1
|
Song Q, Hu C, Zhang X, Ji P, Li Y, Peng H, Zheng Y, Zhang H. Integrated multi-omics approaches reveal the neurotoxicity of triclocarban in mouse brain. ENVIRONMENT INTERNATIONAL 2024; 191:108987. [PMID: 39217723 DOI: 10.1016/j.envint.2024.108987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Triclocarban (TCC) is an antimicrobial ingredient that commonly incorporated in many household and personal care products, raising public concerns about its potential health risks. Previous research has showed that TCC could cross the blood-brain barrier, but to date our understanding of its potential neurotoxicity at human-relevant concentrations remains lacking. In this study, we observed anxiety-like behaviors in mice with continuous percutaneous exposure to TCC. Subsequently, we combined lipidomic, proteomic, and metabolic landscapes to investigate the underlying mechanisms of TCC-related neurotoxicity. The results showed that TCC exposure dysregulated the proteins involved in endocytosis and neurodegenerative disorders in mouse cerebrum. Brain energy homeostasis was also altered, as evidenced by the perturbation of pyruvate metabolism, TCA cycle, and oxidative phosphorylation, which in turn caused mitochondrial dysfunction. Meanwhile, the changing trends of sphingolipid signaling pathway and overproduction of mitochondrial reactive oxygen species (mROS) could enhance the neural apoptosis. The in vitro approach further demonstrated that TCC exposure promoted apoptosis, accompanied by the overproduction of mROS and alteration in the mitochondrial membrane potential in N2A cells. Together, dysregulated endocytosis, mROS-related mitochondrial dysfunction and neural cell apoptosis are considered to be crucial factors for TCC-induced neurotoxicity, which may contribute to the occurrence and development of neurodegenerative disorders. Our findings provide novel perspectives for the mechanisms of TCC-triggered neurotoxicity.
Collapse
Affiliation(s)
- Qian Song
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Chengchen Hu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xueying Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Pengweilin Ji
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yansong Li
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hanyong Peng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongna Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
2
|
Wisessaowapak C, Niyomchan A, Visitnonthachai D, Leelaprachakul N, Watcharasit P, Satayavivad J. Arsenic-induced IGF-1 signaling impairment and neurite shortening: The protective roles of IGF-1 through the PI3K/Akt axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:1119-1128. [PMID: 37853848 DOI: 10.1002/tox.23995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/12/2023] [Accepted: 10/07/2023] [Indexed: 10/20/2023]
Abstract
We recently reported that arsenic caused insulin resistance in differentiated human neuroblastoma SH-SY5Y cells. Herein, we further investigated the effects of sodium arsenite on IGF-1 signaling, which shares downstream signaling with insulin. A time-course experiment revealed that sodium arsenite began to decrease IGF-1-stimulated Akt phosphorylation on Day 3 after treatment, indicating that prolonged sodium arsenite treatment disrupted the neuronal IGF-1 response. Additionally, sodium arsenite decreased IGF-1-stimulated tyrosine phosphorylation of the IGF-1 receptor β (IGF-1Rβ) and its downstream target, insulin receptor substrate 1 (IRS1). These results suggested that sodium arsenite impaired the intrinsic tyrosine kinase activity of IGF-1Rβ, ultimately resulting in a reduction in tyrosine-phosphorylated IRS1. Sodium arsenite also reduced IGF-1 stimulated tyrosine phosphorylation of insulin receptor β (IRβ), indicating the potential inhibition of IGF-1R/IR crosstalk by sodium arsenite. Interestingly, sodium arsenite also induced neurite shortening at the same concentrations that caused IGF-1 signaling impairment. A 24-h IGF-1 treatment partially rescued neurite shortening caused by sodium arsenite. Moreover, the reduction in Akt phosphorylation by sodium arsenite was attenuated by IGF-1. Inhibition of PI3K/Akt by LY294002 diminished the protective effects of IGF-1 against sodium arsenite-induced neurite retraction. Together, our findings suggested that sodium arsenite-impaired IGF-1 signaling, leading to neurite shortening through IGF-1/PI3K/Akt.
Collapse
Affiliation(s)
- Churaibhon Wisessaowapak
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | - Apichaya Niyomchan
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Naphada Leelaprachakul
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Piyajit Watcharasit
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, Thailand
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 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
|
4
|
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
|
5
|
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
|
6
|
Zhang L, Li S, Xia M. High-Throughput Neurite Outgrowth Assay Using GFP-Labeled iPSC-Derived Neurons. Curr Protoc 2022; 2:e542. [PMID: 36102902 PMCID: PMC9635626 DOI: 10.1002/cpz1.542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The potential neurotoxicity from an increasing number of drugs and untested environmental chemicals creates a need to develop reliable and efficient in vitro methods for identifying chemicals that may adversely affect the nervous system. An important process in neurodevelopment is neurite outgrowth, which can be affected by developmental neurotoxicity. Currently, neurite outgrowth assays rely mainly on staining, which requires multiple sample processing steps, particularly washing steps, that may introduce variation and limit throughput. Here, we describe a neurite outgrowth assay that uses induced pluripotent stem cell (iPSC)-derived human cortical glutamatergic neurons and/or spinal motor neurons labeled with green fluorescent protein (GFP) to test compounds in a high-content and high-throughput format. This method enables live and time-lapse imaging of GFP-labeled neurons using an assay plate that is continuously imaged at multiple times after chemical treatment. In this article, we describe how to thaw frozen GFP-labeled neurons, culture them, treat them with a compound of interest, and analyze neurite outgrowth using a high-content imaging platform. In this assay, GFP-labeled iPSC-derived human neurons represent a promising tool for identifying and prioritizing compounds with potential developmental neurotoxicity for further hazard characterization. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA. Basic Protocol 1: Thawing and seeding of iPSC-derived neurons Basic Protocol 2: Compound plate preparation and treatment of neurons Basic Protocol 3: High-content imaging and analysis.
Collapse
Affiliation(s)
- Li Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Shuaizhang Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Taheri Zadeh Z, Esmaeilpour K, Aminzadeh A, Heidari MR, Joushi S. Resveratrol Attenuates Learning, Memory, and Social Interaction Impairments in Rats Exposed to Arsenic. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9993873. [PMID: 34621902 PMCID: PMC8492247 DOI: 10.1155/2021/9993873] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/27/2021] [Accepted: 08/18/2021] [Indexed: 12/23/2022]
Abstract
Arsenic (As) toxicity has deleterious effects on human health causing disorder in the brain. The aim of this study was to investigate the possible neuroprotective effect of resveratrol (RSV) on arsenic-induced neurotoxicity in rats. Neurotoxicity in rats was developed by treating As 10 mg/kg/day for 21 days orally. Animals were put into seven groups: control, vehicle, As, As+RSV10, As+RSV20 mg/kg, RSV10, and RSV20 mg/kg. Behavioral assessments such as the social interaction test, novel object recognition test, elevated plus maze, open field, the Morris water maze, in addition to assessment of biomarkers such as ferric reducing ability of plasma assay, glutathione assay, and malondialdehyde assay, were used to evaluate the effects of RSV on cognitive impairment and molecular changes induced by As. The results showed that cognitive performance impaired in As rats. RSV20 mg/kg significantly could ameliorate behavioral changes like spatial learning in days 3 and 4 (p < 0.05), recognition learning and memory (p < 0.01), disabilities in motor coordination and stress (p < 0.05), increased anxiety (p < 0.05), and social interaction deficit (sociability (p < 0.001) and social memory (p < 0.05)). RSV20 mg/kg also attenuated molecular modifications like decreased antioxidant power (p < 0.001), reduced glutathione content (p < 0.05), and increased malondialdehyde level (p < 0.05) induced by As. In addition to oxidative stress assessments, RSV10 mg/kg could significantly increase FRAP (p < 0.01) and GSH (p < 0.05); however, MDA was not significantly increased. Our current behavioral findings suggest that RSV has neuroprotective effects against AS toxicity.
Collapse
Affiliation(s)
- Zahra Taheri Zadeh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Azadeh Aminzadeh
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahmoud Reza Heidari
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Joushi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
Salter DM, Wei W, Nahar PP, Marques E, Slitt AL. Perfluorooctanesulfonic Acid (PFOS) Thwarts the Beneficial Effects of Calorie Restriction and Metformin. Toxicol Sci 2021; 182:82-95. [PMID: 33844015 DOI: 10.1093/toxsci/kfab043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A combination of calorie restriction (CR), dietary modification, and exercise is the recommended therapy to reverse obesity and nonalcoholic fatty liver disease. In the liver, CR shifts hepatic metabolism from lipid storage to lipid utilization pathways, such as AMP-activated protein kinase (AMPK). Perfluorooctanesulfonic acid (PFOS), a fluorosurfactant previously used in stain repellents and anti-stick materials, can increase hepatic lipids in mice following relatively low-dose exposures. To test the hypothesis that PFOS administration interferes with CR, adult male C57BL/6N mice were fed ad libitum or a 25% reduced calorie diet concomitant with either vehicle (water) or 100 μg PFOS/kg/day via oral gavage for 6 weeks. CR alone improved hepatic lipids and glucose tolerance. PFOS did not significantly alter CR-induced weight loss, white adipose tissue mass, or liver weight over 6 weeks. However, PFOS increased hepatic triglyceride accumulation, in both mice fed ad libitum and subjected to CR. This was associated with decreased phosphorylated AMPK expression in liver. Glucagon (100 nM) treatment induced glucose production in hepatocytes, which was further upregulated with PFOS (2.5 μM) co-treatment. Next, to explore whether the observed changes were related to AMPK signaling, HepG2 cells were treated with metformin or AICAR alone or in combination with PFOS (25 μM). PFOS interfered with glucose-lowering effects of metformin, and AICAR treatment partially impaired PFOS-induced increase in glucose production. In 3T3-L1 adipocytes, metformin was less effective with PFOS co-treatment. Overall, PFOS administration disrupted hepatic lipid and glucose homeostasis and interfered with beneficial glucose-lowering effects of CR and metformin.
Collapse
Affiliation(s)
- Deanna M Salter
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Wei Wei
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Pragati P Nahar
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Emily Marques
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| |
Collapse
|
9
|
Azubuike-Osu SO, Famurewa AC, David JC, Abi I, Ogbu PN, Oparaji CK, Nwaeze KG, Akunna GG. Virgin Coconut Oil Resists Arsenic-Induced Cerebral Neurotoxicity and Cholesterol Imbalance via Suppression of Oxidative Stress, Adenosine Deaminase and Acetylcholinesterase Activities in Rats. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211016962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Arsenic (As) is a classic neurotoxicant; its pathogenesis is associated with oxidative stress and oxidative stress-mediated cholinergic deficits. This study explored antioxidant activity of virgin coconut oil (VCO) against sodium arsenite-induced oxidative stress-mediated cerebral neurotoxicity in rats. Eighteen rats were divided into 3 groups- Normal control, As control and VCO + As. The VCO (5 mL/kg) was given once daily by oral gavage from day 1 to day 21, while As (10 mg/kg) was given once daily by oral gavage from day 15 to day 21. Cerebral superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), malondialdehyde (MDA), adenosine deaminase (ADA) and acetylcholinesterase (AchE) activities were analysed. Nitric oxide (NO), lipid profile, phospholipid (PL), and reduced glutathione (GSH) were also evaluated in cerebral homogenate. The cerebrum was sectioned for histological analysis. Administration of As induced significant depressions in antioxidant enzymes, GSH, PL, and HDL-c compared to normal control. Levels of MDA, NO, total cholesterol and activities of ADA, AchE in the cerebrum were markedly increased by As compared to normal rats. Lipid profile indices and PL were prominently altered by As. Histopathological study supported the biochemical findings through extensive cerebral damage. In contrast, oral supplementation of VCO prior to and along with As treatment significantly attenuated the As-induced biochemical alterations and restored near-normal histology. VCO attenuates cerebral neurotoxicity by strengthening endogenous antioxidant defence and cholinergic function via counteracting free-radical-mediated arsenic toxicity.
Collapse
Affiliation(s)
- Sharon O. Azubuike-Osu
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Ademola C. Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Japheth C. David
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Innocent Abi
- Department of Physiology, Benue State University, Makurdi, Nigeria
| | - Patience N. Ogbu
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Chiedozie K. Oparaji
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Konyefom G. Nwaeze
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Godson G. Akunna
- Department of Anatomy, College of Medicine and Health Sciences, Bowen University, Nigeria
| |
Collapse
|
10
|
Li S, Zhang L, Huang R, Xu T, Parham F, Behl M, Xia M. Evaluation of chemical compounds that inhibit neurite outgrowth using GFP-labeled iPSC-derived human neurons. Neurotoxicology 2021; 83:137-145. [PMID: 33508353 PMCID: PMC9444042 DOI: 10.1016/j.neuro.2021.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/02/2020] [Accepted: 01/19/2021] [Indexed: 01/16/2023]
Abstract
Due to the increasing number of drugs and untested environmental compounds introduced into commercial use, there is recognition for a need to develop reliable and efficient screening methods to identify compounds that may adversely impact the nervous system. One process that has been implicated in neurodevelopment is neurite outgrowth; the disruption of which can result in adverse outcomes that persist later in life. Here, we developed a green fluorescent protein (GFP) labeled neurite outgrowth assay in a high-content, high-throughput format using induced pluripotent stem cell (iPSC) derived human spinal motor neurons and cortical glutamatergic neurons. The assay was optimized for use in a 1536-well plate format. Then, we used this assay to screen a set of 84 unique compounds that have previously been screened in other neurite outgrowth assays. This library consists of known developmental neurotoxicants, environmental compounds with unknown toxicity, and negative controls. Neurons were cultured for 40 h and then treated with compounds at 11 concentrations ranging from 1.56 nM to 92 μM for 24 and 48 h. Effects of compounds on neurite outgrowth were evaluated by quantifying total neurite length, number of segments, and maximum neurite length per cell. Among the 84 tested compounds, neurite outgrowth in cortical neurons and motor neurons were selectively inhibited by 36 and 31 compounds, respectively. Colchicine, rotenone, and methyl mercuric (II) chloride inhibited neurite outgrowth in both cortical and motor neurons. It is interesting to note that some compounds like parathion and bisphenol AF had inhibitory effects on neurite outgrowth specifically in the cortical neurons, while other compounds, such as 2,2',4,4'-tetrabromodiphenyl ether and caffeine, inhibited neurite outgrowth in motor neurons. The data gathered from these studies show that GFP-labeled iPSC-derived human neurons are a promising tool for identifying and prioritizing compounds with developmental neurotoxicity potential for further hazard characterization.
Collapse
Affiliation(s)
- Shuaizhang Li
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Li Zhang
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Ruili Huang
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Tuan Xu
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Fred Parham
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Mamta Behl
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA.
| | - Menghang Xia
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Kaur I, Behl T, Aleya L, Rahman MH, Kumar A, Arora S, Akter R. Role of metallic pollutants in neurodegeneration: effects of aluminum, lead, mercury, and arsenic in mediating brain impairment events and autism spectrum disorder. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:8989-9001. [PMID: 33447979 DOI: 10.1007/s11356-020-12255-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 12/27/2020] [Indexed: 04/16/2023]
Abstract
Autism spectrum disorder (ASD) is a developmental disorder of the brain characterized by shortfall in the social portfolio of an individual and abbreviated interactive and communication aspects rendering stereotypical behavior and pitfalls in a child's memory, thinking, and learning capabilities. The incidence of ASD has accelerated since the past decade, portraying environment as one of the primary assets, comprising of metallic components aiming to curb the neurodevelopmental pathways in an individual. Many regulations like Clean Air Act and critical steps taken by countries all over the globe, like Sweden and the USA, have rendered the necessity to study the effects of environmental metallic components on ASD progression. The review focuses on the primary metallic components present in the environment (aluminum, lead, mercury, and arsenic), responsible for accelerating ASD symptoms by a set of general mechanisms like oxidative stress reduction, glycolysis suppression, microglial activation, and metalloprotein disruption, resulting in apoptotic signaling, neurotoxic effects, and neuroinflammatory responses. The effect of these metals can be retarded by certain protective strategies like chelation, dietary correction, certain agents (curcumin, mangiferin, selenium), and detoxification enhancement, which can necessarily halt the neurodegenerative effects.
Collapse
Affiliation(s)
- Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Paris, France
| | - Md Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Seoul, South Korea
- Department of Pharmacy, Southeast University, Banani, Dhaka, Bangladesh
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Seoul, South Korea
| |
Collapse
|
12
|
Jiang M, Meng J, Zeng F, Qing H, Hook G, Hook V, Wu Z, Ni J. Cathepsin B inhibition blocks neurite outgrowth in cultured neurons by regulating lysosomal trafficking and remodeling. J Neurochem 2020; 155:300-312. [PMID: 32330298 PMCID: PMC7581626 DOI: 10.1111/jnc.15032] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 02/05/2023]
Abstract
Lysosomes are known to mediate neurite outgrowth in neurons. However, the principal lysosomal molecule controlling that outgrowth is unclear. We studied primary mouse neurons in vitro and found that they naturally develop neurite outgrowths over time and as they did so the lysosomal cysteine protease cathepsin B (CTSB) mRNA levels dramatically increased. Surprisingly, we found that treating those neurons with CA-074Me, which inhibits CTSB, prevented neurites. As that compound also inhibits another protease, we evaluated a N2a neuronal cell line in which the CTSB gene was deleted (CTSB knockout, KO) using CRISPR technology and induced their neurite outgrowth by treatment with retinoic acid. We found that CTSB KO N2a cells failed to produce neurite outgrowths but the wild-type (WT) did. CA-074Me is a cell permeable prodrug of CA-074, which is cell impermeable and a specific CTSB inhibitor. Neurite outgrowth was and was not suppressed in WT N2a cells treated with CA-074Me and CA-074, respectively. Lysosome-associated membrane glycoprotein 2-positive lysosomes traffic to the plasma cell membrane in WT but not in CTSB KO N2 a cells. Interestingly, no obvious differences between WT and CTSB KO N2a cells were found in neurite outgrowth regulatory proteins, PI3K/AKT, ERK/MAPK, cJUN, and CREB. These findings show that intracellular CTSB controls neurite outgrowth and that it does so through regulation of lysosomal trafficking and remodeling in neurons. This adds valuable information regarding the physiological function of CTSB in neural development.
Collapse
Affiliation(s)
- Muzhou Jiang
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Jie Meng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fan Zeng
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China
| | - Gregory Hook
- American Life Science Pharmaceuticals, La Jolla, CA, USA
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Junjun Ni
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China
| |
Collapse
|
13
|
David EE, Nwobodo V, Famurewa AC, Igwenyi IO, Egedeigwe-Ekeleme CA, Obeten UN, Obasi DO, Ezeilo UR, Emeribole MN. Effect of parboiling on toxic metal content and nutritional composition of three rice varieties locally produced in Nigeria. SCIENTIFIC AFRICAN 2020. [DOI: 10.1016/j.sciaf.2020.e00580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
14
|
McMichael BD, Perego MC, Darling CL, Perry RL, Coleman SC, Bain LJ. Long-term arsenic exposure impairs differentiation in mouse embryonal stem cells. J Appl Toxicol 2020; 41:1089-1102. [PMID: 33124703 DOI: 10.1002/jat.4095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 11/12/2022]
Abstract
Arsenic is a contaminant found in many foods and drinking water. Exposure to arsenic during development can cause improper neuronal progenitor cell development, differentiation, and function, while in vitro studies have determined that acute arsenic exposure to stem and progenitor cells reduced their ability to differentiate. In the current study, P19 mouse embryonal stem cells were exposed continuously to 0.1-μM (7.5 ppb) arsenic for 32 weeks. A cell lineage array examining messenger RNA (mRNA) changes after 8 and 32 weeks of exposure showed that genes involved in pluripotency were increased, whereas those involved in differentiation were reduced. Therefore, temporal changes of select pluripotency and neuronal differentiation markers throughout the 32-week chronic arsenic exposure were investigated. Sox2 and Oct4 mRNA expression were increased by 1.9- to 2.5-fold in the arsenic-exposed cells, beginning at Week 12. Sox2 protein expression was similarly increased starting at Week 16 and remained elevated by 1.5-fold to sixfold. One target of Sox2 is N-cadherin, whose expression is a hallmark of epithelial-mesenchymal transitions (EMTs). Exposure to arsenic significantly increased N-cadherin protein levels beginning at Week 20, concurrent with increased grouping of N-cadherin positive cells at the perimeter of the embryoid body. Expression of Zeb1, which helps increase the expression of Sox2, was also increased started at Week 16. In contrast, Gdf3 mRNA expression was reduced by 3.4- to 7.2-fold beginning at Week 16, and expression of its target protein, phospho-Smad2/3, was also reduced. These results suggest that chronic, low-level arsenic exposure may delay neuronal differentiation and maintain pluripotency.
Collapse
Affiliation(s)
- Benjamin D McMichael
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA.,US Environmental Protection Agency, Durham, North Carolina, USA
| | - M Chiara Perego
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Caitlin L Darling
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Rebekah L Perry
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Sarah C Coleman
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Lisa J Bain
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA.,Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
15
|
Mehta K, Kaur B, Pandey KK, Kaler S, Dhar P. Curcumin supplementation shows modulatory influence on functional and morphological features of hippocampus in mice subjected to arsenic trioxide exposure. Anat Cell Biol 2020; 53:355-365. [PMID: 32929054 PMCID: PMC7527119 DOI: 10.5115/acb.18.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 06/13/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022] Open
Abstract
Since, oxidative stress has been suggested as one of the mechanisms underlying arsenic-induced toxicity, the present study focused on the role of antioxidant (curcumin) supplementation on behavioral, biochemical, and morphological alterations with context to mice hippocampus (CA1) following arsenic trioxide (As2O3) administration. Healthy male Swiss albino mice were divided into control and experimental groups. As2O3 (2 mg/kg bw) alone or along with curcumin (100 mg/kg bw) was administered to experimental groups by oral route for 45 days whereas the control groups received either no treatment or vehicle for curcumin. Animals were subjected to behavioral study towards the end of the experimental period (day 33-45). On day 46, the brain samples were obtained and subjected either to immersion fixation (for morphometric observations) or used afresh for biochemical test. Behavioral tests (open field, elevated plus maze, and Morris water maze) revealed enhanced anxiety levels and impairment of cognitive functions in As2O3 alone treated groups whereas a trend of recovery was evident in mice simultaneously treated with As2O3 and curcumin. Morphological observations showed noticeable reduction in stratum pyramidale thickness (CA1), along with decrease in density and size of pyramidal neurons in As2O3 alone exposed group as compared to As2O3+Cu co-treated group. Hippocampal glutathione levels were found to be downregulated in animals receiving As2O3 as against the levels of controls and curcumin supplemented animals, thereby, suggestive of beneficial role of curcumin on As2O3 induced adverse effects.
Collapse
Affiliation(s)
- Kamakshi Mehta
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Balpreet Kaur
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Kamlesh Kumar Pandey
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Saroj Kaler
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Pushpa Dhar
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Patwa J, Flora SJS. Heavy Metal-Induced Cerebral Small Vessel Disease: Insights into Molecular Mechanisms and Possible Reversal Strategies. Int J Mol Sci 2020; 21:ijms21113862. [PMID: 32485831 PMCID: PMC7313017 DOI: 10.3390/ijms21113862] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Heavy metals are considered a continuous threat to humanity, as they cannot be eradicated. Prolonged exposure to heavy metals/metalloids in humans has been associated with several health risks, including neurodegeneration, vascular dysfunction, metabolic disorders, cancer, etc. Small blood vessels are highly vulnerable to heavy metals as they are directly exposed to the blood circulatory system, which has comparatively higher concentration of heavy metals than other organs. Cerebral small vessel disease (CSVD) is an umbrella term used to describe various pathological processes that affect the cerebral small blood vessels and is accepted as a primary contributor in associated disorders, such as dementia, cognitive disabilities, mood disorder, and ischemic, as well as a hemorrhagic stroke. In this review, we discuss the possible implication of heavy metals/metalloid exposure in CSVD and its associated disorders based on in-vitro, preclinical, and clinical evidences. We briefly discuss the CSVD, prevalence, epidemiology, and risk factors for development such as genetic, traditional, and environmental factors. Toxic effects of specific heavy metal/metalloid intoxication (As, Cd, Pb, Hg, and Cu) in the small vessel associated endothelium and vascular dysfunction too have been reviewed. An attempt has been made to highlight the possible molecular mechanism involved in the pathophysiology, such as oxidative stress, inflammatory pathway, matrix metalloproteinases (MMPs) expression, and amyloid angiopathy in the CSVD and related disorders. Finally, we discussed the role of cellular antioxidant defense enzymes to neutralize the toxic effect, and also highlighted the potential reversal strategies to combat heavy metal-induced vascular changes. In conclusion, heavy metals in small vessels are strongly associated with the development as well as the progression of CSVD. Chelation therapy may be an effective strategy to reduce the toxic metal load and the associated complications.
Collapse
|
17
|
Arsenic Toxicity: Molecular Targets and Therapeutic Agents. Biomolecules 2020; 10:biom10020235. [PMID: 32033229 PMCID: PMC7072575 DOI: 10.3390/biom10020235] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
High arsenic (As) levels in food and drinking water, or under some occupational conditions, can precipitate chronic toxicity and in some cases cancer. Millions of people are exposed to unacceptable amounts of As through drinking water and food. Highly exposed individuals may develop acute, subacute, or chronic signs of poisoning, characterized by skin lesions, cardiovascular symptoms, and in some cases, multi-organ failure. Inorganic arsenite(III) and organic arsenicals with the general formula R-As2+ are bound tightly to thiol groups, particularly to vicinal dithiols such as dihydrolipoic acid (DHLA), which together with some seleno-enzymes constitute vulnerable targets for the toxic action of As. In addition, R-As2+-compounds have even higher affinity to selenol groups, e.g., in thioredoxin reductase that also possesses a thiol group vicinal to the selenol. Inhibition of this and other ROS scavenging seleno-enzymes explain the oxidative stress associated with arsenic poisoning. The development of chelating agents, such as the dithiols BAL (dimercaptopropanol), DMPS (dimercapto-propanesulfonate) and DMSA (dimercaptosuccinic acid), took advantage of the fact that As had high affinity towards vicinal dithiols. Primary prevention by reducing exposure of the millions of people exposed to unacceptable As levels should be the prioritized strategy. However, in acute and subacute and even some cases with chronic As poisonings chelation treatment with therapeutic dithiols, in particular DMPS appears promising as regards alleviation of symptoms. In acute cases, initial treatment with BAL combined with DMPS should be considered.
Collapse
|
18
|
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: 2.8] [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
|
19
|
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: 1.4] [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
|
20
|
Chung YP, Yen CC, Tang FC, Lee KI, Liu SH, Wu CC, Hsieh SS, Su CC, Kuo CY, Chen YW. Methylmercury exposure induces ROS/Akt inactivation-triggered endoplasmic reticulum stress-regulated neuronal cell apoptosis. Toxicology 2019; 425:152245. [PMID: 31330229 DOI: 10.1016/j.tox.2019.152245] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/06/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have positively linked mercury exposure and neurodegenerative diseases (ND). Methylmercury (MeHg), an organic form of mercury, is a ubiquitous and potent environmental neurotoxicant that easily crosses the blood-brain barrier and causes irreversible injury to the central nervous system (CNS). However, the molecular mechanisms underlying MeHg-induced neurotoxicity remain unclear. Here, the present study found that Neuro-2a cells underwent apoptosis in response to MeHg (1-5 μM), which was accompanied by increased phosphatidylserine (PS) exposure on the outer cellular membrane leaflets, caspase-3 activity, and the activation of caspase cascades and poly (ADP-ribose) polymerase (PARP). Exposure of Neuro-2a cells to MeHg also triggered endoplasmic reticulum (ER) stress, which was identified via several key molecules (including: glucose-regulated protein (GRP)78, GRP94, C/EBP homologous protein (CHOP) X-box binding protein(XBP)-1, protein kinase R-like ER kinase (PERK), eukaryotic initiation factor 2α (eIF2α), inositol-requiring enzyme(IRE)-1, activation transcription factor(AFT)4, and ATF6. Transfection with GRP78-, GRP94-, CHOP-, and XBP-1-specific small interfering (si)RNA significantly suppressed the expression of these proteins, and attenuated cytotoxicity and caspase-12, -7, and -3 activation in MeHg-exposed cells. Furthermore, MeHg dramatically decreased Akt phosphorylation, and the overexpression of activation of Akt1 (myr-Akt1) could significantly prevent MeHg-induced Akt inactivation, as well as apoptotic and ER stress-related signals. Pretreatment with the antioxidant N-acetylcysteine (NAC) effectively prevented MeHg-induced neuronal cell reactive oxygen species (ROS) generation, apoptotic and ER stress-related signals, and Akt inactivation. Collectively, these results indicate that MeHg exerts its cytotoxicity in neurons by inducing ROS-mediated Akt inactivation up-regulated ER stress, which induces apoptosis and ultimately leads to cell death.
Collapse
Affiliation(s)
- Yao-Pang Chung
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Cheng-Chieh Yen
- Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung, 402, Taiwan; Department of Occupational Medicine, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Feng-Cheng Tang
- Department of Occupational Medicine, Changhua Christian Hospital, Changhua County, 500, Taiwan; Department of Leisure Services Management, Chaoyang University of Technology, Taichung, 413, Taiwan
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taichung, 427, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chin-Ching Wu
- Department of Public Health, China Medical University, Taichung, 404, Taiwan
| | - Shang-Shu Hsieh
- Department of Emergency, Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taichung, 427, Taiwan
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Chun-Ying Kuo
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Ya-Wen Chen
- Department of Physiology and Graduate Institute of Basic Medical Science, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
21
|
Ahmed RG, El-Gareib AW. Gestational Arsenic Trioxide Exposure Acts as a Developing Neuroendocrine-Disruptor by Downregulating Nrf2/PPARγ and Upregulating Caspase-3/NF-ĸB/Cox2/BAX/iNOS/ROS. Dose Response 2019; 17:1559325819858266. [PMID: 31258454 PMCID: PMC6589982 DOI: 10.1177/1559325819858266] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/15/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
The goal of this investigation was to evaluate the effects of gestational administrations of arsenic trioxide (ATO; As2O3) on fetal neuroendocrine development (the thyroid-cerebrum axis). Pregnant Wistar rats were orally administered ATO (5 or 10 mg/kg) from gestation day (GD) 1 to 20. Both doses of ATO diminished free thyroxine and free triiodothyronine levels and augmented thyrotropin level in both dams and fetuses at GD 20. Also, the maternofetal hypothyroidism in both groups caused a dose-dependent reduction in the fetal serum growth hormone, insulin growth factor-I (IGF-I), and IGF-II levels at embryonic day (ED) 20. These disorders perturbed the maternofetal body weight, fetal brain weight, and survival of pregnant and their fetuses. In addition, destructive degeneration, vacuolation, hyperplasia, and edema were observed in the fetal thyroid and cerebrum of both ATO groups at ED 20. These disruptions appear to depend on intensification in the values of lipid peroxidation, nitric oxide, and H2O2, suppression of messenger RNA (mRNA) expression of nuclear factor erythroid 2-related factor 2 and peroxisome proliferator-activated receptor gamma, and activation of mRNA expression of caspase-3, nuclear factor kappa-light-chain-enhancer of activated B cells, cyclooxygenase-2, Bcl-2–associated X protein, and inducible nitric oxide synthase in the fetal cerebrum. These data suggest that gestational ATO may disturb thyroid-cerebrum axis generating fetal neurodevelopmental toxicity.
Collapse
Affiliation(s)
- R G Ahmed
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - A W El-Gareib
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Cairo University, Egypt
| |
Collapse
|
22
|
Bjørklund G, Skalny AV, Rahman MM, Dadar M, Yassa HA, Aaseth J, Chirumbolo S, Skalnaya MG, Tinkov AA. Toxic metal(loid)-based pollutants and their possible role in autism spectrum disorder. ENVIRONMENTAL RESEARCH 2018; 166:234-250. [PMID: 29902778 DOI: 10.1016/j.envres.2018.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction, verbal and non-verbal communication, and stereotypic behaviors. Many studies support a significant relationship between many different environmental factors in ASD etiology. These factors include increased daily exposure to various toxic metal-based environmental pollutants, which represent a cause for concern in public health. This article reviews the most relevant toxic metals, commonly found, environmental pollutants, i.e., lead (Pb), mercury (Hg), aluminum (Al), and the metalloid arsenic (As). Additionally, it discusses how pollutants can be a possible pathogenetic cause of ASD through various mechanisms including neuroinflammation in different regions of the brain, fundamentally occurring through elevation of the proinflammatory profile of cytokines and aberrant expression of nuclear factor kappa B (NF-κB). Due to the worldwide increase in toxic environmental pollution, studies on the role of pollutants in neurodevelopmental disorders, including direct effects on the developing brain and the subjects' genetic susceptibility and polymorphism, are of utmost importance to achieve the best therapeutic approach and preventive strategies.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia
| | - Md Mostafizur Rahman
- Department of Environmental Sciences, Jahangirnagar University, Dhaka, Bangladesh; Graduate School of Environmental Science, Hokkaido University, Japan
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Heba A Yassa
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Jan Aaseth
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, Elverum, Norway; Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | | | - Alexey A Tinkov
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| |
Collapse
|
23
|
R G A, El-Gareib AW. WITHDRAWN: Toxic effects of gestational arsenic trioxide on the neuroendocrine axis of developing rats. Food Chem Toxicol 2018:S0278-6915(18)30663-X. [PMID: 30218683 DOI: 10.1016/j.fct.2018.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 11/19/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Ahmed R G
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - A W El-Gareib
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Cairo University, Egypt
| |
Collapse
|
24
|
Increased frequency of AMP-activated protein kinase-positive spinal motor neurons after sciatic nerve injury in a mouse model. Kaohsiung J Med Sci 2018; 34:301-306. [DOI: 10.1016/j.kjms.2017.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/24/2017] [Accepted: 12/11/2017] [Indexed: 12/13/2022] Open
|
25
|
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: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
26
|
Curtis TM, Hannett JM, Harman RM, Puoplo NA, Van de Walle GR. The secretome of adipose-derived mesenchymal stem cells protects SH-SY5Y cells from arsenic-induced toxicity, independent of a neuron-like differentiation mechanism. Neurotoxicology 2018; 67:54-64. [PMID: 29660375 DOI: 10.1016/j.neuro.2018.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/15/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023]
Abstract
Arsenic exposure through contaminated food, water, and air causes irreversible neural damage and affects millions of people worldwide. Several studies have demonstrated that the secreted factors (secretome) from mesenchymal stromal/stem cells (MSCs) can promote neural recovery after several forms of injury including stroke and neurodegenerative diseases. The present study was conducted to determine if the secretome from adipose-derived MSCs (ADSCs) prevents arsenic damage to SH-SY5Y cells. To this end, human neuroblastoma cells (SH-SY5Y) were pre-treated with the secretome from ADSCs and then challenged with different concentrations of arsenic. After various doses and exposure times, the extent of neuronal injury was assessed using MTT reduction and LDH release assays as well as LIVE/DEAD staining. These data demonstrate that the ADSC secretome protects SH-SY5Y cells from arsenic-induced toxicity. Previous reports have shown that the secretome of MSCs can induce neuroblast differentiation and mature neurons are less susceptible to chemical-induced toxicity. In the current study, proliferation assays, neurite length assessment, and quantitative RT-PCR of differentiation markers indicated that the ADSC secretome does not induce SH-SY5Y differentiation into a mature neuron-like phenotype. In contrast, our results demonstrated that soluble factor(s) in the ADSC secretome enhance SH-SY5Y cell substrate-dependent adhesion. The present study is the first to illustrate that the secretome from ADSCs protects SH-SY5Y cells from arsenic-induced toxicity. Additionally, we showed that protection against arsenic toxicity is not dependent on SH-SY5Y cell differentiation into a mature neuron-like phenotype, but involves soluble factor(s) in the secretome that appear to enhance cell survival by an adhesion-dependent mechanism.
Collapse
Affiliation(s)
- Theresa M Curtis
- Department of Biological Sciences, State University of New York at Cortland, Cortland, NY, United States.
| | - Joseph M Hannett
- Department of Biological Sciences, State University of New York at Cortland, Cortland, NY, United States
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Nicholas A Puoplo
- Department of Biological Sciences, State University of New York at Cortland, Cortland, NY, United States
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
27
|
Garza-Lombó C, Schroder A, Reyes-Reyes EM, Franco R. mTOR/AMPK signaling in the brain: Cell metabolism, proteostasis and survival. CURRENT OPINION IN TOXICOLOGY 2018; 8:102-110. [PMID: 30417160 PMCID: PMC6223325 DOI: 10.1016/j.cotox.2018.05.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The mechanistic (or mammalian) target of rapamycin (mTOR) and the adenosine monophosphate-activated protein kinase (AMPK) regulate cell survival and metabolism in response to diverse stimuli such as variations in amino acid content, changes in cellular bioenergetics, oxygen levels, neurotrophic factors and xenobiotics. This Opinion paper aims to discuss the current state of knowledge regarding how mTOR and AMPK regulate the metabolism and survival of brain cells and the close interrelationship between both signaling cascades. It is now clear that both mTOR and AMPK pathways regulate cellular homeostasis at multiple levels. Studies so far demonstrate that dysregulation in these two pathways is associated with neuronal injury, degeneration and neurotoxicity, but the mechanisms involved remain unclear. Most of the work so far has been focused on their antagonistic regulation of autophagy, but recent findings highlight that changes in protein synthesis, metabolism and mitochondrial function are likely to play a role in the regulatory effects of both mTOR and AMPK on neuronal health. Understanding the role and relationship between these two master regulators of cell metabolism is crucial for future therapeutic approaches to counteract alterations in cell metabolism and survival in brain injury and disease.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Redox Biology Center. University of Nebraska-Lincoln, Lincoln, NE 68588
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México 04510
| | - Annika Schroder
- Redox Biology Center. University of Nebraska-Lincoln, Lincoln, NE 68588
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Elsa M. Reyes-Reyes
- University of Arizona College of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Tucson, AZ 85724
| | - Rodrigo Franco
- Redox Biology Center. University of Nebraska-Lincoln, Lincoln, NE 68588
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| |
Collapse
|
28
|
Liu JT, Bain LJ. Arsenic Induces Members of the mmu-miR-466-669 Cluster Which Reduces NeuroD1 Expression. Toxicol Sci 2018; 162:64-78. [PMID: 29121352 PMCID: PMC6693399 DOI: 10.1093/toxsci/kfx241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Chronic arsenic exposure can result in adverse development effects including decreased intellectual function, reduced birth weight, and altered locomotor activity. Previous in vitro studies have shown that arsenic inhibits stem cell differentiation. MicroRNAs (miRNAs) are small noncoding RNAs that regulate multiple cellular processes including embryonic development and cell differentiation. The purpose of this study was to examine whether altered miRNA expression was a mechanism by which arsenic inhibited cellular differentiation. The pluripotent P19 mouse embryonal carcinoma cells were exposed to 0 or 0.5 μM sodium arsenite for 9 days during cell differentiation, and changes in miRNA expression was analyzed using microarrays. We found that the expression of several miRNAs important in cellular differentiation, such as miR-9 and miR-199 were decreased by 1.9- and 1.6-fold, respectively, following arsenic exposure, while miR-92a, miR-291a, and miR-709 were increased by 3-, 3.7-, and 1.6-fold, respectively. The members of the miR-466-669 cluster and its host gene, Scm-like with 4 Mbt domains 2 (Sfmbt2), were significantly induced by arsenic from 1.5- to 4-fold in a time-dependent manner. Multiple miRNA target prediction programs revealed that several neurogenic transcription factors appear to be targets of the cluster. When consensus anti-miRNAs targeting the miR-466-669 cluster were transfected into P19 cells, arsenic-exposed cells were able to more effectively differentiate. The consensus anti-miRNAs appeared to rescue the inhibitory effects of arsenic on cell differentiation due to an increased expression of NeuroD1. Taken together, we conclude that arsenic induces the miR-466-669 cluster, and that this induction acts to inhibit cellular differentiation in part due to a repression of NeuroD1.
Collapse
Affiliation(s)
| | - Lisa J Bain
- Environmental Toxicology Graduate Program
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634
| |
Collapse
|
29
|
The Adiponectin Homolog Osmotin Enhances Neurite Outgrowth and Synaptic Complexity via AdipoR1/NgR1 Signaling in Alzheimer’s Disease. Mol Neurobiol 2018; 55:6673-6686. [DOI: 10.1007/s12035-017-0847-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/19/2017] [Indexed: 01/05/2023]
|
30
|
Analyzing the effects of estuarine freshwater fluxes on fish abundance using artificial neural network ensembles. Ecol Modell 2017. [DOI: 10.1016/j.ecolmodel.2017.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
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: 1.9] [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
|
32
|
Chan MC, Bautista E, Alvarado-Cruz I, Quintanilla-Vega B, Segovia J. Inorganic mercury prevents the differentiation of SH-SY5Y cells: Amyloid precursor protein, microtubule associated proteins and ROS as potential targets. J Trace Elem Med Biol 2017; 41:119-128. [PMID: 28209268 DOI: 10.1016/j.jtemb.2017.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/16/2017] [Accepted: 02/05/2017] [Indexed: 10/20/2022]
Abstract
Exposure to mercury (Hg) occurs through different pathways and forms including methylmecury (MeHg) from seafood and rice, ethylmercury (EtHg), and elemental Hg (Hg0) from dental amalgams and artisanal gold mining. Once in the brain all these forms are transformed to inorganic Hg (I-Hg), where it bioaccumulates and remains for long periods. Hg is a well-known neurotoxicant, with its most damaging effects reported during brain development, when cellular key events, such as cell differentiation take place. A considerable number of studies report an impairment of neuronal differentiation due to MeHg exposure, however the effects of I-Hg, an important form of Hg found in brain, have received less attention. In this study, we decided to examine the effects of I-Hg exposure (5, 10 and 20μM) on the differentiation of SH-SY5Y cells induced by retinoic acid (RA, 10μM). We observed extension of neuritic processes and increased expression of neuronal markers (MAP2, tubulin-βIII, and Tau) after RA stimulation, all these effects were decreased by the co-exposure to I-Hg. Interestingly, I-Hg increased the levels of reactive oxygen species (ROS) and nitric oxide (NO) accompanied with increased levels of inducible nitric oxide synthase (iNOS) and, dimethylarginine dimethylaminohydrolase 1 (DDHA1). Remarkably I-Hg decreased levels of nitric oxide synthase neuronal (nNOS). Moreover I-Hg reduced the levels of tyrosine hydroxylase (TH) and amyloid precursor protein (APP) a protein recently involved in neuronal differentiation. These data suggest that the exposure to I-Hg impairs cell differentiation, and point to new potential targets of Hg toxicity such as APP and NO signaling.
Collapse
Affiliation(s)
- Miguel Chin Chan
- Departmento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico; Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Campeche, Campeche 4039, Mexico
| | - Elizabeth Bautista
- Departmento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico; Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, 52786, Huixquilucan, Mexico
| | - Isabel Alvarado-Cruz
- Departmento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico
| | - Betzabet Quintanilla-Vega
- Departmento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico
| | - José Segovia
- Departmento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico.
| |
Collapse
|
33
|
Zhou LT, Ye SH, Yang HX, Zhou YT, Zhao QH, Sun WW, Gao MM, Yi YH, Long YS. A novel role of fragile X mental retardation protein in pre-mRNA alternative splicing through RNA-binding protein 14. Neuroscience 2017; 349:64-75. [PMID: 28257890 DOI: 10.1016/j.neuroscience.2017.02.044] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 02/08/2023]
Abstract
Fragile X mental retardation protein (FMRP), an important RNA-binding protein responsible for fragile X syndrome, is involved in posttranscriptional control of gene expression that links with brain development and synaptic functions. Here, we reveal a novel role of FMRP in pre-mRNA alternative splicing, a general event of posttranscriptional regulation. Using co-immunoprecipitation and immunofluorescence assays, we identified that FMRP interacts with an alternative-splicing-associated protein RNA-binding protein 14 (RBM14) in a RNA-dependent fashion, and the two proteins partially colocalize in the nuclei of hippocampal neurons. We show that the relative skipping/inclusion ratio of the micro-exon L in the Protrudin gene and exon 10 in the Tau gene decreased in the hippocampus of Fmr1 knockout (KO) mice. Knockdown of either FMRP or RBM14 alters the relative skipping/inclusion ratio of Protrudin and Tau in cultured Neuro-2a cells, similar to that in the Fmr1 KO mice. Furthermore, overexpression of FMRP leads to an opposite pattern of the splicing, which can be offset by RBM14 knockdown. RNA immunoprecipitation assays indicate that FMRP promotes RBM14's binding to the mRNA targets. In addition, overexpression of the long form of Protrudin or the short form of Tau promotes protrusion growth of the retinoic acid-treated, neuronal-differentiated Neuro-2a cells. Together, these data suggest a novel function of FMRP in the regulation of pre-mRNA alternative splicing through RBM14 that may be associated with normal brain function and FMRP-related neurological disorders.
Collapse
Affiliation(s)
- Lin-Tao Zhou
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Shun-Hua Ye
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Hai-Xuan Yang
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Yong-Ting Zhou
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Qi-Hua Zhao
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Wei-Wen Sun
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Mei-Mei Gao
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Yong-Hong Yi
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| | - Yue-Sheng Long
- Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China.
| |
Collapse
|
34
|
Ameer SS, Engström K, Hossain MB, Concha G, Vahter M, Broberg K. Arsenic exposure from drinking water is associated with decreased gene expression and increased DNA methylation in peripheral blood. Toxicol Appl Pharmacol 2017; 321:57-66. [PMID: 28242323 DOI: 10.1016/j.taap.2017.02.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/03/2017] [Accepted: 02/22/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Exposure to inorganic arsenic increases the risk of cancer and non-malignant diseases. Inefficient arsenic metabolism is a marker for susceptibility to arsenic toxicity. Arsenic may alter gene expression, possibly by altering DNA methylation. OBJECTIVES To elucidate the associations between arsenic exposure, gene expression, and DNA methylation in peripheral blood, and the modifying effects of arsenic metabolism. METHODS The study participants, women from the Andes, Argentina, were exposed to arsenic via drinking water. Arsenic exposure was assessed as the sum of arsenic metabolites in urine (U-As), using high performance liquid-chromatography hydride-generation inductively-coupled-plasma-mass-spectrometry, and arsenic metabolism efficiency was assessed by the urinary fractions (%) of the individual metabolites. Genome-wide gene expression (N=80 women) and DNA methylation (N=93; 80 overlapping with gene expression) in peripheral blood were measured using Illumina DirectHyb HumanHT-12 v4.0 and Infinium Human-Methylation 450K BeadChip, respectively. RESULTS U-As concentrations, ranging 10-1251μg/L, was associated with decreased gene expression: 64% of the top 1000 differentially expressed genes were down-regulated with increasing U-As. U-As was also associated with hypermethylation: 87% of the top 1000CpGs were hypermethylated with increasing U-As. The expression of six genes and six individual CpG sites were significantly associated with increased U-As concentration. Pathway analyses revealed enrichment of genes related to cell death and cancer. The pathways differed somewhat depending on arsenic metabolism efficiency. We found no overlap between arsenic-related gene expression and DNA methylation for individual genes. CONCLUSIONS Increased arsenic exposure was associated with lower gene expression and hypermethylation in peripheral blood, but with no evident overlap.
Collapse
Affiliation(s)
- Syeda Shegufta Ameer
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Karin Engström
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden; Institute of Environmental Medicine, Unit of Metals & Health, Karolinska Institutet, Stockholm, Sweden
| | - Mohammad Bakhtiar Hossain
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Gabriela Concha
- Science Department, Risk Benefit Assessment Unit, National Food Agency, Uppsala, Sweden
| | - Marie Vahter
- Institute of Environmental Medicine, Unit of Metals & Health, Karolinska Institutet, Stockholm, Sweden
| | - Karin Broberg
- Institute of Environmental Medicine, Unit of Metals & Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
35
|
Bai L, Mei X, Shen Z, Bi Y, Yuan Y, Guo Z, Wang H, Zhao H, Zhou Z, Wang C, Zhu K, Li G, Lv G. Netrin-1 Improves Functional Recovery through Autophagy Regulation by Activating the AMPK/mTOR Signaling Pathway in Rats with Spinal Cord Injury. Sci Rep 2017; 7:42288. [PMID: 28186165 PMCID: PMC5301251 DOI: 10.1038/srep42288] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/04/2017] [Indexed: 01/08/2023] Open
Abstract
Autophagy is an process for the degradation of cytoplasmic aggregated proteins and damaged organelles and plays an important role in the development of SCI. In this study, we investigated the therapeutic effect of Netrin-1 and its potential mechanism for autophagy regulation after SCI. A rat model of SCI was established and used for analysis. Results showed that administration of Netrin-1 not only significantly enhanced the phosphorylation of AMP-activated protein kinase (AMPK) but also reduced the phosphorylation of mammalian target of rapamycin (mTOR) and P70S6K. In addition, the expression of Beclin-1 and the ratio of the light-chain 3B-II (LC3B-II)/LC3B-I in the injured spinal cord significantly increased in Netrin-1 group than those in SCI group. Moreover, the ratio of apoptotic neurons in the anterior horn of the spinal cord and the cavity area of spinal cord significantly decreased in Netrin-1 group compared with those in SCI group. In addition, Netrin-1 not only preserved motor neurons but also significantly improved motor fuction of injured rats. These results suggest that Netrin-1 improved functional recovery through autophagy stimulation by activating the AMPK/mTOR signaling pathway in rats with SCI. Thus, Netrin-1 treatment could be a novel therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Liangjie Bai
- Department of Orthopedics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xifan Mei
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Zhaoliang Shen
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Yunlong Bi
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Yajiang Yuan
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Zhanpeng Guo
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Hongyu Wang
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Haosen Zhao
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Zipeng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Chen Wang
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Kunming Zhu
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Gang Li
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, China
| | - Gang Lv
- Department of Orthopedics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
36
|
Tyler CR, Labrecque MT, Solomon ER, Guo X, Allan AM. Prenatal arsenic exposure alters REST/NRSF and microRNA regulators of embryonic neural stem cell fate in a sex-dependent manner. Neurotoxicol Teratol 2016; 59:1-15. [PMID: 27751817 DOI: 10.1016/j.ntt.2016.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/30/2016] [Accepted: 10/13/2016] [Indexed: 11/29/2022]
Abstract
Exposure to arsenic, a common environmental toxin found in drinking water, leads to a host of neurological pathologies. We have previously demonstrated that developmental exposure to a low level of arsenic (50ppb) alters epigenetic processes that underlie deficits in adult hippocampal neurogenesis leading to aberrant behavior. It is unclear if arsenic impacts the programming and regulation of embryonic neurogenesis during development when exposure occurs. The master negative regulator of neural-lineage, REST/NRSF, controls the precise timing of fate specification and differentiation of neural stem cells (NSCs). Early in development (embryonic day 14), we observed increased expression of Rest, its co-repressor, CoREST, and the inhibitory RNA binding/splicing protein, Ptbp1, and altered expression of mRNA spliced isoforms of Pbx1 that are directly regulated by these factors in the male brain in response to prenatal 50ppb arsenic exposure. These increases were concurrent with decreased expression of microRNA-9 (miR-9), miR-9*, and miR-124, all of which are REST/NRSF targets and inversely regulate Rest expression to allow for maturation of NSCs. Exposure to arsenic decreased the formation of neuroblasts in vitro from NSCs derived from male pup brains. The female response to arsenic was limited to increased expression of CoREST and Ptbp2, an RNA binding protein that allows for appropriate splicing of genes involved in the progression of neurogenesis. These changes were accompanied by increased neuroblast formation in vitro from NSCs derived from female pups. Unexposed male mice express transcriptomic factors to induce differentiation earlier in development compared to unexposed females. Thus, arsenic exposure likely delays differentiation of NSCs in males while potentially inducing precocious differentiation in females early in development. These effects are mitigated by embryonic day 18 of development. Arsenic-induced dysregulation of the regulatory loop formed by REST/NRSF, its target microRNAs, miR-9 and miR-124, and RNA splicing proteins, PTBP1 and 2, leads to aberrant programming of NSC function that is perhaps perpetuated into adulthood inducing deficits in differentiation we have previously observed.
Collapse
Affiliation(s)
- Christina R Tyler
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, United States
| | - Matthew T Labrecque
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Elizabeth R Solomon
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Xun Guo
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Andrea M Allan
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
37
|
Kim HY, Wegner SH, Van Ness KP, Park JJ, Pacheco SE, Workman T, Hong S, Griffith W, Faustman EM. Differential epigenetic effects of chlorpyrifos and arsenic in proliferating and differentiating human neural progenitor cells. Reprod Toxicol 2016; 65:212-223. [PMID: 27523287 DOI: 10.1016/j.reprotox.2016.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 07/21/2016] [Accepted: 08/10/2016] [Indexed: 12/16/2022]
Abstract
Understanding the underlying temporal and mechanistic responses to neurotoxicant exposures during sensitive periods of neuronal development are critical for assessing the impact of these exposures on developmental processes. To investigate the importance of timing of neurotoxicant exposure for perturbation of epigenetic regulation, we exposed human neuronal progenitor cells (hNPCs) to chlorpyrifos (CP) and sodium arsenite (As; positive control) during proliferation and differentiation. CP or As treatment effects on hNPCs morphology, cell viability, and changes in protein expression levels of neural differentiation and cell stress markers, and histone H3 modifications were examined. Cell viability, proliferation/differentiation status, and epigenetic results suggest that hNPCs cultures respond to CP and As treatment with different degrees of sensitivity. Histone modifications, as measured by changes in histone H3 phosphorylation, acetylation and methylation, varied for each toxicant and growth condition, suggesting that differentiation status can influence the epigenetic effects of CP and As exposures.
Collapse
Affiliation(s)
- Hee Yeon Kim
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Susanna H Wegner
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Kirk P Van Ness
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Julie Juyoung Park
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Sara E Pacheco
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Tomomi Workman
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Sungwoo Hong
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - William Griffith
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States
| | - Elaine M Faustman
- Department of Environmental and Occupational Health, Institute of Risk Analysis and Risk Communication, University of Washington, 4225 Roosevelt Way NE, Seattle, WA, United States.
| |
Collapse
|
38
|
Hijacking microglial glutathione by inorganic arsenic impels bystander death of immature neurons through extracellular cystine/glutamate imbalance. Sci Rep 2016; 6:30601. [PMID: 27477106 PMCID: PMC4967897 DOI: 10.1038/srep30601] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/04/2016] [Indexed: 12/21/2022] Open
Abstract
Arsenic-induced altered microglial activity leads to neuronal death, but the causative mechanism remains unclear. The present study showed, arsenic-exposed (10 μM) microglial (N9) culture supernatant induced bystander death of neuro-2a (N2a), which was further validated with primary microglia and immature neuronal cultures. Results indicated that arsenic-induced GSH synthesis by N9 unfavorably modified the extracellular milieu for N2a by lowering cystine and increasing glutamate concentration. Similar result was observed in N9-N2a co-culture. Co-exposure of arsenic and 250 μM glutamate, less than the level (265 μM) detected in arsenic-exposed N9 culture supernatant, compromised N2a viability which was rescued by cystine supplementation. Therefore, microglia executes bystander N2a death by competitive inhibition of system Xc- (xCT) through extracellular cystine/glutamate imbalance. We confirmed the role of xCT in mediating bystander N2a death by siRNA inhibition studies. Ex-vivo primary microglia culture supernatant from gestationally exposed mice measured to contain lower cystine and higher glutamate compared to control and N-acetyl cysteine co-treated group. Immunofluorescence staining of brain cryosections from treated group showed more dead immature neurons with no such effect on microglia. Collectively, we showed, in presence of arsenic microglia alters cystine/glutamate balance through xCT in extracellular milieu leading to bystander death of immature neurons.
Collapse
|
39
|
Doolaanea AA, Mansor N'I, Mohd Nor NH, Mohamed F. Co-encapsulation ofNigella sativaoil and plasmid DNA for enhanced gene therapy of Alzheimer’s disease. J Microencapsul 2016; 33:114-26. [DOI: 10.3109/02652048.2015.1134689] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
40
|
Takser L, Benachour N, Husk B, Cabana H, Gris D. Cyanotoxins at low doses induce apoptosis and inflammatory effects in murine brain cells: Potential implications for neurodegenerative diseases. Toxicol Rep 2016; 3:180-189. [PMID: 28959538 PMCID: PMC5615428 DOI: 10.1016/j.toxrep.2015.12.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 12/17/2015] [Accepted: 12/24/2015] [Indexed: 11/25/2022] Open
Abstract
Cyanotoxins have been shown to be highly toxic for mammalian cells, including brain cells. However, little is known about their effect on inflammatory pathways. This study investigated whether mammalian brain and immune cells can be a target of certain cyanotoxins, at doses approximating those in the guideline levels for drinking water, either alone or in mixtures. We examined the effects on cellular viability, apoptosis and inflammation signalling of several toxins on murine macrophage-like RAW264.7, microglial BV-2 and neuroblastoma N2a cell lines. We tested cylindrospermopsin (CYN), microcystin-LR (MC-LR), and anatoxin-a (ATX-a), individually as well as their mixture. In addition, we studied the neurotoxins β-N-methylamino-l-alanine (BMAA) and its isomer 2,4-diaminobutyric acid (DAB), as well as the mixture of both. Cellular viability was determined by the MTT assay. Apoptosis induction was assessed by measuring the activation of caspases 3/7. Cell death and inflammation are the hallmarks of neurodegenerative diseases. Thus, our final step was to quantify the expression of a major proinflammatory cytokine TNF-α by ELISA. Our results show that CYN, MC-LR and ATX-a, but not BMAA and DAB, at low doses, especially when present in a mixture at threefold less concentrations than individual compounds are 3–15 times more potent at inducing apoptosis and inflammation. Our results suggest that common cyanotoxins at low doses have a potential to induce inflammation and apoptosis in immune and brain cells. Further research of the neuroinflammatory effects of these compounds in vivo is needed to improve safety limit levels for cyanotoxins in drinking water and food.
Collapse
Affiliation(s)
- Larissa Takser
- Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, J1H 5N4 Quebec, Canada
| | - Nora Benachour
- Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, J1H 5N4 Quebec, Canada
| | - Barry Husk
- BlueLeaf Inc., 310 Chapleau Street, Drummondville, J2B 5E9 Quebec, Canada
| | - Hubert Cabana
- Environmental Engineering Laboratory, Department of Civil Engineering, University of Sherbrooke, Sherbrooke, J1K 2R1 Quebec, Canada
| | - Denis Gris
- Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, J1H 5N4 Quebec, Canada
| |
Collapse
|
41
|
Bain LJ, Liu JT, League RE. Arsenic inhibits stem cell differentiation by altering the interplay between the Wnt3a and Notch signaling pathways. Toxicol Rep 2016; 3:405-413. [PMID: 27158593 PMCID: PMC4855706 DOI: 10.1016/j.toxrep.2016.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
data indicates that arsenic exposure inhibits stem cell differentiation. This study investigated whether arsenic disrupted the Wnt3a signaling pathway, critical in the formation of myotubes and neurons, during the differentiation in P19 mouse embryonic stem cells. Cells were exposed to 0, 0.1, or 0.5 μM arsenite, with or without exogenous Wnt3a, for up to 9 days of differentiation. Arsenic exposure alone inhibits the differentiation of stem cells into neurons and skeletal myotubes, and reduces the expression of both β-catenin and GSK3β mRNA to ~55% of control levels. Co-culture of the arsenic-exposed cells with exogenous Wnt3a rescues the morphological phenotype, but does not alter transcript, protein, or phosphorylation status of GSK3β or β-catenin. However, arsenic exposure maintains high levels of Hes5 and decreases the expression of MASH1 by 2.2-fold, which are anti- and pro-myogenic and neurogenic genes, respectively, in the Notch signaling pathway. While rescue with exogenous Wnt3a reduced Hes5 levels, MASH1 levels stay repressed. Thus, while Wnt3a can partially rescue the inhibition of differentiation from arsenic, it does so by also modulating Notch target genes rather than only working through the canonical Wnt signaling pathway. These results indicate that arsenic alters the interplay between multiple signaling pathways, leading to reduced stem cell differentiation.
Collapse
Affiliation(s)
- Lisa J Bain
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA; Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 23964, USA
| | - Jui-Tung Liu
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Ryan E League
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| |
Collapse
|
42
|
Feng S, Lu L, Zan X, Wu Y, Lin Y, Wang Q. Genetically Engineered Plant Viral Nanoparticles Direct Neural Cells Differentiation and Orientation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:9402-9409. [PMID: 26247572 DOI: 10.1021/acs.langmuir.5b02226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
An important aim of tissue engineering is to design biomimetic materials with specific cell binding motifs and precisely controlled structural organization, thereby providing biochemical and physical cues for desired cellular behaviors. Previously, our group generated genetically modified tobacco mosaic virus (TMV) displaying integrin binding motifs, RGD1, RGD7, PSHRN3, P15, and DGEA. The resulting rod-like virus particles displaying integrin binding motifs were biocompatible with Neuro 2A (N2a), a mouse neural crest-derived cell line, and could promote the neurite outgrowth of N2a. The genetically modified viruses could be assembled with aligned orientation in the capillary by applying a shear force. The resulting aligned substrates were able to dictate directional neurite outgrowth of N2a cells. Therefore, this method could be potentially applied for neural tissue engineering, as a neural conduit for repairing peripheral nerve injuries.
Collapse
Affiliation(s)
- Sheng Feng
- Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina 29208, United States
| | - Lin Lu
- Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina 29208, United States
| | - Xingjie Zan
- Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina 29208, United States
| | - Yehong Wu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, P. R. China
| | - Yuan Lin
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, P. R. China
| | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina 29208, United States
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, P. R. China
| |
Collapse
|
43
|
Feng X, Yu W, Liang R, Shi C, Zhao Z, Guo J. Receptor-interacting protein 140 overexpression promotes neuro-2a neuronal differentiation by ERK1/2 signaling. Chin Med J (Engl) 2015; 128:119-24. [PMID: 25563324 PMCID: PMC4837806 DOI: 10.4103/0366-6999.147850] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background: Abnormal neuronal differentiation plays an important role in central nervous system (CNS) development abnormalities such as Down syndrome (DS), a disorder that results directly from overexpression of genes in trisomic cells. Receptor-interacting protein 140 (RIP140) is significantly upregulated in DS brains, suggesting its involvement in DS CNS development abnormalities. However, the role of RIP140 in neuronal differentiation is still not clear. The current study aimed to investigate the effect of RIP140 overexpression on the differentiation of neuro-2a (N2a) neuroblastoma cells, in vitro. Methods: Stably RIP140-overexpressing N2a (N2a-RIP140) cells were used as a neurodevelopmental model, and were constructed by lipofection and overexpression validated by real-time polymerase chain reaction and Western blot. Retinoic acid (RA) was used to stimulate N2a differentiation. Combining the expression of Tuj1 at the mRNA and protein levels, the percentage of cells baring neurites, and the number of neurites per cell body was semi-quantified to determine the effect of RIP140 on differentiation of N2a cells. Furthermore, western blot and the ERK1/2 inhibitor U0126 were used to identify the specific signaling pathway by which RIP140 induces differentiation of N2a cells. Statistical significance of the differences between groups was determined by one-way analysis of variance followed by the Dunnett test. Results: Compared to untransfected N2a cells RIPl40 expression in N2a-RIP140 cells was remarkably upregulated at both the mRNA and protein levels. N2a-RIP140 cells had a significantly increased percentage of cells baring neurites, and numbers of neurites per cell, as compared to N2a cells, in the absence and presence of RA (P < 0.05). In addition, Tuj1, a neuronal biomarker, was strongly upregulated in N2a-RIP140 cells (P < 0.05) and phosphorylated ERK1/2 (p-ERK1/2) levels in N2a-RIP140 cells were dramatically increased, while differentiation was inhibited by the ERK1/2-specific inhibitor U0126. Conclusions: RIP140 overexpression promotes N2a cell neuronal differentiation by activating the ERK1/2 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Jingzhu Guo
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
44
|
McCoy CR, Stadelman BS, Brumaghim JL, Liu JT, Bain LJ. Arsenic and Its Methylated Metabolites Inhibit the Differentiation of Neural Plate Border Specifier Cells. Chem Res Toxicol 2015; 28:1409-21. [PMID: 26024302 DOI: 10.1021/acs.chemrestox.5b00036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Exposure to arsenic in food and drinking water has been correlated with adverse developmental outcomes, such as reductions in birth weight and neurological deficits. Additionally, studies have shown that arsenic suppresses sensory neuron formation and skeletal muscle myogenesis, although the reason why arsenic targets both of these cell types in unclear. Thus, P19 mouse embryonic stem cells were used to investigate the mechanisms by which arsenic could inhibit cellular differentiation. P19 cells were exposed to 0, 0.1, or 0.5 μM sodium arsenite and induced to form embryoid bodies over a period of 5 days. The expression of transcription factors necessary to form neural plate border specifier (NPBS) cells, neural crest cells and their progenitors, and myocytes and their progenitors were examined. Early during differentiation, arsenic significantly reduced the transcript and protein expression of Msx1 and Pax3, both needed for NPBS cell formation. Arsenic also significantly reduced the protein expression of Sox 10, needed for neural crest progenitor cell production, by 31-50%, and downregulated the protein and mRNA levels of NeuroD1, needed for neural crest cell differentiation, in a time- and dose-dependent manner. While the overall protein expression of transcription factors in the skeletal muscle lineage was not changed, arsenic did alter their nuclear localization. MyoD nuclear translocation was significantly reduced on days 2-5 between 15 and 70%. At a 10-fold lower concentration, monomethylarsonous acid (MMA III) appeared to be just as potent as inorganic arsenic at reducing the mRNA levels Pax3 (79% vs84%), Sox10 (49% vs 65%), and Msx1 (56% vs 56%). Dimethylarsinous acid (DMA III) also reduced protein and transcript expression, but the changes were less dramatic than those with MMA or arsenite. All three arsenic species reduced the nuclear localization of MyoD and NeuroD1 in a similar manner. The early changes in the differentiation of neural plate border specifier cells may provide a mechanism for arsenic to suppress both neurogenesis and myogenesis.
Collapse
Affiliation(s)
- Christopher R McCoy
- †Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States
| | - Bradley S Stadelman
- ‡Department of Chemistry, Clemson University, 219 Hunter Laboratories, Clemson, South Carolina 29634, United States
| | - Julia L Brumaghim
- ‡Department of Chemistry, Clemson University, 219 Hunter Laboratories, Clemson, South Carolina 29634, United States
| | - Jui-Tung Liu
- §Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States
| | - Lisa J Bain
- †Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States.,§Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States
| |
Collapse
|
45
|
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: 0.8] [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
|
46
|
Zogovic N, Tovilovic-Kovacevic G, Misirkic-Marjanovic M, Vucicevic L, Janjetovic K, Harhaji-Trajkovic L, Trajkovic V. Coordinated activation of AMP-activated protein kinase, extracellular signal-regulated kinase, and autophagy regulates phorbol myristate acetate-induced differentiation of SH-SY5Y neuroblastoma cells. J Neurochem 2015; 133:223-32. [PMID: 25348263 DOI: 10.1111/jnc.12980] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/02/2014] [Accepted: 10/10/2014] [Indexed: 01/10/2023]
Abstract
We explored the interplay between the intracellular energy sensor AMP-activated protein kinase (AMPK), extracellular signal-regulated kinase (ERK), and autophagy in phorbol myristate acetate (PMA)-induced neuronal differentiation of SH-SY5Y human neuroblastoma cells. PMA-triggered expression of neuronal markers (dopamine transporter, microtubule-associated protein 2, β-tubulin) was associated with an autophagic response, measured by the conversion of microtubule-associated protein light chain 3 (LC3)-I to autophagosome-bound LC3-II, increase in autophagic flux, and expression of autophagy-related (Atg) proteins Atg7 and beclin-1. This coincided with the transient activation of AMPK and sustained activation of ERK. Pharmacological inhibition or RNA interference-mediated silencing of AMPK suppressed PMA-induced expression of neuronal markers, as well as ERK activation and autophagy. A selective pharmacological blockade of ERK prevented PMA-induced neuronal differentiation and autophagy induction without affecting AMPK phosphorylation. Conversely, the inhibition of autophagy downstream of AMPK/ERK, either by pharmacological agents or LC3 knockdown, promoted the expression of neuronal markers, thus indicating a role of autophagy in the suppression of PMA-induced differentiation of SH-SY5Y cells. Therefore, PMA-induced neuronal differentiation of SH-SY5Y cells depends on a complex interplay between AMPK, ERK, and autophagy, in which the stimulatory effects of AMPK/ERK signaling are counteracted by the coinciding autophagic response. Phorbol myristate acetate (PMA) induces the expression of dopamine transporter, microtubule-associated protein 2, and β-tubulin, and subsequent neuronal differentiation of SH-SY5Y neuroblastoma cells through AMP-activated protein kinase (AMPK)-dependent activation of extracellular signal-regulated kinase (ERK). The activation of AMPK/ERK axis also induces the expression of beclin-1 and Atg7, and increases LC3 conversion, thereby triggering the autophagic response that counteracts differentiation process.
Collapse
Affiliation(s)
- Nevena Zogovic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | | | |
Collapse
|
47
|
Smirnova L, Hogberg HT, Leist M, Hartung T. Developmental neurotoxicity - challenges in the 21st century and in vitro opportunities. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2015; 31:129-56. [PMID: 24687333 DOI: 10.14573/altex.1403271] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 11/23/2022]
Abstract
In recent years neurodevelopmental problems in children have increased at a rate that suggests lifestyle factors and chemical exposures as likely contributors. When environmental chemicals contribute to neurodevelopmental disorders developmental neurotoxicity (DNT) becomes an enormous concern. But how can it be tackled? Current animal test- based guidelines are prohibitively expensive, at $ 1.4 million per substance, while their predictivity for human health effects may be limited, and mechanistic data that would help species extrapolation are not available. A broader screening for substances of concern requires a reliable testing strategy, applicable to larger numbers of substances, and sufficiently predictive to warrant further testing. This review discusses the evidence for possible contributions of environmental chemicals to DNT, limitations of the current test paradigm, emerging concepts and technologies pertinent to in vitro DNT testing and assay evaluation, as well as the prospect of a paradigm shift based on 21st century technologies.
Collapse
Affiliation(s)
- Lena Smirnova
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, USA
| | | | | | | |
Collapse
|
48
|
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: 1.1] [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
|
49
|
Chou CT, Lin HT, Hwang PA, Wang ST, Hsieh CH, Hwang DF. Taurine resumed neuronal differentiation in arsenite-treated N2a cells through reducing oxidative stress, endoplasmic reticulum stress, and mitochondrial dysfunction. Amino Acids 2014; 47:735-44. [DOI: 10.1007/s00726-014-1901-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/11/2014] [Indexed: 11/30/2022]
|
50
|
Liu JT, Bain LJ. Arsenic inhibits hedgehog signaling during P19 cell differentiation. Toxicol Appl Pharmacol 2014; 281:243-53. [PMID: 25448440 DOI: 10.1016/j.taap.2014.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/02/2014] [Accepted: 10/14/2014] [Indexed: 11/30/2022]
Abstract
Arsenic is a toxicant found in ground water around the world, and human exposure mainly comes from drinking water or from crops grown in areas containing arsenic in soils or water. Epidemiological studies have shown that arsenic exposure during development decreased intellectual function, reduced birth weight, and altered locomotor activity, while in vitro studies have shown that arsenite decreased muscle and neuronal cell differentiation. The sonic hedgehog (Shh) signaling pathway plays an important role during the differentiation of both neurons and skeletal muscle. The purpose of this study was to investigate whether arsenic can disrupt Shh signaling in P19 mouse embryonic stem cells, leading to changes muscle and neuronal cell differentiation. P19 embryonic stem cells were exposed to 0, 0.25, or 0.5 μM of sodium arsenite for up to 9 days during cell differentiation. We found that arsenite exposure significantly reduced transcript levels of genes in the Shh pathway in both a time and dose-dependent manner. This included the Shh ligand, which was decreased 2- to 3-fold, the Gli2 transcription factor, which was decreased 2- to 3-fold, and its downstream target gene Ascl1, which was decreased 5-fold. GLI2 protein levels and transcriptional activity were also reduced. However, arsenic did not alter GLI2 primary cilium accumulation or nuclear translocation. Moreover, additional extracellular SHH rescued the inhibitory effects of arsenic on cellular differentiation due to an increase in GLI binding activity. Taken together, we conclude that arsenic exposure affected Shh signaling, ultimately decreasing the expression of the Gli2 transcription factor. These results suggest a mechanism by which arsenic disrupts cell differentiation.
Collapse
Affiliation(s)
- Jui Tung Liu
- Environmental Toxicology Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Lisa J Bain
- Environmental Toxicology Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA; Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA.
| |
Collapse
|