocytes isolated from the cerebral cortex of mice. Cultured astrocytes from transgenic mice expressing fluorescent ubiquitination-based cell cycle indicator (Fucci) were subjected to live imaging analysis to assess the effects of NaAsO2 (0, 1, 2, and 4 μM) on the cell cycle and number of cells. Fucci was designed to express monomeric Kusabira Orange2 (mKO2) fused with the ubiquitylation domain of hCdt1, a marker of G1 phase, and monomeric Azami Green (mAG) fused with the ubiquitylation domain of hGem, a marker of S, G2, and M phases. NaAsO2 concentration-dependently decreased the peak levels of the mAG/mKO2 emission ratio when the ratio had reached a peak in astrocytes without NaAsO2 exposure, which was due to attenuating the increase in the mAG-expressing cell number. In contrast, the mAG/mKO2 emission ratio and number of mAG-expressing cells were concentration-dependently increased by NaAsO2 before their peak levels, indicating unscheduled S phase entry. We further examined the fate of cells forced to enter S phase by NaAsO2. We found that most of these cells died up to the end of live imaging. In addition, quantification of the copy number of the glial fibrillary acidic protein gene expressed specifically in astrocytes revealed a concentration-dependent decrease caused by NaAsO2. However, NaAsO2 did not increase the amount of nucleosomes generated from DNA fragmentation and failed to alter the gene expression of molecules relevant to unscheduled S phase entry-coupled apoptosis (p21, p53, E2F1, E2F4, and Gm36566). These findings suggest that NaAsO2 adversely affects the cell cycle and viability of astrocytes by inducing unscheduled S phase entry coupled with cell death that may be caused by mechanisms other than apoptosis.
Collapse
Affiliation(s)
- Nang T T Htike
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Fumihiko Maekawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies Tsukuba, Japan
| | - Haruka Soutome
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Kazuhiro Sano
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies Tsukuba, Japan
| | - Sho Maejima
- Area of Life-NanoBio, Division of Strategy Research, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Kyaw H Aung
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Masaaki Tokuda
- Department of Cell Physiology, Faculty of Medicine/Graduate School of Medicine, Kagawa University Kagawa, Japan
| | - Shinji Tsukahara
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama UniversitySaitama, Japan; Area of Life-NanoBio, Division of Strategy Research, Graduate School of Science and Engineering, Saitama UniversitySaitama, Japan
| |
Collapse
|
26
|
Aung KH, Kyi-Tha-Thu C, Sano K, Nakamura K, Tanoue A, Nohara K, Kakeyama M, Tohyama C, Tsukahara S, Maekawa F. Prenatal Exposure to Arsenic Impairs Behavioral Flexibility and Cortical Structure in Mice. Front Neurosci 2016; 10:137. [PMID: 27064386 PMCID: PMC4814721 DOI: 10.3389/fnins.2016.00137] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/17/2016] [Indexed: 12/20/2022] Open
Abstract
Exposure to arsenic from well water in developing countries is suspected to cause developmental neurotoxicity. Although, it has been demonstrated that exposure to sodium arsenite (NaAsO2) suppresses neurite outgrowth of cortical neurons in vitro, it is largely unknown how developmental exposure to NaAsO2 impairs higher brain function and affects cortical histology. Here, we investigated the effect of prenatal NaAsO2 exposure on the behavior of mice in adulthood, and evaluated histological changes in the prelimbic cortex (PrL), which is a part of the medial prefrontal cortex that is critically involved in cognition. Drinking water with or without NaAsO2 (85 ppm) was provided to pregnant C3H mice from gestational days 8 to 18, and offspring of both sexes were subjected to cognitive behavioral analyses at 60 weeks of age. The brains of female offspring were subsequently harvested and used for morphometrical analyses. We found that both male and female mice prenatally exposed to NaAsO2 displayed an impaired adaptation to repetitive reversal tasks. In morphometrical analyses of Nissl- or Golgi-stained tissue sections, we found that NaAsO2 exposure was associated with a significant increase in the number of pyramidal neurons in layers V and VI of the PrL, but not other layers of the PrL. More strikingly, prenatal NaAsO2 exposure was associated with a significant decrease in neurite length but not dendrite spine density in all layers of the PrL. Taken together, our results indicate that prenatal exposure to NaAsO2 leads to behavioral inflexibility in adulthood and cortical disarrangement in the PrL might contribute to this behavioral impairment.
Collapse
Affiliation(s)
- Kyaw H Aung
- Division of Life Science, Saitama UniversitySaitama, Japan; Department of Pharmacology, National Research Institute for Child Health and DevelopmentSetagaya, Japan
| | | | - Kazuhiro Sano
- Molecular Toxicology Section, National Institute for Environmental Studies Tsukuba, Japan
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development Setagaya, Japan
| | - Akito Tanoue
- Department of Pharmacology, National Research Institute for Child Health and Development Setagaya, Japan
| | - Keiko Nohara
- Molecular Toxicology Section, National Institute for Environmental Studies Tsukuba, Japan
| | - Masaki Kakeyama
- Faculty of Human Sciences, Waseda University Tokorozawa, Japan
| | | | | | - Fumihiko Maekawa
- Molecular Toxicology Section, National Institute for Environmental Studies Tsukuba, Japan
| |
Collapse
|
27
|
Zhou H, Liu Y, Tan XJ, Wang YC, Liu KY, Cui YX. Inhibitory effect of arsenic trioxide on neuronal migration in vitro and its potential molecular mechanism. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:671-677. [PMID: 26407229 DOI: 10.1016/j.etap.2015.08.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 06/05/2023]
Abstract
Primary neuron cultures were established from the brains of neonatal rats and the effects of arsenic trioxide (As2O3) on the migration of neurons and the potential mechanism of As2O3 were investigated. Boyden chamber assay was used to detect the effect of AS2O3 on neuronal migration. Matrix metalloproteinase-2 (MMP-2) and MMP-9 RNA expression and doublecortin (DCX) protein expression were measured. Neuronal migration ability was significantly lower in the 20 μmol/L group compared with the other three groups (all p < 0.001). The expression of both MMP-2 and MMP-9 was significantly inversely correlated with As2O3 concentration. The expression of DCX was significantly higher in the control group compared with the other three groups (all p ≤ 0.003). Thus, the inhibitory effect of As2O3 on the migration of primary neurons might be related to the reduction in MMP-2 and MMP-9 activities and decrease in β-actin and DCX expression.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Pediatrics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Ye Liu
- Department of Otorhinolaryngology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Xin-Jie Tan
- Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yu-Chuan Wang
- Department of Pediatrics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Kai-Yu Liu
- Department of Pediatrics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yu-Xia Cui
- Department of Pediatrics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China.
| |
Collapse
|
28
|
Prakash C, Soni M, Kumar V. Mitochondrial oxidative stress and dysfunction in arsenic neurotoxicity: A review. J Appl Toxicol 2015; 36:179-88. [DOI: 10.1002/jat.3256] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/01/2015] [Accepted: 09/28/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Chandra Prakash
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| | - Manisha Soni
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| | - Vijay Kumar
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| |
Collapse
|
29
|
Teng YC, Tai YI, Huang HJ, Lin AMY. Melatonin Ameliorates Arsenite-Induced Neurotoxicity: Involvement of Autophagy and Mitochondria. Mol Neurobiol 2015; 52:1015-22. [DOI: 10.1007/s12035-015-9250-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Niyomchan A, Watcharasit P, Visitnonthachai D, Homkajorn B, Thiantanawat A, Satayavivad J. Insulin attenuates arsenic-induced neurite outgrowth impairments by activating the PI3K/Akt/SIRT1 signaling pathway. Toxicol Lett 2015; 236:138-44. [PMID: 25982963 DOI: 10.1016/j.toxlet.2015.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/07/2015] [Accepted: 05/11/2015] [Indexed: 10/23/2022]
Abstract
Arsenic neurotoxicity has a broad range of adverse effects on human health, which are induced in part by inhibition of neurite outgrowth. Insulin has been reported to promote neurite extension. The present study investigated whether insulin can protect neurons from impaired neurite outgrowth induced by arsenic, and examined the signaling pathway involved in this action. The study demonstrated that NaAsO2 caused inhibition of neurite outgrowth in differentiated SH-SY5Y cells indicating its neurotoxicity. This inhibitory effect of NaAsO2 was attenuated by insulin. It was found that blocking PI3K or Akt by selective inhibitors canceled the protective effect of insulin against NaAsO2-induced neurite outgrowth impairment suggesting the essential role of active PI3K and Akt in insulin's protective action. Inhibition of GSK3, which mimics an effect of insulin stimulation, had no effect on the impairment of neurite outgrowth by NaAsO2 implying that the insulin protective action is probably not due to its mediation of GSK3 inhibition ability. Moreover, NaAsO2 decreased the Akt activity, as it caused reduction in Akt phosphorylation, and downregulated expression of SIRT1. Additionally, the reduction of these signals by NaAsO2 was attenuated by insulin. Taken together, these results show that insulin attenuates arsenic-induced neurite outgrowth impairment possibly via activation of PI3K/Akt/SIRT1 signaling, and arsenic may exert neurite outgrowth inhibition through a mechanism involving reduction of signaling molecules downstream from insulin, PI3K/Akt/SIRT1. Our findings raise the possibility of using insulin to combat arsenic neurotoxicity.
Collapse
Affiliation(s)
- Apichaya Niyomchan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand
| | - Piyajit Watcharasit
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Higher Education Commission, Thailand.
| | | | | | - Apinya Thiantanawat
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Higher Education Commission, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Thailand; Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6 Rd, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Higher Education Commission, Thailand
| |
Collapse
|
31
|
Teng YC, Jeng CJ, Huang HJ, Lin AMY. Role of autophagy in arsenite-induced neurotoxicity: The involvement of α-synuclein. Toxicol Lett 2015; 233:239-45. [DOI: 10.1016/j.toxlet.2015.01.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 11/28/2022]
|
32
|
Aung KH, Tsukahara S, Maekawa F, Nohara K, Nakamura K, Tanoue A. Role of Environmental Chemical Insult in Neuronal Cell Death and Cytoskeleton Damage. Biol Pharm Bull 2015; 38:1109-12. [DOI: 10.1248/bpb.b14-00890] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kyaw Htet Aung
- Department of Pharmacology, National Research Institute for Child Health and Development
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Fumihiko Maekawa
- Molecular Toxicology Section, Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Keiko Nohara
- Molecular Toxicology Section, Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development
| | - Akito Tanoue
- Department of Pharmacology, National Research Institute for Child Health and Development
| |
Collapse
|
33
|
Tolins M, Ruchirawat M, Landrigan P. The developmental neurotoxicity of arsenic: cognitive and behavioral consequences of early life exposure. Ann Glob Health 2014; 80:303-14. [PMID: 25459332 DOI: 10.1016/j.aogh.2014.09.005] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND More than 200 million people worldwide are chronically exposed to arsenic. Arsenic is a known human carcinogen, and its carcinogenic and systemic toxicity have been extensively studied. By contrast, the developmental neurotoxicity of arsenic has been less well described. The aim of this review was to provide a comprehensive review of the developmental neurotoxicity of arsenic. METHODS We reviewed the published epidemiological and toxicological literature on the developmental neurotoxicity of arsenic. RESULTS Arsenic is able to gain access to the developing brain and cause neurotoxic effects. Animal models link prenatal and early postnatal exposure to reduction in brain weight, reductions in numbers of glia and neurons, and alterations in neurotransmitter systems. Animal and in vitro studies both suggest that oxidative stress may be a mechanism of arsenic neurotoxicity. Fifteen epidemiological studies indicate that early life exposure is associated with deficits in intelligence and memory. These effects may occur at levels of exposure below current safety guidelines, and some neurocognitive consequences may become manifest only later in life. Sex, concomitant exposures, and timing of exposure appear to modify the developmental neurotoxicity of arsenic. Four epidemiological studies failed to show behavioral outcomes of arsenic exposure. CONCLUSIONS The published literature indicates that arsenic is a human developmental neurotoxicant. Ongoing and future prospective birth cohort studies will allow more precise definition of the developmental consequences of arsenic exposure in early life.
Collapse
Affiliation(s)
- Molly Tolins
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok, Thailand; Departments of Preventive Medicine and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok, Thailand
| | - Philip Landrigan
- Departments of Preventive Medicine and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
34
|
Kanaya M, Tsuda MC, Sagoshi S, Nagata K, Morimoto C, Tha Thu CK, Toda K, Kato S, Ogawa S, Tsukahara S. Regional difference in sex steroid action on formation of morphological sex differences in the anteroventral periventricular nucleus and principal nucleus of the bed nucleus of the stria terminalis. PLoS One 2014; 9:e112616. [PMID: 25398007 PMCID: PMC4232352 DOI: 10.1371/journal.pone.0112616] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/02/2014] [Indexed: 11/18/2022] Open
Abstract
Sex steroid action is critical to form sexually dimorphic nuclei, although it is not fully understood. We previously reported that masculinization of the principal nucleus of the bed nucleus of the stria terminalis (BNSTp), which is larger and has more neurons in males than in females, involves aromatized testosterone that acts via estrogen receptor-α (ERα), but not estrogen receptor-β (ERβ). Here, we examined sex steroid action on the formation of the anteroventral periventricular nucleus (AVPV) that is larger and has more neurons in females. Morphometrical analysis of transgenic mice lacking aromatase, ERα, or ERβ genes revealed that the volume and neuron number of the male AVPV were significantly increased by deletion of aromatase and ERα genes, but not the ERβ gene. We further examined the AVPV and BNSTp of androgen receptor knockout (ARKO) mice. The volume and neuron number of the male BNSTp were smaller in ARKO mice than those in wild-type mice, while no significant effect of ARKO was found on the AVPV and female BNSTp. We also examined aromatase, ERα, and AR mRNA levels in the AVPV and BNSTp of wild-type and ARKO mice on embryonic day (ED) 18 and postnatal day (PD) 4. AR mRNA in the BNSTp and AVPV of wild-type mice was not expressed on ED18 and emerged on PD4. In the AVPV, the aromatase mRNA level was higher on ED18, although the ERα mRNA level was higher on PD4 without any effect of AR gene deletion. Aromatase and ERα mRNA levels in the male BNSTp were significantly increased on PD4 by AR gene deletion. These results suggest that estradiol signaling via ERα during the perinatal period and testosterone signaling via AR during the postnatal period are required for masculinization of the BNSTp, whereas the former is sufficient to defeminize the AVPV.
Collapse
Affiliation(s)
- Moeko Kanaya
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Mumeko C. Tsuda
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Shoko Sagoshi
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Kazuyo Nagata
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Chihiro Morimoto
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Chaw Kyi Tha Thu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Katsumi Toda
- Department of Biochemistry, School of Medicine, Kochi University, Nankoku, Japan
| | | | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
- * E-mail:
| |
Collapse
|
35
|
Senuma M, Mori C, Ogawa T, Kuwagata M. Prenatal sodium arsenite affects early development of serotonergic neurons in the fetal rat brain. Int J Dev Neurosci 2014; 38:204-12. [PMID: 25291237 DOI: 10.1016/j.ijdevneu.2014.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 09/26/2014] [Accepted: 09/26/2014] [Indexed: 10/24/2022] Open
Abstract
Prenatal arsenite exposure has been associated with developmental disorders in children, including reduced IQ and language abnormalities. Animal experiments have also shown that exposure to arsenite during development induced developmental neurotoxicity after birth. However, the evidence is not enough, and the mechanism is poorly understood, especially on the exposure during early brain development. This study assessed effects of sodium (meta) arsenite shortly after exposure on early developing fetal rat brains. Pregnant rats were administered 50 mg/L arsenite in their drinking water or 20 mg/kg arsenite orally using a gastric tube, on gestational days (GD) 9-15. Fetal brains were examined on GD16. Pregnant rats administered 20 mg/kg arsenite showed reductions in maternal body weight gain and food consumption during treatment, but not with 50 mg/L arsenite. Arsenite did not affect fetal development, as determined by body weight, mortality and brain size. Arsenite also did not induce excessive cell death or affect neural cell division in any region of the fetal neuroepithelium. Thyrosine hydroxylase immunohistochemistry revealed no difference in the distribution of catecholaminergic neurons between fetuses of arsenite treated and control rats. However, reductions in the number of serotonin positive cells in the fetal median and dorsal raphe nuclei were observed following maternal treatment with 20mg/kg arsenite. Image analysis showed that the serotonin positive areas decreased in all fetal mid- and hind-brain areas without altering distribution patterns. Maternal stress induced by arsenite toxicity did not alter fetal development. These results suggest that arsenite-induced neurodevelopmental toxicity involves defects in the early development of the serotonin nervous system.
Collapse
Affiliation(s)
- Mika Senuma
- Hatano Research Institute, Food and Drug Safety Center, 729-5 Ochiai, Hadano, Kanagawa 257-8523, Japan.
| | - Chisato Mori
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-8670, Japan.
| | - Tetsuo Ogawa
- Department of Biology, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan.
| | - Makiko Kuwagata
- Hatano Research Institute, Food and Drug Safety Center, 729-5 Ochiai, Hadano, Kanagawa 257-8523, Japan.
| |
Collapse
|
36
|
Aung KH, Win-Shwe TT, Kanaya M, Takano H, Tsukahara S. Involvement of hemeoxygenase-1 in di(2-ethylhexyl) phthalate (DEHP)-induced apoptosis of Neuro-2a cells. J Toxicol Sci 2014; 39:217-29. [DOI: 10.2131/jts.39.217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Kyaw Htet Aung
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Tin-Tin Win-Shwe
- Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Moeko Kanaya
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Hirohisa Takano
- Department of Environmental Engineering, Graduate School of Engineering, Kyoto University
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| |
Collapse
|
37
|
Inorganic Arsenic Exposure and Children’s Neurodevelopment: A Review of the Evidence. TOXICS 2013. [DOI: 10.3390/toxics1010002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Tyler CR, Allan AM. Adult hippocampal neurogenesis and mRNA expression are altered by perinatal arsenic exposure in mice and restored by brief exposure to enrichment. PLoS One 2013; 8:e73720. [PMID: 24019935 PMCID: PMC3760820 DOI: 10.1371/journal.pone.0073720] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/23/2013] [Indexed: 01/30/2023] Open
Abstract
Arsenic is a common and pervasive environmental contaminant found in drinking water in varying concentrations depending on region. Exposure to arsenic induces behavioral and cognitive deficits in both human populations and in rodent models. The Environmental Protection Agency (EPA) standard for the allotment of arsenic in drinking water is in the parts-per-billion range, yet our lab has shown that 50 ppb arsenic exposure during development can have far-reaching consequences into adulthood, including deficits in learning and memory, which have been linked to altered adult neurogenesis. Given that the morphological impact of developmental arsenic exposure on the hippocampus is unknown, we sought to evaluate proliferation and differentiation of adult neural progenitor cells in the dentate gyrus after 50 ppb arsenic exposure throughout the perinatal period of development in mice (equivalent to all three trimesters in humans) using a BrdU pulse-chase assay. Proliferation of the neural progenitor population was decreased by 13% in arsenic-exposed mice, but was not significant. However, the number of differentiated cells was significantly decreased by 41% in arsenic-exposed mice compared to controls. Brief, daily exposure to environmental enrichment significantly increased proliferation and differentiation in both control and arsenic-exposed animals. Expression levels of 31% of neurogenesis-related genes including those involved in Alzheimer's disease, apoptosis, axonogenesis, growth, Notch signaling, and transcription factors were altered after arsenic exposure and restored after enrichment. Using a concentration previously considered safe by the EPA, perinatal arsenic exposure altered hippocampal morphology and gene expression, but did not inhibit the cellular neurogenic response to enrichment. It is possible that behavioral deficits observed during adulthood in animals exposed to arsenic during development derive from the lack of differentiated neural progenitor cells necessary for hippocampal-dependent learning. This study is the first to determine the impact of arsenic exposure during development on adult hippocampal neurogenesis and related gene expression.
Collapse
Affiliation(s)
- Christina R. Tyler
- Department of Neuroscience, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Andrea M. Allan
- Department of Neuroscience, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
39
|
Maekawa F, Tsuboi T, Oya M, Aung KH, Tsukahara S, Pellerin L, Nohara K. Effects of sodium arsenite on neurite outgrowth and glutamate AMPA receptor expression in mouse cortical neurons. Neurotoxicology 2013; 37:197-206. [DOI: 10.1016/j.neuro.2013.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 05/10/2013] [Accepted: 05/10/2013] [Indexed: 11/28/2022]
|