1
|
Kang B, Wang J, Guo S, Yang L. Mercury-induced toxicity: Mechanisms, molecular pathways, and gene regulation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173577. [PMID: 38852866 DOI: 10.1016/j.scitotenv.2024.173577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/01/2024] [Accepted: 05/25/2024] [Indexed: 06/11/2024]
Abstract
Mercury is a well-known neurotoxicant for humans and wildlife. The epidemic of mercury poisoning in Japan has clearly demonstrated that chronic exposure to methylmercury (MeHg) results in serious neurological damage to the cerebral and cerebellar cortex, leading to the dysfunction of the central nervous system (CNS), especially in infants exposed to MeHg in utero. The occurrences of poisoning have caused a wide public concern regarding the health risk emanating from MeHg exposure; particularly those eating large amounts of fish may experience the low-level and long-term exposure. There is growing evidence that MeHg at environmentally relevant concentrations can affect the health of biota in the ecosystem. Although extensive in vivo and in vitro studies have demonstrated that the disruption of redox homeostasis and microtube assembly is mainly responsible for mercurial toxicity leading to adverse health outcomes, it is still unclear whether we could quantitively determine the occurrence of interaction between mercurial and thiols and/or selenols groups of proteins linked directly to outcomes, especially at very low levels of exposure. Furthermore, intracellular calcium homeostasis, cytoskeleton, mitochondrial function, oxidative stress, neurotransmitter release, and DNA methylation may be the targets of mercury compounds; however, the primary targets associated with the adverse outcomes remain to be elucidated. Considering these knowledge gaps, in this article, we conducted a comprehensive review of mercurial toxicity, focusing mainly on the mechanism, and genes/proteins expression. We speculated that comprehensive analyses of transcriptomics, proteomics, and metabolomics could enhance interpretation of "omics" profiles, which may reveal specific biomarkers obviously correlated with specific pathways that mediate selective neurotoxicity.
Collapse
Affiliation(s)
- Bolun Kang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Jinghan Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China.
| |
Collapse
|
2
|
Panzenhagen AC, Petry FDS, Alves-Teixeira A, Santos L, Carazza-Kessler FG, Gelain DP, Moreira JCF. Biomarkers of methylmercury neurotoxicity and neurodevelopmental features: A systematic review. Food Chem Toxicol 2024; 191:114851. [PMID: 38986832 DOI: 10.1016/j.fct.2024.114851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
The issue of MeHg contamination is a significant concern due to its detrimental impact on the environment. This study aimed to thoroughly investigate the effects of MeHg on neurodevelopmental biomarkers, as there is a lack of systematic reviews in this area. We conducted a comprehensive search of three databases (PubMed, Scopus, and Web of Science) and found 522 records, which were then meticulously reviewed by two independent reviewers. A total of 66 studies were included, with biomarkers related to oxidative stress, neurotransmission, inflammation, epigenetics, and apoptosis being the most prominent. The results of both in vitro and in vivo models indicate that antioxidant enzymes and other oxidative stress-related markers are indeed, altered following MeHg exposure. Moreover, MeHg exposure causes significant disruptions to neurotransmitter levels, activities of neurotransmitter synthesis enzymes, receptor densities, and proteins involved in synaptic function. Proinflammatory biomarkers are consistently overexpressed in both MeHg-treated cells and the brains of exposed rats. Furthermore, studies on DNA methylation and biomarker activity suggest that MeHg exposure may lead to neurotoxicity and neurodevelopmental issues via perturbations to epigenetic markers and the apoptosis pathway.
Collapse
Affiliation(s)
- Alana Castro Panzenhagen
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| | - Fernanda Dos Santos Petry
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Alexsander Alves-Teixeira
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Lucas Santos
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Flávio Gabriel Carazza-Kessler
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| |
Collapse
|
3
|
Tam LM, Rand MD. Review: myogenic and muscle toxicity targets of environmental methylmercury exposure. Arch Toxicol 2024; 98:1645-1658. [PMID: 38546836 PMCID: PMC11105986 DOI: 10.1007/s00204-024-03724-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/29/2024] [Indexed: 05/01/2024]
Abstract
A number of environmental toxicants are noted for their activity that leads to declined motor function. However, the role of muscle as a proximal toxicity target organ for environmental agents has received considerably less attention than the toxicity targets in the nervous system. Nonetheless, the effects of conventional neurotoxicants on processes of myogenesis and muscle maintenance are beginning to resolve a concerted role of muscle as a susceptible toxicity target. A large body of evidence from epidemiological, animal, and in vitro studies has established that methylmercury (MeHg) is a potent developmental toxicant, with the nervous system being a preferred target. Despite its well-recognized status as a neurotoxicant, there is accumulating evidence that MeHg also targets muscle and neuromuscular development as well as contributes to the etiology of motor defects with prenatal MeHg exposure. Here, we summarize evidence for targets of MeHg in the morphogenesis and maintenance of skeletal muscle that reveal effects on MeHg distribution, myogenesis, myotube formation, myotendinous junction formation, neuromuscular junction formation, and satellite cell-mediated muscle repair. We briefly recapitulate the molecular and cellular mechanisms of skeletal muscle development and highlight the pragmatic role of alternative model organisms, Drosophila and zebrafish, in delineating the molecular underpinnings of muscle development and MeHg-mediated myotoxicity. Finally, we discuss how toxicity targets in muscle development may inform the developmental origins of health and disease theory to explain the etiology of environmentally induced adult motor deficits and accelerated decline in muscle fitness with aging.
Collapse
Affiliation(s)
- Lok Ming Tam
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY, 14642, USA.
- Clinical and Translational Science Institute, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY, 14642, USA
| |
Collapse
|
4
|
Lin H, Wei Y, Li S, Mao X, Qin J, Su S, He T. Changes in transcriptome regulations of a marine rotifer Brachionus plicatilis under methylmercury stress. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 49:101177. [PMID: 38104474 DOI: 10.1016/j.cbd.2023.101177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Mercury (Hg), a heavy metal pollutant worldwide, can be transformed into methylmercury (MeHg) by various aquatic microorganisms in water, thus accumulating along the aquatic food chain and posing a particular challenge to human health. Zooplankton plays a crucial role in aquatic ecosystems and serves as a major component of the food chain. To evaluate the effects of MeHg on the rotifer Brachionus plicatilis and reveal the underlying mechanism of these effects, we exposed B. plicatilis to MeHg by either direct immersion or by feeding with MeHg-poisoned Chlorella pyrenoidesa, respectively, and conducted a transcriptomic analysis. The results showed that B. plicatilis directly exposed to MeHg by immersion showed significant enrichment of the glutathione metabolism pathway for detoxification of MeHg. In addition, the exposure to MeHg by feeding induced a significant enrichment of lysosome and notch signaling pathways of rotifers, supporting the hypothesis that MeHg can induce autophagy dysfunction in cells and disturb the nervous system of rotifers. In two different routes of MeHg exposure, the pathway of cytochrome P450 in rotifers showed significant enrichment for resisting MeHg toxicity. Our results suggest further studies on the potential mechanism and biological responses of MeHg toxicity in other links of the aquatic food chain.
Collapse
Affiliation(s)
- Hangyu Lin
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, 400715, China
| | - Yanlin Wei
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, 400715, China
| | - Songzhang Li
- College of Fisheries, Southwest University, Chongqing 400715, China
| | - Xiaodong Mao
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, 400715, China
| | - Jianguang Qin
- College of Science and Engineering, Flinders University, South Australia 5001, Australia
| | - Shengqi Su
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, 400715, China.
| | - Tao He
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, 400715, China.
| |
Collapse
|
5
|
Chen Y, Li H, Yi TC, Shen J, Zhang J. Notch Signaling in Insect Development: A Simple Pathway with Diverse Functions. Int J Mol Sci 2023; 24:14028. [PMID: 37762331 PMCID: PMC10530718 DOI: 10.3390/ijms241814028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Notch signaling is an evolutionarily conserved pathway which functions between adjacent cells to establish their distinct identities. Despite operating in a simple mechanism, Notch signaling plays remarkably diverse roles in development to regulate cell fate determination, organ growth and tissue patterning. While initially discovered and characterized in the model insect Drosophila melanogaster, recent studies across various insect species have revealed the broad involvement of Notch signaling in shaping insect tissues. This review focuses on providing a comprehensive picture regarding the roles of the Notch pathway in insect development. The roles of Notch in the formation and patterning of the insect embryo, wing, leg, ovary and several specific structures, as well as in physiological responses, are summarized. These results are discussed within the developmental context, aiming to deepen our understanding of the diversified functions of the Notch signaling pathway in different insect species.
Collapse
Affiliation(s)
- Yao Chen
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| | - Haomiao Li
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| | - Tian-Ci Yi
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, Institute of Entomology, Guizhou University, Guiyang 550025, China
| | - Jie Shen
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| | - Junzheng Zhang
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| |
Collapse
|
6
|
Koch K, Bartmann K, Hartmann J, Kapr J, Klose J, Kuchovská E, Pahl M, Schlüppmann K, Zühr E, Fritsche E. Scientific Validation of Human Neurosphere Assays for Developmental Neurotoxicity Evaluation. FRONTIERS IN TOXICOLOGY 2022; 4:816370. [PMID: 35295221 PMCID: PMC8915868 DOI: 10.3389/ftox.2022.816370] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/21/2022] [Indexed: 01/06/2023] Open
Abstract
There is a call for a paradigm shift in developmental neurotoxicity (DNT) evaluation, which demands the implementation of faster, more cost-efficient, and human-relevant test systems than current in vivo guideline studies. Under the umbrella of the Organisation for Economic Co-operation and Development (OECD), a guidance document is currently being prepared that instructs on the regulatory use of a DNT in vitro battery (DNT IVB) for fit-for-purpose applications. One crucial issue for OECD application of methods is validation, which for new approach methods (NAMs) requires novel approaches. Here, mechanistic information previously identified in vivo, as well as reported neurodevelopmental adversities in response to disturbances on the cellular and tissue level, are of central importance. In this study, we scientifically validate the Neurosphere Assay, which is based on human primary neural progenitor cells (hNPCs) and an integral part of the DNT IVB. It assesses neurodevelopmental key events (KEs) like NPC proliferation (NPC1ab), radial glia cell migration (NPC2a), neuronal differentiation (NPC3), neurite outgrowth (NPC4), oligodendrocyte differentiation (NPC5), and thyroid hormone-dependent oligodendrocyte maturation (NPC6). In addition, we extend our work from the hNPCs to human induced pluripotent stem cell-derived NPCs (hiNPCs) for the NPC proliferation (iNPC1ab) and radial glia assays (iNPC2a). The validation process we report for the endpoints studied with the Neurosphere Assays is based on 1) describing the relevance of the respective endpoints for brain development, 2) the confirmation of the cell type-specific morphologies observed in vitro, 3) expressions of cell type-specific markers consistent with those morphologies, 4) appropriate anticipated responses to physiological pertinent signaling stimuli and 5) alterations in specific in vitro endpoints upon challenges with confirmed DNT compounds. With these strong mechanistic underpinnings, we posit that the Neurosphere Assay as an integral part of the DNT in vitro screening battery is well poised for DNT evaluation for regulatory purposes.
Collapse
Affiliation(s)
- Katharina Koch
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Kristina Bartmann
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Julia Hartmann
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Julia Kapr
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Jördis Klose
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Eliška Kuchovská
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Melanie Pahl
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Kevin Schlüppmann
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Etta Zühr
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
- Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| |
Collapse
|
7
|
Olsvik PA, Azad AM, Yadetie F. Bioaccumulation of mercury and transcriptional responses in tusk (Brosme brosme), a deep-water fish from a Norwegian fjord. CHEMOSPHERE 2021; 279:130588. [PMID: 33901891 DOI: 10.1016/j.chemosphere.2021.130588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 06/12/2023]
Abstract
High concentrations of mercury (Hg) have been documented in deep-water fish species from some Norwegian fjords. In this study, tusk (Brosme brosme) was sampled from four locations in the innermost parts of Sognefjorden in Western Norway. Total Hg and methylmercury (MeHg) levels were measured in liver tissue. To search for potential sublethal effects of Hg, we characterized the hepatic transcriptome in tusk with high and low levels of Hg bioaccumulation using global transcriptomics analysis (RNA-seq). The results showed that there was a significant correlation between fish weight and accumulated concentrations of MeHg but not total Hg. MeHg accounted for 30-40% of total Hg in liver of most of the fish, although at concentrations above 2-3 mg Hg/kg wet weight the percentage of MeHg dropped considerably. Transcriptome analysis resulted in hundreds of differentially expressed genes in the liver of tusk with high Hg levels. Functional enrichment analysis suggested that the top affected pathways are associated with protein folding, adipogenesis, notch signaling, and lipid metabolism (beta-oxidation and phospholipids). Based on transcriptional responses pointing to well-known effects of Hg compounds in fish, the study suggests that tusk in Sognefjorden could be negatively impacted by Hg bioaccumulation.
Collapse
Affiliation(s)
- Pål A Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway; Institute of Marine Research, Nordnes, Bergen, Norway.
| | - Atabak M Azad
- Institute of Marine Research, Nordnes, Bergen, Norway
| | - Fekadu Yadetie
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Abbott LC, Nigussie F. Mercury Toxicity and Neurogenesis in the Mammalian Brain. Int J Mol Sci 2021; 22:ijms22147520. [PMID: 34299140 PMCID: PMC8305137 DOI: 10.3390/ijms22147520] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/12/2021] [Accepted: 07/05/2021] [Indexed: 01/21/2023] Open
Abstract
The mammalian brain is formed from billions of cells that include a wide array of neuronal and glial subtypes. Neural progenitor cells give rise to the vast majority of these cells during embryonic, fetal, and early postnatal developmental periods. The process of embryonic neurogenesis includes proliferation, differentiation, migration, the programmed death of some newly formed cells, and the final integration of differentiated neurons into neural networks. Adult neurogenesis also occurs in the mammalian brain, but adult neurogenesis is beyond the scope of this review. Developing embryonic neurons are particularly susceptible to neurotoxicants and especially mercury toxicity. This review focused on observations concerning how mercury, and in particular, methylmercury, affects neurogenesis in the developing mammalian brain. We summarized information on models used to study developmental mercury toxicity, theories of pathogenesis, and treatments that could be used to reduce the toxic effects of mercury on developing neurons.
Collapse
Affiliation(s)
- Louise C. Abbott
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, 4458 TAMU, College Station, TX 77843-4458, USA
- Correspondence: ; Tel.: +1-541-254-0779
| | - Fikru Nigussie
- College of Veterinary Medicine, Oregon State University, 700 SW 30th Street, Corvallis, OR 97331, USA;
| |
Collapse
|
9
|
Mechanisms of oxidative stress in methylmercury-induced neurodevelopmental toxicity. Neurotoxicology 2021; 85:33-46. [PMID: 33964343 DOI: 10.1016/j.neuro.2021.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022]
Abstract
Methylmercury (MeHg) is a long-lasting organic environmental pollutant that poses a great threat to human health. Ingestion of seafood containing MeHg is the most important way by which it comes into contact with human body, where the central nervous system (CNS) is the primary target of MeHg toxicity. During periods of pre-plus postnatal, in particular, the brain of offspring is vulnerable to specific developmental insults that result in abnormal neurobehavioral development, even without symptoms in mothers. While many studies on neurotoxic effects of MeHg on the developing brain have been conducted, the mechanisms of oxidative stress in MeHg-induced neurodevelopmental toxicity is less clear. Hitherto, no single process can explain the many effects observed in MeHg-induced neurodevelopmental toxicity. This review summarizes the possible mechanisms of oxidative stress in MeHg-induced neurodevelopmental toxicity, highlighting modulation of Nrf2/Keap1/Notch1, PI3K/AKT, and PKC/MAPK molecular pathways as well as some preventive drugs, and thus contributes to the discovery of endogenous and exogenous molecules that can counteract MeHg-induced neurodevelopmental toxicity.
Collapse
|
10
|
Liao Y, Peng S, He L, Wang Y, Li Y, Ma D, Wang Y, Sun L, Zheng H, Yang W, Dai F, Zhao J. Methylmercury cytotoxicity and possible mechanisms in human trophoblastic HTR-8/SVneo cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 207:111520. [PMID: 33254395 DOI: 10.1016/j.ecoenv.2020.111520] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/22/2020] [Accepted: 10/13/2020] [Indexed: 06/12/2023]
Abstract
Methylmercury (MeHg) exposure during pregnancy can lead to adverse outcomes, including miscarriage and intrauterine growth retardation. In this study, MeHg cytotoxicity and its mechanisms in HTR-8/SVneo cells were investigated. MeHg inhibited HTR-8/SVneo cell viability and severely disrupted the cellular submicrostructure, showing a time-dose effect relationship. After MeHg treatment, the reactive oxygen species levels, malondialdehyde content, and superoxide dismutase (SOD) and catalase activities in the HTR-8/SVneo cells increased significantly with increased MeHg concentration (P<0.05). Similarly, MeHg also induced HTR-8/SVneo cell apoptosis in a dose-dependent manner. The proportion of cells in G1 phase decreased with increasing MeHg concentration, while that in the S and G2/M phases gradually increased. Moreover, cell migration and invasion capacities gradually decreased with increasing MeHg concentration, showing a significant difference between the MeHg-treated and control groups. Genes related to oxidative stress (HSPA6, HSPA1A, Nrf2, SOD1, HO-1, NQO1, OSGIN1, and gPX1), cell cycle (P21 and CDC25A), apoptosis (CYCS and AIFM2), and migration and invasion (CXCL8, CXCL3, CLU, IL24, COL3A1, MAPT, and ITGA7) were differentially expressed in the MeHg-treated group, indicating MeHg toxicity and mechanism of action. This study will provide insights into the prevention and treatment of pregnancy-related diseases caused by MeHg.
Collapse
Affiliation(s)
- Ying Liao
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Shiqin Peng
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Lei He
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Yu Wang
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Yang Li
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Danwei Ma
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Yanan Wang
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Liang Sun
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Hong Zheng
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Wenke Yang
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Fengyan Dai
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China
| | - Jiayuan Zhao
- College of Life Science, Sichuan Normal University, Chengdu, 610101 Sichuan, PR China.
| |
Collapse
|
11
|
Salazar JL, Yang SA, Yamamoto S. Post-Developmental Roles of Notch Signaling in the Nervous System. Biomolecules 2020; 10:biom10070985. [PMID: 32630239 PMCID: PMC7408554 DOI: 10.3390/biom10070985] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Since its discovery in Drosophila, the Notch signaling pathway has been studied in numerous developmental contexts in diverse multicellular organisms. The role of Notch signaling in nervous system development has been extensively investigated by numerous scientists, partially because many of the core Notch signaling components were initially identified through their dramatic ‘neurogenic’ phenotype of developing fruit fly embryos. Components of the Notch signaling pathway continue to be expressed in mature neurons and glia cells, which is suggestive of a role in the post-developmental nervous system. The Notch pathway has been, so far, implicated in learning and memory, social behavior, addiction, and other complex behaviors using genetic model organisms including Drosophila and mice. Additionally, Notch signaling has been shown to play a modulatory role in several neurodegenerative disease model animals and in mediating neural toxicity of several environmental factors. In this paper, we summarize the knowledge pertaining to the post-developmental roles of Notch signaling in the nervous system with a focus on discoveries made using the fruit fly as a model system as well as relevant studies in C elegans, mouse, rat, and cellular models. Since components of this pathway have been implicated in the pathogenesis of numerous psychiatric and neurodegenerative disorders in human, understanding the role of Notch signaling in the mature brain using model organisms will likely provide novel insights into the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
- Department of Neuroscience, BCM, Houston, TX 77030, USA
- Program in Developmental Biology, BCM, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8119
| |
Collapse
|
12
|
Yang L, Zhang Y, Wang F, Luo Z, Guo S, Strähle U. Toxicity of mercury: Molecular evidence. CHEMOSPHERE 2020; 245:125586. [PMID: 31881386 DOI: 10.1016/j.chemosphere.2019.125586] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/28/2019] [Accepted: 12/08/2019] [Indexed: 05/25/2023]
Abstract
Minamata disease in Japan and the large-scale poisoning by methylmercury (MeHg) in Iraq caused wide public concerns about the risk emanating from mercury for human health. Nowadays, it is widely known that all forms of mercury induce toxic effects in mammals, and increasing evidence supports the concern that environmentally relevant levels of MeHg could impact normal biological functions in wildlife. The information of mechanism involved in mercurial toxicity is growing but knowledge gaps still exist between the adverse effects and mechanisms of action, especially at the molecular level. A body of data obtained from experimental studies on mechanisms of mercurial toxicity in vivo and in vitro points to that disruption of the antioxidant system may play an important role in the mercurial toxic effects. Moreover, the accumulating evidence indicates that signaling transduction, protein or/and enzyme activity, and gene regulation are involving in mediating toxic and adaptive response to mercury exposure. We conducted here a comprehensive review of mercurial toxic effects on wildlife and human, in particular synthesized key findings of molecular pathways involved in mercurial toxicity from the cells to human. We discuss the molecular evidence related mercurial toxicity to the adverse effects, with particular emphasis on the gene regulation. The further studies relying on Omic analysis connected to adverse effects and modes of action of mercury will aid in the evaluation and validation of causative relationship between health outcomes and gene expression.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| | - Yuanyuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Zidie Luo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Uwe Strähle
- Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
13
|
Rand MD, Vorojeikina D, Peppriell A, Gunderson J, Prince LM. Drosophotoxicology: Elucidating Kinetic and Dynamic Pathways of Methylmercury Toxicity in a Drosophila Model. Front Genet 2019; 10:666. [PMID: 31447878 PMCID: PMC6695472 DOI: 10.3389/fgene.2019.00666] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/25/2019] [Indexed: 01/18/2023] Open
Abstract
The risks of methylmercury (MeHg) toxicity are greatest during early life where it has long been appreciated that the developing nervous system is an especially sensitive target. Yet, understanding the discrete mechanisms of MeHg toxicity have been obscured by the wide variation in the nature and severity of developmental outcomes that are typically seen across individuals in MeHg exposed populations. Some insight has come from studies aimed at identifying a role for genetic background as a modifier of MeHg toxicity, which have predominantly focused on factors influencing MeHg toxicokinetics, notably, polymorphisms in genes related to glutathione (GSH) metabolism. For example, variants in genes encoding the catalytic and modifier subunits of glutamyl-cysteine ligase (GCLc and GCLm), the rate limiting enzyme for GSH synthesis, have been reported to associate with Hg body burden (Hg levels in blood or hair) in humans. However, GSH can facilitate both toxicokinetics and toxicodynamics of MeHg by forming MeHg-GSH conjugates, which are readily transported and excreted, and by acting indirectly as an anti-oxidant. In this study, we refine a model to distinguish kinetic and dynamic traits of MeHg toxicity using a paradigm of Drosophotoxicolgy. First, we identify that the pupal stage is selectively sensitive to MeHg toxicity. Using a protocol of larval feeding, measurements of Hg body burden, and assays of development to adulthood (pupal eclosion), we identify strain-dependent variation in MeHg elimination as a potential kinetic determinant of differential tolerance to MeHg. We also find that global upregulation of GSH levels, with GCLc trans-gene expression, can induce MeHg tolerance and reduce Hg body burden. However, we demonstrate that MeHg tolerance can also be achieved independently of reducing Hg body burden, in both wild-derived strains and with targeted expression of GCLc in developing neuronal and muscle tissue, pointing to a robust toxicodynamic mechanism. Our findings have important implications for understanding variation in MeHg toxic potential on an individual basis and for informing the process of relating a measurement of Hg body burden to the potential for adverse developmental outcome.
Collapse
Affiliation(s)
- Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Ashley Peppriell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Jakob Gunderson
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Lisa M Prince
- School of Human Health Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
14
|
Raciti M, Salma J, Spulber S, Gaudenzi G, Khalajzeyqami Z, Conti M, Anderlid BM, Falk A, Hermanson O, Ceccatelli S. NRXN1 Deletion and Exposure to Methylmercury Increase Astrocyte Differentiation by Different Notch-Dependent Transcriptional Mechanisms. Front Genet 2019; 10:593. [PMID: 31316548 PMCID: PMC6610538 DOI: 10.3389/fgene.2019.00593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/05/2019] [Indexed: 01/11/2023] Open
Abstract
Controversial evidence points to a possible involvement of methylmercury (MeHg) in the etiopathogenesis of autism spectrum disorders (ASD). In the present study, we used human neuroepithelial stem cells from healthy donors and from an autistic patient bearing a bi-allelic deletion in the gene encoding for NRXN1 to evaluate whether MeHg would induce cellular changes comparable to those seen in cells derived from the ASD patient. In healthy cells, a subcytotoxic concentration of MeHg enhanced astroglial differentiation similarly to what observed in the diseased cells (N1), as shown by the number of GFAP positive cells and immunofluorescence signal intensity. In both healthy MeHg-treated and N1 untreated cells, aberrations in Notch pathway activity seemed to play a critical role in promoting the differentiation toward glia. Accordingly, treatment with the established Notch inhibitor DAPT reversed the altered differentiation. Although our data are not conclusive since only one of the genes involved in ASD is considered, the results provide novel evidence suggesting that developmental exposure to MeHg, even at subcytotoxic concentrations, induces alterations in astroglial differentiation similar to those observed in ASD.
Collapse
Affiliation(s)
- Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jahan Salma
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Gaudenzi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mirko Conti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Centre for Molecular Medicine, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Prince LM, Rand MD. Methylmercury exposure causes a persistent inhibition of myogenin expression and C2C12 myoblast differentiation. Toxicology 2018; 393:113-122. [PMID: 29104120 PMCID: PMC5757876 DOI: 10.1016/j.tox.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxicant, best known for its selective targeting of the developing nervous system. MeHg exposure has been shown to cause motor deficits such as impaired gait and coordination, muscle weakness, and muscle atrophy, which have been associated with disruption of motor neurons. However, recent studies have suggested that muscle may also be a target of MeHg toxicity, both in the context of developmental myogenic events and of low-level chronic exposures affecting muscle wasting in aging. We therefore investigated the effects of MeHg on myotube formation, using the C2C12 mouse myoblast model. We found that MeHg inhibits both differentiation and fusion, in a concentration-dependent manner. Furthermore, MeHg specifically and persistently inhibits myogenin (MyoG), a transcription factor involved in myocyte differentiation, within the first six hours of exposure. MeHg-induced reduction in MyoG expression is contemporaneous with a reduction of a number of factors involved in mitochondrial biogenesis and mtDNA transcription and translation, which may implicate a role for mitochondria in mediating MeHg-induced change in the differentiation program. Unexpectedly, inhibition of myoblast differentiation with MeHg parallels inhibition of Notch receptor signaling. Our research establishes muscle cell differentiation as a target for MeHg toxicity, which may contribute to the underlying etiology of motor deficits with MeHg toxicity.
Collapse
Affiliation(s)
- Lisa M Prince
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| |
Collapse
|
16
|
Prince LM, Rand MD. Notch Target Gene E(spl)mδ Is a Mediator of Methylmercury-Induced Myotoxicity in Drosophila. Front Genet 2018; 8:233. [PMID: 29379520 PMCID: PMC5775289 DOI: 10.3389/fgene.2017.00233] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/22/2017] [Indexed: 01/09/2023] Open
Abstract
Methylmercury (MeHg) is a ubiquitous environmental contaminant and neurotoxicant that has long been known to cause a variety of motor deficits. These motor deficits have primarily been attributed to MeHg targeting of developing neurons and induction of oxidative stress and calcium dysregulation. Few studies have looked at how MeHg may be affecting fundamental signaling mechanisms in development, particularly in developing muscle. Studies in Drosophila recently revealed that MeHg perturbs embryonic muscle formation and upregulates Notch target genes, reflected predominantly by expression of the downstream transcriptional repressor Enhancer of Split mdelta [E(spl)mδ]. An E(spl)mδ reporter gene shows expression primarily in the myogenic domain, and both MeHg exposure and genetic upregulation of E(spl)mδ can disrupt embryonic muscle development. Here, we tested the hypothesis that developing muscle is targeted by MeHg via upregulation of E(spl)mδ using genetic modulation of E(spl)mδ expression in combination with MeHg exposure in developing flies. Developmental MeHg exposure causes a decreased rate of eclosion that parallels gross disruption of indirect flight muscle (IFM) development. An increase in E(spl) expression across the pupal stages, with preferential E(spl)mδ upregulation occurring at early (p5) stages, is also observed. E(spl)mδ overexpression in myogenic lineages under the Mef2 promoter was seen to phenocopy eclosion and IFM effects of developmental MeHg exposure; whereas reduced expression of E(spl)mδ shows rescue of eclosion and IFM morphology effects of MeHg exposure. No effects were seen on eclosion with E(spl)mδ overexpression in neural and gut tissues. Our data indicate that muscle development is a target for MeHg and that E(spl)mδ is a muscle-specific mediator of this myotoxicity. This research advances our knowledge of the target pathways that mediate susceptibility to MeHg toxicity, as well as a potential muscle development-specific role for E(spl)mδ.
Collapse
Affiliation(s)
- Lisa M Prince
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| | - Matthew D Rand
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| |
Collapse
|
17
|
The road less traveled: from genotype to phenotype in flies and humans. Mamm Genome 2017; 29:5-23. [DOI: 10.1007/s00335-017-9722-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/05/2017] [Indexed: 12/20/2022]
|
18
|
Edoff K, Raciti M, Moors M, Sundström E, Ceccatelli S. Gestational Age and Sex Influence the Susceptibility of Human Neural Progenitor Cells to Low Levels of MeHg. Neurotox Res 2017; 32:683-693. [PMID: 28756503 PMCID: PMC5602033 DOI: 10.1007/s12640-017-9786-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 07/09/2017] [Accepted: 07/12/2017] [Indexed: 01/13/2023]
Abstract
The developing nervous system is highly susceptible to methylmercury (MeHg), a widespread environmental neurotoxic contaminant. A wide range of morphological and functional outcomes have been described; however, there are still open questions regarding the mechanisms behind the developmental neurotoxic effects induced by low-level exposure. In the present study, we have examined the effects of nanomolar concentrations of MeHg on primary fetal human progenitor cells (hNPCs) with special focus on the role played by developmental stage and sex on the neurotoxic outcome. We found that neurospheres derived from earlier gestational time points exhibit higher susceptibility to MeHg, as they undergo apoptosis at a much lower dose (25 nM) as compared to neurospheres established from older fetuses (100 nM). At subapoptotic concentrations (10 nM), MeHg inhibited neuronal differentiation and maturation of hNPCs, as shown by a reduced number of Tuj1-positive cells and a visible reduction in neurite extension and cell migration, associated with a misregulation of Notch1 and BDNF signaling pathways. Interestingly, cells derived from male fetuses showed more severe alterations of neuronal morphology as compared to cells from females, indicating that the MeHg-induced impairment of neurite extension and cell migration is sex-dependent. Accordingly, the expression of the CDKL5 gene, a major factor regulating neurite outgrowth, was significantly more downregulated in male-derived cells. Altogether, gestational age and sex appear to be critical factors influencing in vitro hNPC sensitivity to low levels of MeHg.
Collapse
Affiliation(s)
- Karin Edoff
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77, Stockholm, Sweden
| | - Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77, Stockholm, Sweden.
| | - Michaela Moors
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Geriatrik-lab plan 5, SE-141 52, Huddinge, Sweden
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77, Stockholm, Sweden
| |
Collapse
|
19
|
Le Manh H, Guio L, Merenciano M, Rovira Q, Barrón MG, González J. Natural and laboratory mutations in kuzbanian are associated with zinc stress phenotypes in Drosophila melanogaster. Sci Rep 2017; 7:42663. [PMID: 28218276 PMCID: PMC5316978 DOI: 10.1038/srep42663] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/13/2017] [Indexed: 11/09/2022] Open
Abstract
Organisms must cope with altered environmental conditions such as high concentrations of heavy metals. Stress response to heavy metals is mediated by the metal-responsive transcription factor 1 (MTF-1), which is conserved from Drosophila to humans. MTF-1 binds to metal response elements (MREs) and changes the expression of target genes. kuzbanian (kuz), a metalloendopeptidase that activates the evolutionary conserved Notch signaling pathway, has been identified as an MTF-1 target gene. We have previously identified a putatively adaptive transposable element in the Drosophila melanogaster genome, named FBti0019170, inserted in a kuz intron. In this work, we investigated whether a laboratory mutant stock overexpressing kuz is associated with zinc stress phenotypes. We found that both embryos and adult flies overexpressing kuz are more tolerant to zinc compared with wild-type flies. On the other hand, we found that the effect of FBti0019170 on zinc stress tolerance depends on developmental stage and genetic background. Moreover, in the majority of the genetic backgrounds analyzed, FBti0019170 has a deleterious effect in unpolluted environments in pre-adult stages. These results highlight the complexity of natural mutations and suggest that besides laboratory mutations, natural mutations should be studied in order to accurately characterize gene function and evolution.
Collapse
Affiliation(s)
- Hung Le Manh
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49. 08003 Barcelona. Spain
- Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st, Hanoi, Vietnam
| | - Lain Guio
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49. 08003 Barcelona. Spain
| | - Miriam Merenciano
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49. 08003 Barcelona. Spain
| | - Quirze Rovira
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49. 08003 Barcelona. Spain
| | - Maite G. Barrón
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49. 08003 Barcelona. Spain
| | - Josefa González
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta 37-49. 08003 Barcelona. Spain
| |
Collapse
|
20
|
Long-term consequences of prenatal stress and neurotoxicants exposure on neurodevelopment. Prog Neurobiol 2016; 155:21-35. [PMID: 27236051 DOI: 10.1016/j.pneurobio.2016.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 02/19/2016] [Accepted: 05/24/2016] [Indexed: 11/20/2022]
Abstract
There is a large consensus that the prenatal environment determines the susceptibility to pathological conditions later in life. The hypothesis most widely accepted is that exposure to insults inducing adverse conditions in-utero may have negative effects on the development of target organs, disrupting homeostasis and increasing the risk of diseases at adulthood. Several models have been proposed to investigate the fetal origins of adult diseases, but although these approaches hold true for almost all diseases, particular attention has been focused on disorders related to the central nervous system, since the brain is particularly sensitive to alterations of the microenvironment during early development. Neurobiological disorders can be broadly divided into developmental, neurodegenerative and neuropsychiatric disorders. Even though most of these diseases share genetic risk factors, the onset of the disorders cannot be explained solely by inheritance. Therefore, current understanding presumes that the interactions of environmental input, may lead to different disorders. Among the insults that can play a direct or indirect role in the development of neurobiological disorders are stress, infections, drug abuse, and environmental contaminants. Our laboratories have been involved in the study of the neurobiological impact of gestational stress on the offspring (Dr. Antonelli's lab) and on the effect of gestational exposure to toxicants, mainly methyl mercury (MeHg) and perfluorinated compounds (PFCs) (Dr. Ceccatelli's lab). In this focused review, we will review the specialized literature but we will concentrate mostly on our own work on the long term neurodevelopmental consequences of gestational exposure to stress and neurotoxicants.
Collapse
|
21
|
Tong J, Wang Y, Lu Y. In vitro evaluation of inorganic and methyl mercury mediated cytotoxic effect on neural cells derived from different animal species. J Environ Sci (China) 2016; 41:138-145. [PMID: 26969059 DOI: 10.1016/j.jes.2015.04.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/24/2015] [Accepted: 04/15/2015] [Indexed: 05/15/2023]
Abstract
To extend the current understanding of the mercury-mediated cytotoxic effect, five neural cell lines established from different animal species were comparatively analyzed using three different endpoint bioassays: thiazolyl blue tetrazolium bromide, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay (MTT), neutral red uptake assay (NRU), and Coomassie blue assay (CB). Following a 24-hr exposure to selected concentrations of mercury chloride (HgCl2) and methylmercury (II) chloride (MeHgCl), the cytotoxic effect on test cells was characterized by comparing their 50% inhibition concentration (IC50) values. Experimental results indicated that both these forms of mercury were toxic to all the neural cells, but at very different degrees. The IC50 values of MeHgCl among these cell lines ranged from 1.15±0.22 to 10.31±0.70μmol/L while the IC50 values for HgCl2 were much higher, ranging from 6.44±0.36 to 160.97±19.63μmol/L, indicating the more toxic nature of MeHgCl. The IC50 ratio between HgCl2 and MeHgCl ranged from 1.75 to 96.0, which confirms that organic mercury is much more toxic to these neural cells than inorganic mercury. Among these cell lines, HGST-BR and TriG44 derived from marine sea turtles showed a significantly high tolerance to HgCl2 as compared to the three mammalian neural cells. Among these neural cells, SK-N-SH represented the most sensitive cells to both chemical forms of mercury.
Collapse
Affiliation(s)
- Jing Tong
- Institute of TCM & Natural Products, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Youwei Wang
- Institute of TCM & Natural Products, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuanan Lu
- Global Health Center, School of Public Health, Wuhan University, Wuhan 430071, China; Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| |
Collapse
|
22
|
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: 62] [Impact Index Per Article: 6.2] [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
|
23
|
Engel GL, Rand MD. The Notch target E(spl)mδ is a muscle-specific gene involved in methylmercury toxicity in motor neuron development. Neurotoxicol Teratol 2014; 43:11-8. [PMID: 24632433 DOI: 10.1016/j.ntt.2014.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/19/2014] [Accepted: 03/04/2014] [Indexed: 01/02/2023]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxin that has a selective and potent impact on the nervous system, particularly during neural development yet, the mechanisms for its apparent neurodevelopmental specificity are unknown. The Notch receptor pathway has been implicated as a MeHg target in several studies. Notch signaling mediates cell-cell signals in a number of developmental contexts including neurogenesis and myogenesis, where it fundamentally acts to repress differentiation. Previous work in our lab has shown that MeHg causes preferential upregulation of a canonical Notch response gene, E(spl)mδ, in Drosophila embryos. In parallel, MeHg is seen to disrupt outgrowth of embryonic intersegmental motor nerves (ISN), which can be mimicked by expression of activated Notch in embryonic neurons. However, overexpression of E(spl)mδ in developing neurons fails to elicit motor neuron outgrowth defects, pointing to a non-autonomous role for E(spl)mδ in motor axon development. In this study we investigate a role for E(spl)mδ in conveying the toxicity of MeHg in the embryo. We find that endogenous expression of the E(spl)mδ gene localizes to developing somatic muscles in embryos. Notably, E(spl)mδ expression is seen in several muscles that are known synaptic targets for both the ISN and the segmental motor nerve (SN). We also demonstrate that the SN, similar to the ISN, exhibits disrupted axon outgrowth in response to MeHg. E(spl)mδ can induce a SN motor neuron phenotype, similar to MeHg treatment; but, only when E(spl)mδ expression is targeted to developing muscles. E(spl)mδ overexpression in developing muscles also results in aberrant muscle morphology, which is not apparent with expression of the closely related E(spl)mγ in developing muscles. Our data point to a role for the Notch target E(spl)mδ in mediating MeHg toxicity in embryonic development by disrupting the coordinated targeting of motor neurons to their muscle targets.
Collapse
Affiliation(s)
- Gregory L Engel
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box EHSC, Rochester, NY 14622, United States
| |
Collapse
|
24
|
|
25
|
Mercury specifically induces LINE-1 activity in a human neuroblastoma cell line. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2013; 759:9-20. [PMID: 24240092 DOI: 10.1016/j.mrgentox.2013.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/25/2013] [Accepted: 07/05/2013] [Indexed: 11/21/2022]
Abstract
L1 retro-elements comprise 17% of the human genome. Approximately 100 copies of these autonomous mobile elements are active in our DNA and can cause mutations, gene disruptions, and genomic instability. Therefore, human cells control the activities of L1 elements, in order to prevent their deleterious effects through different mechanisms. However, some toxic agents increase the retrotransposition activity of L1 elements in somatic cells. In order to identify specific effects of neurotoxic metals on L1 activity in neuronal cells, we studied the effects of mercury and cobalt on L1-retroelement activity by measuring levels of cellular transcription, protein expression, and genomic retrotransposition in a neuroblastoma cell line compared with the effects in three non-neuronal cell lines. Our results show that mercury increased the expression of L1 RNA, the activity of the L1 5'UTR, and L1 retrotransposition exclusively in the neuroblastoma cell line but not in non-neuronal cell lines. However, cobalt increased the expression of L1 RNA in neuroblastoma cells, HeLa cells, and wild-type human fibroblasts, and also increased the activity of the L1 5'UTR as well as the SV40 promoter in HeLa cells but not in neuroblastoma cells. Exposure to cobalt did not result in increased retrotransposition activity in HeLa cells or neuroblastoma cells. We conclude that non-toxic levels of the neurotoxic agent mercury could influence DNA by increasing L1 activities, specifically in neuronal cells, and may make these cells susceptible to neurodegeneration over time.
Collapse
|
26
|
Croes K, De Coster S, De Galan S, Morrens B, Loots I, Van de Mieroop E, Nelen V, Sioen I, Bruckers L, Nawrot T, Colles A, Den Hond E, Schoeters G, van Larebeke N, Baeyens W, Gao Y. Health effects in the Flemish population in relation to low levels of mercury exposure: from organ to transcriptome level. Int J Hyg Environ Health 2013; 217:239-47. [PMID: 23920476 DOI: 10.1016/j.ijheh.2013.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 04/29/2013] [Accepted: 06/04/2013] [Indexed: 11/30/2022]
Abstract
Due to possible health risks, quantification of mercury accumulation in humans was included in the Flemish biomonitoring programmes FLEHS I (2002-2006) and FLEHS II (2007-2011). The general objective of FLEHS I was to assess regional exposure levels in order to link possible differences in these internal exposure levels to different types of local environmental pressure. Therefore, Hg and MMHg (methylmercury) were only measured in pooled blood samples per region and per age class. In FLEHS II, mercury concentrations were measured in hair of each participant. About 200 adolescents and 250 mothers (reference group) and two times 200 adolescents (2 hotspots) were screened. The main objectives of the FLEHS II study were: (1) to determine reference levels of mercury in hair for Flanders; (2) to assess relations between mercury exposure and possible sources like fish consumption; (3) to assess dose-effect relations between mercury exposure and health effect markers. The results showed that mercury concentrations in the Flemish population were rather low compared to other studies. Mercury levels in the Flemish populations were strongly related to the age of the participants and consumption of fish. Significant negative associations were observed between mercury in hair and asthma, having received breast feeding as a newborn, age at menarche in girls, allergy for animals and free testosterone levels. Significant correlations were also observed between mercury in hair and genes JAK2, ARID4A, Hist1HA4L (boys) and HLAdrb5, PIAS2, MANN1B1, GIT and ABCA1 (girls).
Collapse
Affiliation(s)
- Kim Croes
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium
| | - Sam De Coster
- Ghent University Hospital, Study Centre for Carcinogenesis and Primary Prevention of Cancer, De Pintelaan 185, 9000 Ghent, Belgium
| | - Sandra De Galan
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium
| | - Bert Morrens
- University of Antwerp, Faculty of Political and Social Sciences, Department of Sociology, Sint Jacobstraat 2, 2000 Antwerp, Belgium
| | - Ilse Loots
- University of Antwerp, Faculty of Political and Social Sciences, Department of Sociology, Sint Jacobstraat 2, 2000 Antwerp, Belgium
| | - Els Van de Mieroop
- Provincial Institute for Hygiene, Kronenburgstraat 45, 2000 Antwerp, Belgium
| | - Vera Nelen
- Provincial Institute for Hygiene, Kronenburgstraat 45, 2000 Antwerp, Belgium
| | - Isabelle Sioen
- Ghent University, Department of Public Health, UZ-2 Blok A, De Pintelaan 185, 9000 Ghent, Belgium; Research Foundation - Flanders, Egmontstraat 5, 1000 Brussels, Belgium
| | - Liesbeth Bruckers
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Tim Nawrot
- School of Public Health, Occupational & Environmental Medicine, K.U. Leuven, Herestraat 49 (O&N 706), 3000 Leuven, Belgium; Centre for Environmental Sciences, Hasselt University, Agoralaan Gebouw D, 3590 Diepenbeek, Belgium
| | - Ann Colles
- Flemish Institute for Technological Research (VITO), Environmental Health and Risk, Boeretang 200, 2400 Mol, Belgium
| | - Elly Den Hond
- Flemish Institute for Technological Research (VITO), Environmental Health and Risk, Boeretang 200, 2400 Mol, Belgium
| | - Greet Schoeters
- Flemish Institute for Technological Research (VITO), Environmental Health and Risk, Boeretang 200, 2400 Mol, Belgium
| | - Nicolas van Larebeke
- Ghent University Hospital, Study Centre for Carcinogenesis and Primary Prevention of Cancer, De Pintelaan 185, 9000 Ghent, Belgium
| | - Willy Baeyens
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium
| | - Yue Gao
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
27
|
Engel GL, Delwig A, Rand MD. The effects of methylmercury on Notch signaling during embryonic neural development in Drosophila melanogaster. Toxicol In Vitro 2011; 26:485-92. [PMID: 22230562 DOI: 10.1016/j.tiv.2011.12.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 12/19/2011] [Accepted: 12/20/2011] [Indexed: 10/14/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous toxicant that targets the developing fetal nervous system. MeHg interacts with the Notch signaling pathway, a highly-conserved intercellular signaling mechanism required for normal development. Notch signaling is conveyed by activation of the genes in the enhancer of split (E(spl)) locus in Drosophila. We have previously shown that acute high doses of MeHg upregulate several E(spl) genes in Drosophila neural-derived C6 cells. Furthermore, MeHg induction of E(spl) can occur independent of the Notch receptor itself. We now show that MeHg, unlike inorganic mercury (HgCl2), preferentially upregulates E(spl)mδ and E(spl)mγ in Drosophila C6 cells. This is distinct from Delta ligand-induced Notch signaling in which no induction of E(spl)mδ is seen. MeHg is also seen to specifically upregulate E(spl)mδ in Drosophila embryos where HgCl2 showed no such effect. Additionally, treatment of embryos with MeHg caused a consistent failure in axonal outgrowth of the intersegmental nerve (ISN). This ISN phenotype was partially replicated by genetic activation of the Notch pathway, but was not replicated by increasing expression of E(spl)mδ. These data suggest a role for Notch signaling and the E(spl)mδ target gene in MeHg toxicity, however, the site of action for E(spl)mδ in this system remains to be elucidated.
Collapse
Affiliation(s)
- G L Engel
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, USA
| | | | | |
Collapse
|
28
|
Alattia J, Kuraishi T, Dimitrov M, Chang I, Lemaitre B, Fraering PC. Mercury is a direct and potent γ‐secretase inhibitor affecting Notch processing and development in
Drosophila. FASEB J 2011; 25:2287-95. [DOI: 10.1096/fj.10-174078] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | - Takayuki Kuraishi
- Global Health Institute, School of Life Sciences, Swiss Federal Institute of TechnologyLausanneSwitzerland
| | | | | | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Swiss Federal Institute of TechnologyLausanneSwitzerland
| | | |
Collapse
|
29
|
Robinson JF, van Beelen VA, Verhoef A, Renkens MFJ, Luijten M, van Herwijnen MHM, Westerman A, Pennings JLA, Piersma AH. Embryotoxicant-Specific Transcriptomic Responses in Rat Postimplantation Whole-Embryo Culture. Toxicol Sci 2010; 118:675-85. [DOI: 10.1093/toxsci/kfq292] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
30
|
Mahapatra CT, Bond J, Rand DM, Rand MD. Identification of methylmercury tolerance gene candidates in Drosophila. Toxicol Sci 2010; 116:225-38. [PMID: 20375079 PMCID: PMC2902855 DOI: 10.1093/toxsci/kfq097] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 03/19/2010] [Indexed: 11/14/2022] Open
Abstract
Methylmercury (MeHg) is a ubiquitous environmental contaminant that preferentially targets the developing nervous system. Variable outcomes of prenatal MeHg exposure within a population point to a genetic component that regulates MeHg toxicity. We therefore sought to identify fundamental MeHg tolerance genes using the Drosophila model for genetic and molecular dissection of a MeHg tolerance trait. We observe autosomal dominance in a MeHg tolerance trait (development on MeHg food) in both wild-derived and laboratory-selected MeHg-tolerant strains of flies. We performed whole-genome transcript profiling of larval brains of tolerant (laboratory selected) and nontolerant (control) strains in the presence and absence of MeHg stress. Pairwise transcriptome comparisons of four conditions (+/-selection and +/-MeHg) identified a "down-down-up" expression signature, whereby MeHg alone and selection alone resulted in a greater number of downregulated transcripts, and the combination of selection + MeHg resulted in a greater number of upregulated transcripts. Functional annotation cluster analyses showed enrichment for monooxygenases/oxidoreductases, which include cytochrome P450 (CYP) family members. Among the 10 CYPs upregulated with selection + MeHg in tolerant strains, CYP6g1, previously identified as the dichlorodiphenyl trichloroethane resistance allele in flies, was the most highly expressed and responsive to MeHg. Among all the genes, Turandot A (TotA), an immune pathway-regulated humoral response gene, showed the greatest upregulation with selection + MeHg. Neural-specific transgenic overexpression of TotA enhanced MeHg tolerance during pupal development. Identification of TotA and CYP genes as MeHg tolerance genes is an inroad to investigating the conserved function of immune signaling and phase I metabolism pathways in MeHg toxicity and tolerance in higher organisms.
Collapse
Affiliation(s)
- Cecon T. Mahapatra
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont
| | - Jeffrey Bond
- Bioinformatics Core, Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington, Vermont 05405
| | - David M. Rand
- Department of Ecology & Evolutionary Biology, Brown University, Providence, Rhode Island 02912
| | - Matthew D. Rand
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont
| |
Collapse
|
31
|
Are neuropathological conditions relevant to ethylmercury exposure? Neurotox Res 2009; 18:59-68. [PMID: 19756911 DOI: 10.1007/s12640-009-9113-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 08/31/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
Abstract
Mercury and mercurial compounds are among the environmentally ubiquitous substances most toxic to both wildlife and humans. Once released into the environment from both natural and anthropogenic sources, mercury exists mainly as three different molecular species: elemental, inorganic, and organic. Potential health risks have been reported from exposure to all forms; however, of particular concern for human exposure relate to the potent neurotoxic effects of methylmercury (MeHg), especially for the developing nervous system. The general population is primarily exposed to MeHg by seafood consumption. In addition, some pharmaceuticals, including vaccines, have been, and some continue to be, a ubiquitous source of exposure to mercurials. A significant controversy has been whether the vaccine preservative ethylmercury thiosalicylate, commonly known as thimerosal, could cause the development of autism. In this review, we have discussed the hypothesis that exposure to thimerosal during childhood may be a primary cause of autism. The conclusion is that there are no reliable data indicating that administration of vaccines containing thimerosal is a primary cause of autism. However, one cannot rule out the possibility that the individual gene profile and/or gene-environment interactions may play a role in modulating the response to acquired risk by modifying the individual susceptibility.
Collapse
|
32
|
Rand MD, Dao JC, Clason TA. Methylmercury disruption of embryonic neural development in Drosophila. Neurotoxicology 2009; 30:794-802. [PMID: 19409416 PMCID: PMC2774130 DOI: 10.1016/j.neuro.2009.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 04/21/2009] [Accepted: 04/21/2009] [Indexed: 11/15/2022]
Abstract
Methylmercury (MeHg) is a potent environmental neurotoxin that preferentially targets the developing embryonic nervous system. While a number of cytotoxic mechanisms of MeHg have been characterized in differentiated cells its mode of action in the developing nervous system in vivo is less clear. Studies in primate and rodent models demonstrate aberrant cell migration and disorganized patterning of cortical layers in the brain following MeHg exposure. However, defining the molecular and cellular pathways targeted by MeHg will require more genetically accessible animal models. In this study, we instigate a method of in vitro MeHg exposure using Drosophila embryos. We demonstrate dose-dependent inhibition of embryonic development with MeHg revealed by a failure of embryos to hatch to the larval stage. In addition, we document definitive phenotypes in neural development showing abnormalities in neuronal and glial cell patterning consistent with disrupted migration. We observe pronounced defects in neurite outgrowth in both central and peripheral neurons. Ectopic expression of the Nrf2 transcription factor in embryos, a core factor in the antioxidant response element (ARE) pathway, enhances embryonic development and hatching in the presence of MeHg, illustrating the power of this model for investigation of candidate MeHg tolerance genes. Our data establish a utility for the Drosophila embryo model as a platform for elucidating MeHg sensitive pathways in neural development.
Collapse
Affiliation(s)
- Matthew D Rand
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, United States.
| | | | | |
Collapse
|
33
|
Abstract
A central mechanism in activation of the Notch signaling pathway is cleavage of the Notch receptor by ADAM metalloproteases. ADAMs also cleave Delta, the ligand for Notch, thereby downregulating Notch signals. Two ADAMs, Kuzbanian (Kuz) and TNF-alpha converting enzyme (TACE), are known to process both Delta and Notch, yet the role of these cleavages in signal propagation has remained controversial. Using an in vitro model, we show that Kuz regulates Notch signaling primarily by activating the receptor and has little overall effect on signaling via disabling Delta. We confirm that Kuz-dependent activation of Notch requires stimulation of Notch by Delta. However, over-expression of Kuz gives ligand-independent Notch activation. In contrast, TACE, which is elevated in expression in the developing Drosophila nervous system, can efficiently activate Notch in a ligand-independent manner. Altogether, these data demonstrate the potential for Kuz and TACE to participate in context- and mechanism-specific modes of Notch activation.
Collapse
Affiliation(s)
- A. Delwig
- Department of Anatomy and Neurobiology, University of Vermont, HSRF 426C, 149 Beaumont Ave., Burlington, VT 05405 USA
| | - M. D. Rand
- Department of Anatomy and Neurobiology, University of Vermont, HSRF 426C, 149 Beaumont Ave., Burlington, VT 05405 USA
| |
Collapse
|
34
|
Developmental exposure to polychlorinated biphenyls or methylmercury, but not to its combination, impairs the glutamate-nitric oxide-cyclic GMP pathway and learning in 3-month-old rats. Neuroscience 2008; 154:1408-16. [PMID: 18556134 DOI: 10.1016/j.neuroscience.2008.05.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 05/12/2008] [Accepted: 05/13/2008] [Indexed: 11/21/2022]
Abstract
Prenatal exposure to polychlorinated biphenyls (PCBs) or methylmercury (MeHg) contaminated food may affect brain development, leading to long-term alterations in cognitive function. Both types of contaminants, PCBs and MeHg, are often found together contaminating food, especially fish in some polluted areas. Exposure to combinations of neurotoxicants may exert different effects on the developing nervous system than exposure to individual contaminants. Developmental exposure (during pregnancy and lactation) to PCB126 or PCB153 impairs learning ability when the rats are 3 months old. Impairment of learning seems to be a consequence of impairment of the function of the glutamate-nitric oxide (NO)-cGMP pathway in brain in vivo. The aims of the present work were 1) to assess whether perinatal exposure to MeHg also affects the function of the glutamate-NO-cGMP pathway in brain in vivo analyzed by in vivo brain microdialysis and/or the ability to learn the Y maze task when the rats are 3 months old, and 2) to assess whether perinatal exposure to combinations of MeHg with PCB153 or PCB126 potentiates, decreases or does not modify the effects of the individual neurotoxicants. Perinatal exposure to PCB126, PCB153 or MeHg impaired the function of the glutamate-NO-cGMP pathway in cerebellum and learning ability. However, co-exposure to PCB126+MeHg or PCB153+MeHg inhibits the impairment of the pathway or learning ability. These results support that the function of this pathway modulates learning of the Y maze task. Moreover, they show that co-exposure to these PCBs and MeHg does not exacerbate, but reduces the effects on the ability to learn this task.
Collapse
|
35
|
Abstract
The Notch receptor is essential for neural stem cell (NSC) characteristics. Relatively high concentrations (micromolar) of methylmercury (MeHg) activate Notch signalling in Drosophila cell lines; however, exposure of MeHg at such concentrations is rare, and the implications for mammalian cells are unclear. We have shown that MeHg at a nanomolar range inhibits neuronal differentiation of rodent embryonic NSCs. Here we show that low MeHg levels (2.5-10 nM) activated Notch signalling in NSCs, as assessed by the increased activity in a specific Notch-reporter assay and by the increased cleavage of the Notch intracellular domain. Importantly, pretreatment with Notch cleavage inhibitor reversed the MeHg-induced repression of neuronal differentiation, suggesting that Notch activation is involved in the inhibition of NSC differentiation by environmentally relevant levels of MeHg.
Collapse
|
36
|
Rand MD, Bland CE, Bond J. Methylmercury activates enhancer-of-split and bearded complex genes independent of the notch receptor. Toxicol Sci 2008; 104:163-76. [PMID: 18367466 DOI: 10.1093/toxsci/kfn060] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmercury (MeHg) is a persistent environmental toxin that has targeted effects on fetal neural development. Although a number of cytotoxic mechanisms of MeHg have been characterized in cultured cells, its mode of action in the developing nervous system in vivo is less clear. Studies of MeHg-affected rodent and human brains show disrupted cortical and cerebellar architecture suggestive of mechanisms that augment cell signaling pathways potentially affecting cell migration and proliferation. We previously identified the Notch receptor pathway, a highly conserved signaling mechanism fundamental for neural development, as a target for MeHg-induced signaling in Drosophila neural cell lines. Here we have expanded our use of the Drosophila model to resolve a broader spectrum of transcriptional changes resulting from MeHg exposure in vivo and in vitro. Several Notch target genes within the Enhancer-of-split (E(spl)C) and Bearded (BrdC) complexes are upregulated with MeHg exposure in the embryo and in cultured neural cells. However, the profile of MeHg-induced E(spl)C and BrdC gene expression differs significantly from that seen with activation of the Notch receptor. Targeted knockdown of Notch and of the downstream coactivator Suppressor of Hairless (Su(H)), shows no effect on MeHg-induced transcription, indicating a novel Notch-independent mechanism of action for MeHg. MeHg transcriptional activation is partially mimicked by iodoacetamide but not by N-ethylmaleimide, two thiol-specific electrophiles, revealing a degree of specificity of cellular thiol targets in MeHg-induced transcriptional events.
Collapse
Affiliation(s)
- Matthew D Rand
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, Burlington, VT 05405, USA.
| | | | | |
Collapse
|
37
|
Guzzi G, La Porta CAM. Molecular mechanisms triggered by mercury. Toxicology 2007; 244:1-12. [PMID: 18077077 DOI: 10.1016/j.tox.2007.11.002] [Citation(s) in RCA: 187] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Accepted: 11/02/2007] [Indexed: 11/24/2022]
Abstract
Mercury is an ubiquitous environmental toxin that causes a wide range of adverse health effects in humans. Three forms of mercury exist: elemental, inorganic and organic. Each of them has its own profile of toxicity. Exposure to mercury typically occurs by inhalation or ingestion. Mercury can be an indoor air pollutant, however industry emission remains the most important source of inhaled mercury. Furthermore, fresh water and ocean fish may contain large amounts of mercury and dental amalgam can be another important source of inorganic and mercury vapor. The present review discusses the current information on mercury toxicity and the distinct toxicologic profile of the three forms of mercury. The existing therapeutics, new therapeutics development or agents for treating mercury poisoning will also discussed. Since in general low levels of mercurial are tolerable, herein, we also discuss the defensive mechanisms developed by the cell to protect itself against mercury injury. This aspect may be useful to provide a biological protection against toxic effects exerted by mercury or by specific forms of mercury in view of a medicinal purposes.
Collapse
Affiliation(s)
- GianPaolo Guzzi
- AIRMEB Italian Association for Metals and Biocompatibility Research, Italy
| | | |
Collapse
|
38
|
Johansson C, Castoldi AF, Onishchenko N, Manzo L, Vahter M, Ceccatelli S. Neurobehavioural and molecular changes induced by methylmercury exposure during development. Neurotox Res 2007; 11:241-60. [PMID: 17449462 DOI: 10.1007/bf03033570] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is an increasing body of evidence on the possible environmental influence on neurodevelopmental and neurodegenerative disorders. Both experimental and epidemiological studies have demonstrated the distinctive susceptibility of the developing brain to environmental factors such as lead, mercury and polychlorinated biphenyls at levels of exposure that have no detectable effects in adults. Methylmercury (MeHg) has long been known to affect neurodevelopment in both humans and experimental animals. Neurobehavioural effects reported include altered motoric function and memory and learning disabilities. In addition, there is evidence from recent experimental neurodevelopmental studies that MeHg can induce depression-like behaviour. Several mechanisms have been suggested from in vivo- and in vitro-studies, such as effects on neurotransmitter systems, induction of oxidative stress and disruption of microtubules and intracellular calcium homeostasis. Recent in vitro data show that very low levels of MeHg can inhibit neuronal differentiation of neural stem cells. This review summarises what is currently known about the neurodevelopmental effects of MeHg and consider the strength of different experimental approaches to study the effects of environmentally relevant exposure in vivo and in vitro.
Collapse
Affiliation(s)
- Carolina Johansson
- Division of Toxicology and Neurotoxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|