1
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
2
|
Leal-Nazaré CG, Arrifano GP, Lopes-Araújo A, Santos-Sacramento L, Barthelemy JL, Soares-Silva I, Crespo-Lopez ME, Augusto-Oliveira M. Methylmercury neurotoxicity: Beyond the neurocentric view. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 920:170939. [PMID: 38365040 DOI: 10.1016/j.scitotenv.2024.170939] [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: 12/14/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Mercury is a highly toxic metal widely used in human activities worldwide, therefore considered a global public health problem. Many cases of mercury intoxication have occurred in history and represent a huge challenge nowadays. Of particular importance is its methylated form, methylmercury (MeHg). This mercurial species induces damage to several organs in the human body, especially to the central nervous system. Neurological impairments such as executive, memory, motor and visual deficits are associated with MeHg neurotoxicity. Molecular mechanisms involved in MeHg-induced neurotoxicity include excitotoxicity due to glutamatergic imbalance, disturbance in calcium homeostasis and oxidative balance, failure in synaptic support, and inflammatory response. Although neurons are largely affected by MeHg intoxication, they only represent half of the brain cells. Glial cells represent roughly 50 % of the brain cells and are key elements in the functioning of the central nervous system. Particularly, astrocytes and microglia are deeply involved in MeHg-induced neurotoxicity, resulting in distinct neurological outcomes depending on the context. In this review, we discuss the main findings on astroglial and microglial involvement as mediators of neuroprotective and neurotoxic responses to MeHg intoxication. The literature shows that these responses depend on chemical and morphophysiological features, thus, we present some insights for future investigations, considering the particularities of the context, including time and dose of exposure, brain region, and species of study.
Collapse
Affiliation(s)
- Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Amanda Lopes-Araújo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Jean Ludger Barthelemy
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Isabela Soares-Silva
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| | - Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| |
Collapse
|
3
|
Shinoda Y, Akiyama M, Toyama T. Potential Association between Methylmercury Neurotoxicity and Inflammation. Biol Pharm Bull 2023; 46:1162-1168. [PMID: 37661394 DOI: 10.1248/bpb.b23-00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Methylmercury (MeHg) is the causal substrate of Minamata disease and a major environmental toxicant. MeHg is widely distributed, mainly in the ocean, meaning its bioaccumulation in seafood is a considerable problem for human health. MeHg has been intensively investigated and is known to induce inflammatory responses and neurodegeneration. However, the relationship between MeHg-induced inflammatory responses and neurodegeneration is not understood. In the present review, we first describe recent findings showing an association between inflammatory responses and certain MeHg-unrelated neurological diseases caused by neurodegeneration. In addition, cell-specific MeHg-induced inflammatory responses are summarized for the central nervous system including those of microglia, astrocytes, and neurons. We also describe MeHg-induced inflammatory responses in peripheral cells and tissue, such as macrophages and blood. These findings provide a concept of the relationship between MeHg-induced inflammatory responses and neurodegeneration, as well as direction for future research of MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Masahiro Akiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University
| | - Takashi Toyama
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
4
|
Martins B, Novo JP, Fonseca É, Raposo R, Sardão VA, Pereira F, Oriá RB, Fontes-Ribeiro C, Malva J. Necrotic-like BV-2 microglial cell death due to methylmercury exposure. Front Pharmacol 2022; 13:1003663. [PMID: 36408241 PMCID: PMC9667718 DOI: 10.3389/fphar.2022.1003663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 12/01/2022] Open
Abstract
Methylmercury (MeHg) is a dangerous environmental contaminant with strong bioaccumulation in the food chain and neurotoxic properties. In the nervous system, MeHg may cause neurodevelopment impairment and potentially interfere with immune response, compromising proper control of neuroinflammation and aggravating neurodegeneration. Human populations are exposed to environmental contamination with MeHg, especially in areas with strong mining or industrial activity, raising public health concerns. Taking this into consideration, this work aims to clarify pathways leading to acute toxic effects caused by MeHg exposure in microglial cells. BV-2 mouse microglial cells were incubated with MeHg at different concentrations (0.01, 0.1, 1 and 10 µM) for 1 h prior to continuous Lipopolysaccharide (LPS, 0.5 μg/ml) exposure for 6 or 24 h. After cell exposure, reactive oxygen species (ROS), IL-6 and TNF-α cytokines production, inducible nitric oxide synthase (iNOS) expression, nitric oxide (NO) release, metabolic activity, propidium iodide (PI) uptake, caspase-3 and -9 activities and phagocytic activity were assessed. MeHg 10 µM decreased ROS formation, the production and secretion of pro-inflammatory cytokines IL-6, TNF-α, iNOS immunoreactivity, the release of NO in BV-2 cells. Furthermore, MeHg 10 µM decreased the metabolic activity of BV-2 and increased the number of PI-positive cells (necrotic-like cell death) when compared to the respective control group. Besides, MeHg did not interfere with caspase activity or the phagocytic profile of cells. The short-term effects of a high concentration of MeHg on BV-2 microglial cells lead to impaired production of several pro-inflammatory mediators, as well as a higher microglial cell death via necrosis, compromising their neuroinflammatory response. Clarifying the mechanisms underlying MeHg-induced neurotoxicity and neurodegeneration in brain cells is relevant to better understand acute and long-term chronic neuroinflammatory responses following MeHg exposure.
Collapse
Affiliation(s)
- B. Martins
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - J. P. Novo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - É. Fonseca
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. Raposo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,Experimental Biology Core, Health Sciences Center, University of Fortaleza, Fortaleza, Brazil
| | - V. A. Sardão
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,Center for Neuroscience and Cell Biology (CNC), UC Biotech, University of Coimbra, Cantanhede, Portugal
| | - F. Pereira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. B. Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - C. Fontes-Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - J. Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,*Correspondence: J. Malva,
| |
Collapse
|
5
|
Walker EV, Girgis S, Yuan Y, Goodman KJ. Community-driven research in the canadian arctic: dietary exposure to methylmercury and gastric health outcomes. Int J Circumpolar Health 2021; 80:1889879. [PMID: 33646085 PMCID: PMC7928021 DOI: 10.1080/22423982.2021.1889879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/24/2021] [Accepted: 02/09/2021] [Indexed: 10/31/2022] Open
Abstract
Indigenous Arctic Canadians have a higher prevalence of gastric neoplasms relative to North Americans of European ancestry. We investigated the hypothesis that low-dose methylmercury exposure from eating fish/whale increases the risk of gastric cancer in Arctic communities. We used intermediate endpoints from an established model of gastric carcinogenesis: intestinal metaplasia, atrophy, and severe chronic gastritis. During 2008-2012, we obtained gastric biopsies from participants of community-driven projects in 3 communities. In 2016, we collected hair samples to measure methylmercury levels and interviewed them about diet. In cross-sectional analysis, logistic regression estimated odds ratios for the estimated effect of hair-methylmercury concentration on the prevalence of each pathology outcome stratified by selenium intake. Among 80 participants, prevalence of intestinal metaplasia, atrophy and severe chronic gastritis was 17, 29 and 38%, respectively. Adjusted Odds of severe chronic gastritis and atrophy were highest at hair-methylmercury concentrations ≥1μg/g when estimated selenium intake was 0, and approached 0 for all methylmercury levels as estimated selenium intake increased. Gastric pathology increased with methylmercury exposure when selenium intake was low. Though limited by small numbers, these findings suggest selenium ingested by eating fish/whale may counter harmful effects of methylmercury exposure in Arctic populations.
Collapse
Affiliation(s)
- Emily V. Walker
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Safwat Girgis
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Yan Yuan
- Department of Public Health Sciences, University of Alberta, Edmonton, Canada
| | - Karen J. Goodman
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Public Health Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Novo JP, Martins B, Raposo RS, Pereira FC, Oriá RB, Malva JO, Fontes-Ribeiro C. Cellular and Molecular Mechanisms Mediating Methylmercury Neurotoxicity and Neuroinflammation. Int J Mol Sci 2021; 22:ijms22063101. [PMID: 33803585 PMCID: PMC8003103 DOI: 10.3390/ijms22063101] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Methylmercury (MeHg) toxicity is a major environmental concern. In the aquatic reservoir, MeHg bioaccumulates along the food chain until it is consumed by riverine populations. There has been much interest in the neurotoxicity of MeHg due to recent environmental disasters. Studies have also addressed the implications of long-term MeHg exposure for humans. The central nervous system is particularly susceptible to the deleterious effects of MeHg, as evidenced by clinical symptoms and histopathological changes in poisoned humans. In vitro and in vivo studies have been crucial in deciphering the molecular mechanisms underlying MeHg-induced neurotoxicity. A collection of cellular and molecular alterations including cytokine release, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate dyshomeostasis, and cell death mechanisms are important consequences of brain cells exposure to MeHg. The purpose of this review is to organize an overview of the mercury cycle and MeHg poisoning events and to summarize data from cellular, animal, and human studies focusing on MeHg effects in neurons and glial cells. This review proposes an up-to-date compendium that will serve as a starting point for further studies and a consultation reference of published studies.
Collapse
Affiliation(s)
- João P. Novo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Beatriz Martins
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Ramon S. Raposo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Experimental Biology Core, University of Fortaleza, Health Sciences, Fortaleza 60110-001, Brazil
| | - Frederico C. Pereira
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil;
| | - João O. Malva
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| | - Carlos Fontes-Ribeiro
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| |
Collapse
|
7
|
Bjørklund G, Pivina L, Dadar M, Semenova Y, Chirumbolo S, Aaseth J. Mercury Exposure, Epigenetic Alterations and Brain Tumorigenesis: A Possible Relationship? Curr Med Chem 2020; 27:6596-6610. [DOI: 10.2174/0929867326666190930150159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/11/2019] [Accepted: 08/30/2019] [Indexed: 12/09/2022]
Abstract
The risk assessment of mercury (Hg), in both wildlife and humans, represents an increasing
challenge. Increased production of Reactive Oxygen Species (ROS) is a known Hg-induced
toxic effect, which can be accentuated by other environmental pollutants and by complex interactions
between environmental and genetic factors. Some epidemiological and experimental studies
have investigated a possible correlation between brain tumors and heavy metals. Epigenetic modifications
in brain tumors include aberrant activation of genes, hypomethylation of specific genes,
changes in various histones, and CpG hypermethylation. Also, Hg can decrease the bioavailability
of selenium and induce the generation of reactive oxygen that plays important roles in different
pathological processes. Modification of of metals can induce excess ROS and cause lipid peroxidation,
alteration of proteins, and DNA damage. In this review, we highlight the possible relationship
between Hg exposure, epigenetic alterations, and brain tumors.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | | | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| |
Collapse
|
8
|
Muniroh M. Methylmercury-induced pro-inflammatory cytokines activation and its preventive strategy using anti-inflammation N-acetyl-l-cysteine: a mini-review. REVIEWS ON ENVIRONMENTAL HEALTH 2020; 35:233-238. [PMID: 32710722 DOI: 10.1515/reveh-2020-0026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/09/2020] [Indexed: 06/11/2023]
Abstract
The exposure of methylmercury (MeHg) has become a public health concern because of its neurotoxic effect. Various neurological symptoms were detected in Minamata disease patients, who got intoxicated by MeHg, including paresthesia, ataxia, gait disturbance, sensory disturbances, tremors, visual, and hearing impairments, indicating that MeHg could pass the blood-brain barrier (BBB) and cause impairment of neurons and other brain cells. Previous studies have reported some expected mechanisms of MeHg-induced neurotoxicity including the neuroinflammation pathway. It was characterized by the up-regulation of numerous pro-inflammatory cytokines expression. Therefore, the use of anti-inflammatories such as N-acetyl-l-cysteine (NAC) may act as a preventive compound to protect the brain from MeHg harmful effects. This mini-review will explain detailed information on MeHg-induced pro-inflammatory cytokines activation as well as possible preventive strategies using anti-inflammation NAC to protect brain cells, particularly in in vivo and in vitro studies.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Physiology, Faculty of Medicine Diponegoro University, Semarang, Central Java, 50275, Indonesia
| |
Collapse
|
9
|
Ke T, Gonçalves FM, Gonçalves CL, Dos Santos AA, Rocha JBT, Farina M, Skalny A, Tsatsakis A, Bowman AB, Aschner M. Post-translational modifications in MeHg-induced neurotoxicity. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2068-2081. [PMID: 30385410 DOI: 10.1016/j.bbadis.2018.10.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Mercury (Hg) exposure remains a major public health concern due to its widespread distribution in the environment. Organic mercurials, such as MeHg, have been extensively investigated especially because of their congenital effects. In this context, studies on the molecular mechanism of MeHg-induced neurotoxicity are pivotal to the understanding of its toxic effects and the development of preventive measures. Post-translational modifications (PTMs) of proteins, such as phosphorylation, ubiquitination, and acetylation are essential for the proper function of proteins and play important roles in the regulation of cellular homeostasis. The rapid and transient nature of many PTMs allows efficient signal transduction in response to stress. This review summarizes the current knowledge of PTMs in MeHg-induced neurotoxicity, including the most commonly PTMs, as well as PTMs induced by oxidative stress and PTMs of antioxidant proteins. Though PTMs represent an important molecular mechanism for maintaining cellular homeostasis and are involved in the neurotoxic effects of MeHg, we are far from understanding the complete picture on their role, and further research is warranted to increase our knowledge of PTMs in MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Cinara Ludvig Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | | | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, 97105900 Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040900 Florianópolis, SC, Brazil
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
10
|
Sato M, Toyama T, Lee JY, Miura N, Naganuma A, Hwang GW. Activation of ornithine decarboxylase protects against methylmercury toxicity by increasing putrescine. Toxicol Appl Pharmacol 2018; 356:120-126. [DOI: 10.1016/j.taap.2018.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/13/2018] [Accepted: 08/01/2018] [Indexed: 11/15/2022]
|
11
|
Tucker EK, Nowak RA. Methylmercury alters proliferation, migration, and antioxidant capacity in human HTR8/SV-neo trophoblast cells. Reprod Toxicol 2018; 78:60-68. [PMID: 29581082 DOI: 10.1016/j.reprotox.2018.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/01/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Abstract
Methylmercury, a potent neurotoxin, is able to pass through the placenta, but its effects on the placenta itself have not been elucidated. Using an immortalized human trophoblast cell line, HTR8/SV-neo, we assessed the in vitro toxicity of methylmercury. We found that 1 μg/mL methylmercury decreased viability, proliferation, and migration; and it had effects on antioxidant genes similar to those seen in neural cells. However, methylmercury led to decreased expression of superoxide dismutase 1 and increased expression of surfactant protein D. HTR cells treated 0.01 or 0.1 μg/mL methylmercury had increased migration rates along with decreased expression of an adhesion gene, cadherin 3, suggesting that low doses of methylmercury promote migration in HTR cells. Our results indicate that trophoblast cells react differently to methylmercury relative to neural cell lines, and thus investigation of methylmercury toxicity in placental cells is needed to understand the effects of this heavy metal on the placenta.
Collapse
Affiliation(s)
- Emily K Tucker
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207, W. Gregory Dr., Urbana, Illinois, USA.
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207, W. Gregory Dr., Urbana, Illinois, USA
| |
Collapse
|
12
|
Bjørklund G, Dadar M, Mutter J, Aaseth J. The toxicology of mercury: Current research and emerging trends. ENVIRONMENTAL RESEARCH 2017; 159:545-554. [PMID: 28889024 DOI: 10.1016/j.envres.2017.08.051] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/27/2017] [Accepted: 08/30/2017] [Indexed: 05/16/2023]
Abstract
Mercury (Hg) is a persistent bio-accumulative toxic metal with unique physicochemical properties of public health concern since their natural and anthropogenic diffusions still induce high risk to human and environmental health. The goal of this review was to analyze scientific literature evaluating the role of global concerns over Hg exposure due to human exposure to ingestion of contaminated seafood (methyl-Hg) as well as elemental Hg levels of dental amalgam fillings (metallic Hg), vaccines (ethyl-Hg) and contaminated water and air (Hg chloride). Mercury has been recognized as a neurotoxicant as well as immunotoxic and designated by the World Health Organization as one of the ten most dangerous chemicals to public health. It has been shown that the half-life of inorganic Hg in human brains is several years to several decades. Mercury occurs in the environment under different chemical forms as elemental Hg (metallic), inorganic and organic Hg. Despite the raising understanding of the Hg toxicokinetics, there is still fully justified to further explore the emerging theories about its bioavailability and adverse effects in humans. In this review, we describe current research and emerging trends in Hg toxicity with the purpose of providing up-to-date information for a better understanding of the kinetics of this metal, presenting comprehensive knowledge on published data analyzing its metabolism, interaction with other metals, distribution, internal doses and targets, and reservoir organs.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610 Mo i Rana, Norway.
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | | | - Jan Aaseth
- Innlandet Hospital Trust and Inland Norway University of Applied Sciences, Elverum, Norway
| |
Collapse
|
13
|
The Putative Role of Environmental Mercury in the Pathogenesis and Pathophysiology of Autism Spectrum Disorders and Subtypes. Mol Neurobiol 2017; 55:4834-4856. [DOI: 10.1007/s12035-017-0692-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023]
|
14
|
Risher JF, Tucker P. Alkyl Mercury-Induced Toxicity: Multiple Mechanisms of Action. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2017; 240:105-149. [PMID: 27161558 PMCID: PMC10508330 DOI: 10.1007/398_2016_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
There are a number of mechanisms by which alkylmercury compounds cause toxic action in the body. Collectively, published studies reveal that there are some similarities between the mechanisms of the toxic action of the mono-alkyl mercury compounds methylmercury (MeHg) and ethylmercury (EtHg). This paper represents a summary of some of the studies regarding these mechanisms of action in order to facilitate the understanding of the many varied effects of alkylmercurials in the human body. The similarities in mechanisms of toxicity for MeHg and EtHg are presented and compared. The difference in manifested toxicity of MeHg and EtHg are likely the result of the differences in exposure, metabolism, and elimination from the body, rather than differences in mechanisms of action between the two.
Collapse
Affiliation(s)
- John F Risher
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, 1600 Clifton Road (MS F-58), Atlanta, GA, 30333, USA.
| | - Pamela Tucker
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, 1600 Clifton Road (MS F-58), Atlanta, GA, 30333, USA
| |
Collapse
|
15
|
Muniroh M, Khan N, Koriyama C, Akiba S, Vogel CFA, Yamamoto M. Suppression of methylmercury-induced IL-6 and MCP-1 expressions by N-acetylcysteine in U-87MG human astrocytoma cells. Life Sci 2015; 134:16-21. [PMID: 26006043 DOI: 10.1016/j.lfs.2015.04.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 04/09/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
Abstract
AIMS The aim of this study was to clarify the involvement of oxidative stress in methylmercury (MeHg)-induced pro-inflammatory cytokine expressions and the suppressive effects of N-acetylcysteine (NAC) in MeHg-induced cytokine expression. MATERIALS AND METHODS Using U-87-MG human astrocytoma cell line, interleukin (IL)-6 and monocyte chemoattractant protein (MCP)-1 expressions induced by 4 μM MeHg were measured at mRNA and protein levels. Hydrogen peroxide (H2O2) and superoxide anion (O2(-)) were quantified by flow-cytometry analysis. To examine the suppressive effects of NAC on the cytokine expressions among different timing of NAC treatment, cells were treated with 0.5 or 5mM NAC before, simultaneously, or after MeHg administration. KEY FINDINGS MeHg exposure at 4 μM, a non-cytotoxic concentration, significantly induced MCP-1 and IL-6 expressions at both mRNA and protein levels. A significant increase of H2O2 production but not O2(-) was observed. MeHg-induced expression of MCP-1 and IL-6 mRNA was reduced by 10-20% in the presence of 5mM NAC (co-treatment experiment) compared to cells treated with MeHg only. Pre-treatment of cells with 0.5 or 5mM NAC at 0.5 or 1h and its subsequent washout before MeHg addition suppressed MCP-1 and IL-6 cytokine expressions. Post-treatment of cells with NAC after MeHg addition also suppressed the cytokine induction, but the magnitude of suppression was evidently lower than in co-treated cells even though the H2O2 generation was almost completely suppressed by NAC. SIGNIFICANCE NAC may effectively suppress the MeHg-induced cytokine production through both, inhibition of reactive oxygen species as well as extracellular chelation of MeHg in astrocytes.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan; Department of Physiology, Faculty of Medicine, Diponegoro University, Tembalang, Semarang 50725, Indonesia
| | - Noureen Khan
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan.
| | - Suminori Akiba
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | - Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan
| |
Collapse
|
16
|
Kanda H, Shinkai Y, Kumagai Y. S-Mercuration of cellular proteins by methylmercury and its toxicological implications. J Toxicol Sci 2014; 39:687-700. [DOI: 10.2131/jts.39.687] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Hironori Kanda
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba
| | - Yasuhiro Shinkai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba
| |
Collapse
|
17
|
Ondovcik SL, Preston TJ, McCallum GP, Wells PG. Expression of human oxoguanine glycosylase 1 or formamidopyrimidine glycosylase in human embryonic kidney 293 cells exacerbates methylmercury toxicity in vitro. Toxicol Appl Pharmacol 2013; 271:41-8. [DOI: 10.1016/j.taap.2013.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/19/2013] [Accepted: 04/01/2013] [Indexed: 10/26/2022]
|
18
|
Ondovcik SL, Tamblyn L, McPherson JP, Wells PG. Sensitivity to methylmercury toxicity is enhanced in oxoguanine glycosylase 1 knockout murine embryonic fibroblasts and is dependent on cellular proliferation capacity. Toxicol Appl Pharmacol 2013; 270:23-30. [DOI: 10.1016/j.taap.2013.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 02/20/2013] [Accepted: 02/22/2013] [Indexed: 12/16/2022]
|
19
|
Effect of marine omega 3 fatty acids on methylmercury-induced toxicity in fish and mammalian cells in vitro. J Biomed Biotechnol 2012; 2012:417652. [PMID: 22654480 PMCID: PMC3359764 DOI: 10.1155/2012/417652] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 02/29/2012] [Indexed: 11/18/2022] Open
Abstract
Methylmercury (MeHg) is a ubiquitous environmental contaminant which bioaccumulates in marine biota. Fish constitute an important part of a balanced human diet contributing with health beneficial nutrients but may also contain contaminants such as MeHg. Interactions between the marine n-3 fatty acids eicosapentaenoic acid (20:5n-3, EPA) and docosahexaenoic acid (22:6n-3, DHA) with MeHg-induced toxicity were investigated. Different toxic and metabolic responses were studied in Atlantic salmon kidney (ASK) cell line and the mammalian kidney-derived HEK293 cell line. Both cell lines were preincubated with DHA or EPA prior to MeHg-exposure, and cell toxicity was assessed differently in the cell lines by MeHg-uptake in cells (ASK and HEK293), proliferation (HEK293 and ASK), apoptosis (ASK), oxidation of the red-ox probe roGFP (HEK293), and regulation of selected toxicological and metabolic transcriptional markers (ASK). DHA was observed to decrease the uptake of MeHg in HEK293, but not in ASK cells. DHA also increased, while EPA decreased, MeHg-induced apoptosis in ASK. MeHg exposure induced changes in selected metabolic and known MeHg biomarkers in ASK cells. Both DHA and MeHg, but not EPA, oxidized roGFP in HEK293 cells. In conclusion, marine n-3 fatty acids may ameliorate MeHg toxicity, either by decreasing apoptosis (EPA) or by reducing MeHg uptake (DHA). However, DHA can also augment MeHg toxicity by increasing oxidative stress and apoptosis when combined with MeHg.
Collapse
|
20
|
Abstract
Autism spectrum disorders (ASDs) including classic autism is a group of complex developmental disabilities with core deficits of impaired social interactions, communication difficulties and repetitive behaviors. Although the neurobiology of ASDs has attracted much attention in the last two decades, the role of microglia has been ignored. Existing data are focused on their recognized role in neuroinflammation, which only covers a small part of the pathological repertoire of microglia. This review highlights recent findings on the broader roles of microglia, including their active surveillance of brain microenvironments and regulation of synaptic connectivity, maturation of brain circuitry and neurogenesis. Emerging evidence suggests that microglia respond to pre- and postnatal environmental stimuli through epigenetic interface to change gene expression, thus acting as effectors of experience-dependent synaptic plasticity. Impairments of these microglial functions could substantially contribute to several major etiological factors of autism, such as environmental toxins and cortical underconnectivity. Our recent study on Rett syndrome, a syndromic autistic disorder, provides an example that intrinsic microglial dysfunction due to genetic and epigenetic aberrations could detrimentally affect the developmental trajectory without evoking neuroinflammation. We propose that ASDs provide excellent opportunities to study the influence of microglia on neurodevelopment, and this knowledge could lead to novel therapies.
Collapse
|
21
|
Ondovcik SL, Tamblyn L, McPherson JP, Wells PG. Oxoguanine glycosylase 1 (OGG1) protects cells from DNA double-strand break damage following methylmercury (MeHg) exposure. Toxicol Sci 2012; 128:272-83. [PMID: 22523232 DOI: 10.1093/toxsci/kfs138] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Methylmercury (MeHg) is a potent neurotoxin, teratogen, and probable carcinogen, but the underlying mechanisms of its actions remain unclear. Although MeHg causes several types of DNA damage, the toxicological consequences of this macromolecular damage are unknown. MeHg enhances oxidative stress, which can cause various oxidative DNA lesions that are primarily repaired by oxoguanine glycosylase 1 (OGG1). Herein, we compared the response of wild-type and OGG1 null (Ogg1(-/-)) murine embryonic fibroblasts to environmentally relevant, low micromolar concentrations of MeHg by measuring clonogenic efficiency, cell cycle arrest, DNA double-strand breaks (DSBs), and activation of the DNA damage response pathway.Ogg1(-/-) cells exhibited greater sensitivity to MeHg than wild-type controls, as measured by the clonogenic assay, and showed a greater propensity for MeHg-initiated apoptosis. Both wild-type and Ogg1(-/-) cells underwent cell cycle arrest when exposed to micromolar concentrations of MeHg; however, the extent of DSBs was exacerbated in Ogg1(-/-) cells compared with that in wild-type controls. Pretreatment with the antioxidative enzyme catalase reduced levels of DSBs in both wild-type and Ogg1(-/-) cells but failed to block MeHg-initiated apoptosis at micromolar concentrations. Our findings implicate reactive oxygen species mediated DNA damage in the mechanism of MeHg toxicity; and demonstrate for the first time that impaired DNA repair capacity enhances cellular sensitivity to MeHg. Accordingly, the genotoxic properties of MeHg may contribute to its neurotoxic and teratogenic effects, and an individual's response to oxidative stress and DNA damage may constitute an important determinant of risk.
Collapse
Affiliation(s)
- Stephanie L Ondovcik
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
22
|
Farina M, Aschner M, Rocha JBT. Oxidative stress in MeHg-induced neurotoxicity. Toxicol Appl Pharmacol 2011; 256:405-17. [PMID: 21601588 PMCID: PMC3166649 DOI: 10.1016/j.taap.2011.05.001] [Citation(s) in RCA: 256] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/01/2011] [Accepted: 05/02/2011] [Indexed: 12/20/2022]
Abstract
Methylmercury (MeHg) is an environmental toxicant that leads to long-lasting neurological and developmental deficits in animals and humans. Although the molecular mechanisms mediating MeHg-induced neurotoxicity are not completely understood, several lines of evidence indicate that oxidative stress represents a critical event related to the neurotoxic effects elicited by this toxicant. The objective of this review is to summarize and discuss data from experimental and epidemiological studies that have been important in clarifying the molecular events which mediate MeHg-induced oxidative damage and, consequently, toxicity. Although unanswered questions remain, the electrophilic properties of MeHg and its ability to oxidize thiols have been reported to play decisive roles to the oxidative consequences observed after MeHg exposure. However, a close examination of the relationship between low levels of MeHg necessary to induce oxidative stress and the high amounts of sulfhydryl-containing antioxidants in mammalian cells (e.g., glutathione) have led to the hypothesis that nucleophilic groups with extremely high affinities for MeHg (e.g., selenols) might represent primary targets in MeHg-induced oxidative stress. Indeed, the inhibition of antioxidant selenoproteins during MeHg poisoning in experimental animals has corroborated this hypothesis. The levels of different reactive species (superoxide anion, hydrogen peroxide and nitric oxide) have been reported to be increased in MeHg-exposed systems, and the mechanisms concerning these increments seem to involve a complex sequence of cascading molecular events, such as mitochondrial dysfunction, excitotoxicity, intracellular calcium dyshomeostasis and decreased antioxidant capacity. This review also discusses potential therapeutic strategies to counteract MeHg-induced toxicity and oxidative stress, emphasizing the use of organic selenocompounds, which generally present higher affinity for MeHg when compared to the classically studied agents.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Pediatrics and Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - João B. T. Rocha
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
23
|
Bassett T, Bach P, Chan HM. Effects of methylmercury on the secretion of pro-inflammatory cytokines from primary microglial cells and astrocytes. Neurotoxicology 2011; 33:229-34. [PMID: 22037494 DOI: 10.1016/j.neuro.2011.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 10/10/2011] [Accepted: 10/13/2011] [Indexed: 10/16/2022]
Abstract
Glial cells, including oligodendrocytes, astrocytes and microglia are important to proper central nervous system (CNS) function. Deregulation or changes to CNS populations of astrocytes and microglia in particular are expected to play a role in many neurodegenerative diseases, including Parkinson's disease, amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD). Previous studies have reported methylmercury (MeHg) induced changes in glial cell function; however, the effects of MeHg on these cells remains poorly understood. This study aims to examine the effect of MeHg on the secretion of pro-inflammatory cytokines from microglia and astrocytes. The impact of the microglia/astrocyte ratio on cytokine secretion was also examined. Microglia and astrocytes were cultured from the brains of neo-natal BALB/C mice and dosed with MeHg (0-1 μM) and stimulated with PAM(3)CSK(4) (PAM(3)), a toll-like receptor (TLR) ligand. After this, the secretion of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α) and interleukin-1-beta (IL-1β) was measured by ELISA. MeHg reduced the secretion of IL-6 in a dose dependant manner but did not effect the secretion of TNF-α. No change in IL-1β was observed in any treatments, indicating that PAM(3) cannot induce the secretion of this cytokine from glial cells. Additionally, the ratio of microglia/astrocyte had an effect on the secretion of IL-6 but not TNF-α. These results indicate that MeHg can modify the response of glial cells and the interactions with astrocytes can affect the response of the microglia cells in culture. These results are significant in understanding the potential relationship with MeHg and neurodegenerative diseases and for the interpretation of results of future in vitro studies using monoculture.
Collapse
Affiliation(s)
- Tyler Bassett
- Community Health Sciences Program, University of Northern British Columbia, Prince George, BC, Canada
| | | | | |
Collapse
|
24
|
Godefroy D, Gosselin RD, Yasutake A, Fujimura M, Combadière C, Maury-Brachet R, Laclau M, Rakwal R, Melik-Parsadaniantz S, Bourdineaud JP, Rostène W. The chemokine CCL2 protects against methylmercury neurotoxicity. Toxicol Sci 2011; 125:209-18. [PMID: 21976372 DOI: 10.1093/toxsci/kfr252] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Industrial pollution due to heavy metals such as mercury is a major concern for the environment and public health. Mercury, in particular methylmercury (MeHg), primarily affects brain development and neuronal activity, resulting in neurotoxic effects. Because chemokines can modulate brain functions and are involved in neuroinflammatory and neurodegenerative diseases, we tested the possibility that the neurotoxic effect of MeHg may interfere with the chemokine CCL2. We have used an original protocol in young mice using a MeHg-contaminated fish-based diet for 3 months relevant to human MeHg contamination. We observed that MeHg induced in the mice cortex a decrease in CCL2 concentrations, neuronal cell death, and microglial activation. Knock-out (KO) CCL2 mice fed with a vegetal control food already presented a decrease in cortical neuronal cell density in comparison with wild-type animals under similar diet conditions, suggesting that the presence of CCL2 is required for normal neuronal survival. Moreover, KO CCL2 mice showed a pronounced neuronal cell death in response to MeHg. Using in vitro experiments on pure rat cortical neurons in culture, we observed by blockade of the CCL2/CCR2 neurotransmission an increased neuronal cell death in response to MeHg neurotoxicity. Furthermore, we showed that sod genes are upregulated in brain of wild-type mice fed with MeHg in contrast to KO CCL2 mice and that CCL2 can blunt in vitro the decrease in glutathione levels induced by MeHg. These original findings demonstrate that CCL2 may act as a neuroprotective alarm system in brain deficits due to MeHg intoxication.
Collapse
Affiliation(s)
- David Godefroy
- INSERM UMRS 968, Institut de la Vision, Université Pierre et Marie Curie Paris 6, 75012 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lu TH, Hsieh SY, Yen CC, Wu HC, Chen KL, Hung DZ, Chen CH, Wu CC, Su YC, Chen YW, Liu SH, Huang CF. Involvement of oxidative stress-mediated ERK1/2 and p38 activation regulated mitochondria-dependent apoptotic signals in methylmercury-induced neuronal cell injury. Toxicol Lett 2011; 204:71-80. [PMID: 21549813 DOI: 10.1016/j.toxlet.2011.04.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/16/2011] [Accepted: 04/18/2011] [Indexed: 11/18/2022]
Abstract
Methylmercury (MeHg) is well-known for causing irreversible damage in the central nervous system as well as a risk factor for inducing neuronal degeneration. However, the molecular mechanisms of MeHg-induced neurotoxicity remain unclear. Here, we investigated the effects and possible mechanisms of MeHg in the mouse cerebrum (in vivo) and in cultured Neuro-2a cells (in vitro). In vivo study showed that the levels of LPO in the plasma and cerebral cortex significantly increased after administration of MeHg (50μg/kg/day) for 7 consecutive weeks. MeHg could also decrease glutathione level and increase the expressions of caspase-3, -7, and -9, accompanied by Bcl-2 down-regulation and up-regulation of Bax, Bak, and p53. Moreover, treatment of Neuro-2a cells with MeHg significantly reduced cell viability, increased oxidative stress damage, and induced several features of mitochondria-dependent apoptotic signals, including increased sub-G1 hypodiploids, mitochondrial dysfunctions, and the activation of PARP, and caspase cascades. These MeHg-induced apoptotic-related signals could be remarkably reversed by antioxidant NAC. MeHg also increased the phosphorylation of ERK1/2 and p38, but not JNK. Pharmacological inhibitors NAC, PD98059, and SB203580 attenuated MeHg-induced cytotoxicity, ERK1/2 and p38 activation, MMP loss, and caspase-3 activation in Neuro-2a cells. Taken together, these results suggest that the signals of ROS-mediated ERK1/2 and p38 activation regulated mitochondria-dependent apoptotic pathways that are involved in MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tien-Hui Lu
- Graduate Institute of Drug Safety, College of Pharmacy, China Medical University, Taichung 404, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chang JY. Methylmercury-induced IL-6 release requires phospholipase C activities. Neurosci Lett 2011; 496:152-6. [PMID: 21513770 DOI: 10.1016/j.neulet.2011.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 03/23/2011] [Accepted: 04/05/2011] [Indexed: 10/18/2022]
Abstract
Methylmercury (MeHg) is a neurotoxin capable of causing severe damage to the CNS, especially in the developing fetus. Glia in the CNS release a number of cytokines that are important for proper CNS development and function. We reported earlier that MeHg could induce interleukin-6 (IL-6) release in primary mouse glia. This finding is significant considering previous reports indicating that sustained IL-6 exposure could be detrimental to cerebellar granule neurons, one of the major cellular targets of MeHg cytotoxicity. By using pharmacological antagonists against phophatidycholine- and phosphoinositol-specific phospholipase C, the current study indicated that phospholipase C activity was necessary for MeHg-induced IL-6 release. Results from pharmacological antagonists further suggested that the calcium signaling initiated by phospholipase C appeared essential for this event. In contrast, protein kinase C activity did not appear to be important. Even though mitogen-activated protein kinases were important for IL-6 release in some experimental systems, these enzymes did not appear to be required for MeHg-induced IL-6 release in glia. Based on these data and those reported by us and others, there is a possibility that MeHg-induced phospholipase C activation initiates a calcium signaling that causes phospholipase A(2) activation. This, in turn, leads to arachidonic acid and lysophosphatidyl choline generation, both of which are potent inducers for IL-6 release.
Collapse
Affiliation(s)
- Jason Y Chang
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
27
|
Ni M, Aschner M. Neonatal rat primary microglia: isolation, culturing, and selected applications. ACTA ACUST UNITED AC 2011; Chapter 12:Unit 12.17. [PMID: 20960423 DOI: 10.1002/0471140856.tx1217s43] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Microglial cells elaborate trophic factors and cytokines and remove toxins and debris from the extracellular space in the central nervous system, acting analogously to peripheral macrophages. Over the past two decades, increased attention has been directed at the role of microglia, not only in normal physiology, but also in mediating neurotoxicity. Activation of microglia is inherent to multiple neurodegenerative disorders and exposure to toxic compounds. In large measure, these revelations have come about as a result of technologies that enable researchers to obtain high yield and purity primary cultures of rodent microglia. The mechanical isolation protocol discussed in this unit offers an economical method to isolate large amounts of microglia in a short and not too labor-intensive manner. Most importantly, it ensures a high yield of cells with great reproducibility. Given the ever-increasing importance of microglia to the field of neurotoxicology research, the ability to isolate large quantities of primary microglia makes it possible to investigate the role and mechanisms associated with microglial modulation of neurotoxicity. We provide a detailed description on the methods that are routinely used in our laboratory for the isolation and culture of microglia, with emphasis on the steps that are deemed most critical for obtaining pure and healthy cultures.
Collapse
Affiliation(s)
- Mingwei Ni
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
28
|
Zhang L, Li YY, Zeng HC, Li M, Wan YJ, Schluesener HJ, Zhang ZY, Xu SQ. Perfluorooctane Sulfonate Induces Apoptosis in N9 Microglial Cell Line. Int J Toxicol 2010; 30:207-15. [DOI: 10.1177/1091581810387832] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Perfluorooctane sulfonate (PFOS) is an environmental persistent acid found at low levels in human, wildlife, and environmental media samples. To study the apoptosis effects of PFOS on microglia, murine N9 cell line was used as a model in current research. The results showed that PFOS could reduce the cell viability significantly, and the cellular apoptosis induced by PFOS was closely accompanied with dissipation of mitochondria membrane potential, upregulation messenger RNAs (mRNAs) of p53, Bax, caspase 9, and caspase 3, and decreased expression of Bcl-2 mRNA. These results suggested that PFOS could disturb homeostasis of N9 cells, impact mitochondria, and affect gene expression of apoptotic regulators, all of which resulted in a start-up of apoptosis.
Collapse
Affiliation(s)
- Ling Zhang
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Preventive Medicine, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Yuan-yuan Li
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huai-cai Zeng
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Li
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Jian Wan
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Zhi-yuan Zhang
- Institute of Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Shun-qing Xu
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Zhang Y, Gao D, Bolivar VJ, Lawrence DA. Induction of autoimmunity to brain antigens by developmental mercury exposure. Toxicol Sci 2010; 119:270-80. [PMID: 21071724 DOI: 10.1093/toxsci/kfq334] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A.SW mice, which are known to be prone to mercury (Hg)-induced immune nephritis, were assessed for their ability to develop autoimmunity to brain antigens after developmental exposure to Hg. Maternal drinking water containing subclinical doses of 1.25μM methyl Hg (MeHg) or 50μM Hg chloride (HgCl(2)) were used to evaluate developmental (exposure from gestational day 8 to postnatal day 21) induction of immune responses to brain antigens. Only HgCl(2) induced autoantibody production; the HgCl(2)-exposed offspring showed an increased number of CD4(+) splenic T cells expressing CD25 and V(β) 8.3 chains, and the brain-reactive immunoglobulin G (IgG) antibodies were predominantly against nuclear proteins (30 and 34 kD). The antibodies were deposited in all brain regions. Although male and female A.SW mice exposed to HgCl(2) showed deposition of IgG in multiple brain regions, inflammation responses were observed only in the cerebellum (CB) of female A.SW mice; these responses were associated with increased levels of exploratory behavior. The developmental exposure to MeHg also induced inflammation in the CB and increased exploratory behavior of the female A.SW mice, but the change did not correlate with increased IgG in the brain. Interestingly, the non-Hg-exposed female A.SW mice habituated (adapted to the information and/or stimuli of a new environment) more than the male A.SW mice during exploratory behavior assessment, and the Hg exposure eliminated the habituation (i.e., no changes in behavior with subsequent trials), making the female behaviors more like those of the male A.SW mice. Additionally, gender differences in A.SW brain cytokine expressions prior to Hg exposure were eliminated by the Hg exposure.
Collapse
Affiliation(s)
- Yubin Zhang
- The University at Albany School of Public Health, Albany, NY 12201, USA
| | | | | | | |
Collapse
|
30
|
Helmcke KJ, Aschner M. Hormetic effect of methylmercury on Caenorhabditis elegans. Toxicol Appl Pharmacol 2010; 248:156-64. [PMID: 20691719 DOI: 10.1016/j.taap.2010.07.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 07/23/2010] [Accepted: 07/27/2010] [Indexed: 10/19/2022]
Abstract
Research has demonstrated the toxic effects of methylmercury (MeHg), yet molecular mechanisms underlying its toxicity are not completely understood. Caenorhabditis elegans (C. elegans) offers a unique biological model to explore mechanisms of MeHg toxicity given many advantages associated with its ease of use and genetic power. Since our previous work indicated neurotoxic resistance of C. elegans to MeHg, the present study was designed to examine molecular mechanisms associated with this resistance. We hypothesized MeHg would induce expression of gst, hsp or mtl in vivo since glutathione (GSH), heat shock proteins (HSPs), and metallothioneins (MTs) have shown involvement in MeHg toxicity. Our studies demonstrated a modest, but significant increase in fluorescence in gst-4::GFP and mtl-1::GFP strains at an acute, low L1 MeHg exposure, whereas chronic L4 MeHg exposure induced expression of gst-4::GFP and hsp-4::GFP. Knockout gst-4 animals showed no alterations in lethality sensitivity compared to wildtype animals whereas mtl knockouts displayed increased sensitivity to MeHg exposure. GSH levels were increased by acute MeHg treatment and depleted with chronic exposure. We also demonstrate that MeHg induces hormesis, a phenotype whereby a sublethal exposure to MeHg rendered C. elegans resistant to subsequent exposure to the organometal. The involvement of gst-4, hsp-4, mtl-1, and mtl-2 in hormesis was examined. An increase in gst-4::GFP expression after a low-dose acute exposure to MeHg indicated that gst-4 may be involved in this response. Our results implicate GSH, HSPs, and MTs in protecting C. elegans from MeHg toxicity and show a potential role of gst-4 in MeHg-induced hormesis.
Collapse
Affiliation(s)
- Kirsten J Helmcke
- Pharmacology Department and Center in Molecular Toxicology, Vanderbilt University, Nashville, TN 37232, USA.
| | | |
Collapse
|
31
|
Ni M, Li X, Yin Z, Jiang H, Sidoryk-Wegrzynowicz M, Milatovic D, Cai J, Aschner M. Methylmercury induces acute oxidative stress, altering Nrf2 protein level in primary microglial cells. Toxicol Sci 2010; 116:590-603. [PMID: 20421342 DOI: 10.1093/toxsci/kfq126] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The neurotoxicity of methylmercury (MeHg) is well documented in both humans and animals. MeHg causes acute and chronic damage to multiple organs, most profoundly the central nervous system (CNS). Microglial cells are derived from macrophage cell lineage, making up approximately 12% of cells in the CNS, yet their role in MeHg-induced neurotoxicity is not well defined. The purpose of the present study was to characterize microglial vulnerability to MeHg and their potential adaptive response to acute MeHg exposure. We examined the effects of MeHg on microglial viability, reactive oxygen species (ROS) generation, glutathione (GSH) level, redox homeostasis, and Nrf2 protein expression. Our data showed that MeHg (1-5 microM) treatment caused a rapid (within 1 min) concentration- and time-dependent increase in ROS generation, accompanied by a statistically significant decrease in the ratio of GSH and its oxidized form glutathione disulfide (GSSG) (GSH:GSSG ratio). MeHg increased the cytosolic Nrf2 protein level within 1 min of exposure, followed by its nuclear translocation after 10 min of treatment. Consistent with the nuclear translocation of Nrf2, quantitative real-time PCR revealed a concentration-dependent increase in the messenger RNA level of Ho-1, Nqo1, and xCT 30 min post MeHg exposure, whereas Nrf2 knockdown greatly reduced the upregulation of these genes. Furthermore, we observed increased microglial death upon Nrf2 knockdown by the small hairpin RNA approach. Taken together, our study has demonstrated that microglial cells are exquisitely sensitive to MeHg and respond rapidly to MeHg by upregulating the Nrf2-mediated antioxidant response.
Collapse
Affiliation(s)
- Mingwei Ni
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
von Bernhardi R, Tichauer JE, Eugenín J. Aging-dependent changes of microglial cells and their relevance for neurodegenerative disorders. J Neurochem 2009; 112:1099-114. [PMID: 20002526 DOI: 10.1111/j.1471-4159.2009.06537.x] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Among multiple structural and functional brain changes, aging is accompanied by an increase of inflammatory signaling in the nervous system as well as a dysfunction of the immune system elsewhere. Although the long-held view that aging involves neurocognitive impairment is now dismissed, aging is a major risk factor for neurodegenerative diseases such as Alzheimer;s disease, Parkinson;s disease and Huntington's disease, among others. There are many age-related changes affecting the brain, contributing both to certain declining in function and increased frailty, which could singly and collectively affect neuronal viability and vulnerability. Among those changes, both inflammatory responses in aged brains and the altered regulation of toll like receptors, which appears to be relevant for understanding susceptibility to neurodegenerative processes, are linked to pathogenic mechanisms of several diseases. Here, we review how aging and pro-inflammatory environment could modulate microglial phenotype and its reactivity and contribute to the genesis of neurodegenerative processes. Data support our idea that age-related microglial cell changes, by inducing cytotoxicity in contrast to neuroprotection, could contribute to the onset of neurodegenerative changes. This view can have important implications for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta, Santiago, Chile.
| | | | | |
Collapse
|
33
|
Fujimura M, Usuki F, Sawada M, Takashima A. Methylmercury induces neuropathological changes with tau hyperphosphorylation mainly through the activation of the c-jun-N-terminal kinase pathway in the cerebral cortex, but not in the hippocampus of the mouse brain. Neurotoxicology 2009; 30:1000-7. [PMID: 19666049 DOI: 10.1016/j.neuro.2009.08.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 08/01/2009] [Accepted: 08/02/2009] [Indexed: 11/28/2022]
Abstract
Methylmercury (MeHg) is a well-known neurotoxicant inducing neuronal degeneration in the central nervous system. This in vivo study investigated the involvement of tau hyperphosphorylation in MeHg-induced neuropathological changes in the mouse brain, because abnormal tau hyperphosphorylation causes significant pathological changes associated with some neurodegenerative diseases. Mice that were administrated to 30 ppm MeHg in drinking water for 8 weeks exhibited neuropathological changes, e.g. a decrease in the number of neuron; an increase in the number of migratory astrocytes and microglia/macrophages; necrosis and apoptosis in the cerebral cortex, particularly the deep layer of primary motor cortex and prelimbic cortex. Western blotting revealed that MeHg exposure increased tau phosphorylation at Thr-205, Ser-396 and Ser-422 in the cerebral cortex, consistent with the phosphorylation patterns noted in Alzheimer's disease and frontotemporal dementia. Immunohistochemical analyses revealed that the distribution of tau-phosphorylated (Thr-205) neurons corresponded with the areas showing considerable neuropathological changes. Among the kinases and phosphatases related to tau hyperphosphorylation, the activation of mitogen-activated protein kinase kinase 4 (MKK4) and c-Jun N-terminal kinase (JNK) was recognized. Neither neuropathological changes nor tau hyperphosphorylation was detected in the hippocampus in this study although the mercury concentration here was twice that in the cerebral cortex. These findings suggest that MeHg exposure induces tau hyperphosphorylation at specific sites of tau mainly through the activation of JNK pathways, leading to neuropathological changes in the cerebral cortex selectively, but not in the hippocampus of mouse brain.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan.
| | | | | | | |
Collapse
|
34
|
Watanabe J, Nakamachi T, Ogawa T, Naganuma A, Nakamura M, Shioda S, Nakajo S. Characterization of antioxidant protection of cultured neural progenitor cells (NPC) against methylmercury (MeHg) toxicity. J Toxicol Sci 2009; 34:315-25. [PMID: 19483385 DOI: 10.2131/jts.34.315] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Methylmercury (MeHg) is an environmental pollutant known to cause neurobehavioral defects and is especially toxic to the developing brain. With recent studies showing that fetal exposure to low-dose MeHg causes developmental abnormalities, it is therefore important to find ways to combat its effects as well as to clarify the mechanism(s) underlying MeHg toxicity. In the present study, the effects of MeHg on cultured neural progenitor cells (NPC) derived from mouse embryonic brain were investigated. We first confirmed the vulnerability of embryonic NPC to MeHg toxicity, NPC from the telencephalon were more sensitive to MeHg compared to those from the diencephalon. Buthionine sulfoximine (BSO) which is known to inhibit glutathione synthesis accelerated MeHg toxicity. Furthermore, antioxidants such as N-acetyl cysteine and alpha-tocopherol dramatically rescued the NPC from MeHg's toxic effects. Interestingly, a 12 hr delay in the addition of either antioxidant was still able to prevent the cells from undergoing cell death. Although it is now difficult to avoid MeHg exposure from our environment and contaminated foods, taking anti-oxidants from foods or supplements may prevent or diminish the toxicological effects of MeHg.
Collapse
Affiliation(s)
- Jun Watanabe
- Department of Anatomy, School of Medicine, Showa University
| | | | | | | | | | | | | |
Collapse
|
35
|
Chang JY, Tsai PF. IL-6 release from mouse glia caused by MeHg requires cytosolic phospholipase A2 activation. Neurosci Lett 2009; 461:85-9. [PMID: 19539721 DOI: 10.1016/j.neulet.2009.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Revised: 06/02/2009] [Accepted: 06/11/2009] [Indexed: 11/28/2022]
Abstract
Methylmercury is a potent neurotoxin that causes severe neurological disorders in fetuses and young children. Recent studies indicated that MeHg could alter levels of immune mediators produced by cells of the central nervous system. Results from this study indicated that MeHg could greatly induce IL-6 release from primary mouse glial cultures. This property was not shared by other cytotoxic heavy metals, such as CdCl(2) or HgCl(2). MeHg was known to induce cytosolic phospholipase A(2) (PLA(2)) activation and expression, and this enzyme was required for IL-6 induction in some experimental systems. Further experiments using structurally distinct pharmacological agents were performed to test the hypothesis that MeHg induced PLA(2) activation was necessary for MeHg induced IL-6 release. Results indicated that AACOCF(3) (>or=10 microM), MAFP (>or=0.625 microM) and BEL (>or=0.625 microM) significantly reduced MeHg induced IL-6 release in glia. However, these PLA(2) inhibitors did not block MeHg induced GSH depletion. These results suggested that PLA(2) activation was required for MeHg to induce glial IL-6 release.
Collapse
Affiliation(s)
- Jason Y Chang
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | |
Collapse
|
36
|
Ferraro L, Tomasini MC, Tanganelli S, Mazza R, Coluccia A, Carratù MR, Gaetani S, Cuomo V, Antonelli T. Developmental exposure to methylmercury elicits early cell death in the cerebral cortex and long-term memory deficits in the rat. Int J Dev Neurosci 2008; 27:165-74. [PMID: 19084587 DOI: 10.1016/j.ijdevneu.2008.11.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 11/05/2008] [Accepted: 11/12/2008] [Indexed: 01/24/2023] Open
Abstract
Experiments were performed to assess the neurotoxic effects induced by prenatal acute treatment with methylmercury on cortical neurons. To this purpose, primary neuronal cultures were obtained from cerebral cortex of neonatal rats born to dams treated with methylmercury (4 and 8 mg/kg by gavage) on gestational day 15, the developmental stage critical for cortical neuron proliferation. Prenatal exposure to methylmercury 8 mg/kg significantly reduced cell viability and caused either apoptotic or necrotic neuronal death. Moreover, this exposure level resulted in abnormal neurite outgrowth and retraction or collapse of some neurites, caused by a dissolution of microtubules. The severe and early cortical neuron damage induced by methylmercury 8 mg/kg treatment correlated with long term memory impairment, since adult rats (90 days of age) born to dams treated with this dose level showed a significant deficit in the retention performance when subjected to a passive avoidance task. Prenatal exposure to methylmercury 4 mg/kg significantly increased the neuronal vulnerability to a neurotoxic insult. This was determined by measuring the increment of chromatin condensation induced by glutamate, at a concentration (30 microM) able to induce an excitotoxic damage. This exposure level eliciting apoptotic death did not result in cognitive dysfunctions. In conclusion, the methylmercury-induced disruption of glutamate pathway during critical windows of brain development may interfere with cell fate and proliferation resulting in a more or less severe cortical lesions associated or not with loss of function later in life, depending on the exposure levels. Therefore, the early biochemical effects and long-term behavioral changes elicited by high methylmercury levels suggest that the developing brain is impaired in its ability to recover following toxic insult, and the initial effects on cortical neurons may lead to permanent cognitive dysfunctions.
Collapse
Affiliation(s)
- Luca Ferraro
- Department of Clinical and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chang JY, Tsai PF. Prevention of methylmercury-induced mitochondrial depolarization, glutathione depletion and cell death by 15-deoxy-delta-12,14-prostaglandin J(2). Neurotoxicology 2008; 29:1054-61. [PMID: 18778734 DOI: 10.1016/j.neuro.2008.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2008] [Revised: 08/05/2008] [Accepted: 08/08/2008] [Indexed: 12/31/2022]
Abstract
Methylmercury (MeHg) is an environmental toxin that causes severe neurological complications in humans and experimental animals. In addition to neurons, glia in the central nervous system are very susceptible to MeHg toxicity. Pretreatment of glia with the prostaglandin derivative, 15-deoxy-delta-12,14-prostaglandin J(2) (15d-PGJ(2)), caused a significant protection against MeHg cytotoxicity. Results with the C6 glioma cells demonstrated that the protection was dependent on the duration of pretreatment, suggesting that time was required for the up-regulation of cellular defenses. Subsequent experiments indicated that 15d-PGJ(2) prevented MeHg induced mitochondrial depolarization. Similar protection against MeHg cytotoxicity was observed in primary cultures of mouse glia. Analysis of cellular glutathione (GSH) levels indicated that 15d-PGJ(2) caused an up-regulation of GSH and prevented MeHg-induced GSH depletion. Buthionine sulfoximine (BSO), a GSH synthesis inhibitor, completely inhibited the GSH induction by 15d-PGJ(2). However, BSO did not prevent the stabilization of mitochondrial potential and only partially prevented the protection caused by 15d-PGJ(2). While induction of heme oxygenase-1 was implicated in the cytoprotection by 15d-PGJ(2) under some experimental conditions, additional experiments indicated that this enzyme was not involved in the cytoprotection observed in this system. Together, these results suggested that while up-regulation of GSH by 15d-PGJ(2) might help cells to defend against MeHg toxicity, there may be other yet unidentified mechanism(s) initiated by 15d-PGJ(2) treatment that contributed to its protection against MeHg cytotoxicity.
Collapse
Affiliation(s)
- Jason Y Chang
- Department of Neurobiology & Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | |
Collapse
|
38
|
Filipak Neto F, Zanata SM, Silva de Assis HC, Nakao LS, Randi MAF, Oliveira Ribeiro CA. Toxic effects of DDT and methyl mercury on the hepatocytes from Hoplias malabaricus. Toxicol In Vitro 2008; 22:1705-13. [PMID: 18675336 DOI: 10.1016/j.tiv.2008.07.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Revised: 07/04/2008] [Accepted: 07/07/2008] [Indexed: 10/21/2022]
Abstract
Here, we examined the impact of dichlorodiphenyltrichloroethane (DDT) and monomethyl mercury (MeHg) on the redox milieu and survival of hepatocytes from Hoplias malabaricus (traíra). After isolation and attachment of cells, we established one control and four treatments: DDT (50nM of DDT), MeHg I (0.25microM of MeHg), MeHg II (2.5microM of MeHg) and DDT * MeHg I (combination of 50nM of DDT and 0.25microM of MeHg). After four days the exposed hepatocytes presented significantly increased damage in lipids (all treatments), proteins (DDT * MeHg I and MeHg II) and reduced cell viability (all treatments). Also the antioxidant enzymes catalase, glucose-6-phosphate dehydrogenase (G6PDH), glutathione reductase and superoxide dismutase were affected. The current data showed that despite of some protective responses, the increased disturbs on membrane lipids and proteins, increased hydrogen peroxide levels, and decreased glutathione concentration and cell viability strongly indicate oxidative stress as the reason of hepatotoxicity due to DDT and MeHg exposure. In addition, DDT and MeHg together had greater effect than alone when G6PDH and glutathione-S-transferase activities and lipids damage were considered. These findings are indicative of hepatotoxicity occurring at realistic concentrations of DDT and MeHg found in Amazonian fish tissues.
Collapse
Affiliation(s)
- F Filipak Neto
- Departamento de Biologia Celular, Universidade Federal do Paraná, Cx. Postal 19031, CEP 81.531-990 Curitiba, PR, Brazil.
| | | | | | | | | | | |
Collapse
|
39
|
Sakamoto M, Miyamoto KI, Wu Z, Nakanishi H. Possible involvement of cathepsin B released by microglia in methylmercury-induced cerebellar pathological changes in the adult rat. Neurosci Lett 2008; 442:292-6. [PMID: 18638529 DOI: 10.1016/j.neulet.2008.07.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 07/04/2008] [Accepted: 07/08/2008] [Indexed: 11/24/2022]
Abstract
There is increasing evidence that cathepsin B (CB), a lysosomal cysteine protease, is one of the toxic molecules that are secreted by activated microglia. We herein provide evidence that CB released by activated microglia may play a role in the methylmercury (MeHg)-induced pathological changes observed in the cerebellum of the adult rat. Pathological changes tended to progress slowly after treatment with MeHg (5 mg/kg) for 12 consecutive days. At 5 days after the final treatment of MeHg, there was a mild pyknotic change of the granule cells, whereas a marked accumulation of activated microglia was observed in the granule cell layer of the lingual and central lobe. At 8 days after the final treatment, intense pyknotic changes of the granule cells and the accumulation of activated microglia were observed throughout the cerebellar vermis. CB first significantly increased at 3 days after the final treatment of MeHg as the mature form. CB mainly increased in activated microglia which accumulated in the granule cell layer. The coadministration of CA074, an irreversible CB inhibitor, with MeHg significantly reduced the severity of pyknotic changes of the granule cells. Furthermore, primary cultured microglia secreted the mature CB in the culture medium following cellular activation. These observations strongly suggest that CB secreted by activated microglia is thus closely associated with the MeHg-induced severe pyknotic changes of the cerebellar granule cells. The treatment of CA074 could be a potentially effective therapeutic intervention to prevent the pathological changes in the cerebellum caused by ingestion of MeHg-contaminated food.
Collapse
Affiliation(s)
- Misao Sakamoto
- Laboratory of Oral Aging Science, Faculty of Dental Sciences, Kyushu University, Maidashi 3-1-1, Fukuoka 812-8582, Japan
| | | | | | | |
Collapse
|
40
|
Mazerik JN, Hagele T, Sherwani S, Ciapala V, Butler S, Kuppusamy ML, Hunter M, Kuppusamy P, Marsh CB, Parinandi NL. Phospholipase A2 activation regulates cytotoxicity of methylmercury in vascular endothelial cells. Int J Toxicol 2008; 26:553-69. [PMID: 18066971 DOI: 10.1080/10915810701707759] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mercury has been identified as a risk factor for cardiovascular disease among humans. Through diet, mainly fish consumption, humans are exposed to methylmercury, the biomethylated organic form of environmental mercury. As the endothelium is an important player in homeostasis of the cardiovascular system, here, the authors tested their hypothesis that methylmercury activates the lipid signaling enzyme phospholipase A(2) (PLA(2)) in vascular endothelial cells (ECs), causing upstream regulation of cytotoxicity. To test this hypothesis, the authors used bovine pulmonary artery ECs (BPAECs) cultured in monolayers, following labeling of their membrane phospholipids with [(3)H]arachidonic acid (AA). The cells were exposed to methylmercury chloride (MMC) and then the release of free AA (index of PLA(2) activity) and lactate dehydrogenase (LDH; index of cytotoxicity) were determined by liquid scintillation counting and spectrophotometry, respectively. MMC significantly activated PLA(2) in a dose-dependent (5 to 15 microM) and time-dependent (0 to 60 min) fashion. Sulfhydryl (thiol-protective) agents, calcium chelators, antioxidants, and PLA(2)-specific inhibitors attenuated the MMC-induced PLA(2) activation, suggesting the role of thiols, reactive oxygen species (ROS), and calcium in the activation of PLA(2) in BPAECs. MMC also induced the loss of thiols and increase of lipid peroxidation in BPAECs. MMC induced cytotoxicity in BPAECs as observed by the altered cell morphology and LDH leak, which was significantly attenuated by PLA(2) inhibitors. This study established that PLA(2) activation through thiols, calcium, and oxidative stress was associated with the cytotoxicity of MMC in BPAECs, drawing attention to the involvement of PLA(2) signaling in the methylmercury-induced vascular endothelial dysfunctions.
Collapse
Affiliation(s)
- Jessica N Mazerik
- Lipid Signaling and Lipidomics Laboratory, Division of Pulmonary, Allergy, Critical Care, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kane CJM, Chang JY, Roberson PK, Garg TK, Han L. Ethanol exposure of neonatal rats does not increase biomarkers of oxidative stress in isolated cerebellar granule neurons. Alcohol 2008; 42:29-36. [PMID: 18249267 DOI: 10.1016/j.alcohol.2007.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 03/20/2007] [Accepted: 10/09/2007] [Indexed: 11/29/2022]
Abstract
Oxidative stress is a candidate mechanism for ethanol neuropathology in fetal alcohol spectrum disorders. Oxidative stress often involves production of reactive oxygen species (ROS), deterioration of the mitochondrial membrane potential (MMP), and cell death. Previous studies have produced conflicting results regarding the role of oxidative stress and the benefit of antioxidants in ethanol neuropathology in the developing brain. This study investigated the hypothesis that ethanol neurotoxicity involves production of ROS with negative downstream consequences for MMP and neuron survival. This was modeled in neonatal rats at postnatal day 4 (P4) and P14. It is well established that granule neurons in the rat cerebellar cortex are more vulnerable to ethanol neurotoxicity on P4 than at later ages. Thus, it was hypothesized that ethanol produces more oxidative stress and its negative consequences on P4 than on P14. A novel experimental approach was used in which ethanol was administered to animals in vivo (gavage 6g/kg), granule neurons were isolated 2-24h post-treatment, and ROS production and relative MMP were immediately assessed in the viable cells. Cells were also placed in culture and survival was measured 24h later. The results revealed that ethanol did not induce granule cells to produce ROS, cause deterioration of neuronal MMP, or cause neuron death when compared to vehicle controls. Further, granule neurons from neither P4 nor P14 animals mounted an oxidative response to ethanol. These findings do not support the hypothesis that oxidative stress is obligate to granule neuron death after ethanol exposure in the neonatal rat brain. Other investigators have reached a similar conclusion using either brain homogenates or cell cultures. In this context, it is likely that oxidative stress is not the sole and perhaps not the principal mechanism of ethanol neurotoxicity for cerebellar granule neurons during this stage of brain development.
Collapse
Affiliation(s)
- Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | | | | | | | | |
Collapse
|
42
|
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: 112] [Impact Index Per Article: 6.2] [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
|
43
|
Falluel-Morel A, Sokolowski K, Sisti HM, Zhou X, Shors TJ, DiCicco-Bloom E. Developmental mercury exposure elicits acute hippocampal cell death, reductions in neurogenesis, and severe learning deficits during puberty. J Neurochem 2007; 103:1968-81. [PMID: 17760861 PMCID: PMC3363963 DOI: 10.1111/j.1471-4159.2007.04882.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Normal brain development requires coordinated regulation of several processes including proliferation, differentiation, and cell death. Multiple factors from endogenous and exogenous sources interact to elicit positive as well as negative regulation of these processes. In particular, the perinatal rat brain is highly vulnerable to specific developmental insults that produce later cognitive abnormalities. We used this model to examine the developmental effects of an exogenous factor of great concern, methylmercury (MeHg). Seven-day-old rats received a single injection of MeHg (5 microg/gbw). MeHg inhibited DNA synthesis by 44% and reduced levels of cyclins D1, D3, and E at 24 h in the hippocampus, but not the cerebellum. Toxicity was associated acutely with caspase-dependent programmed cell death. MeHg exposure led to reductions in hippocampal size (21%) and cell numbers 2 weeks later, especially in the granule cell layer (16%) and hilus (50%) of the dentate gyrus defined stereologically, suggesting that neurons might be particularly vulnerable. Consistent with this, perinatal exposure led to profound deficits in juvenile hippocampal-dependent learning during training on a spatial navigation task. In aggregate, these studies indicate that exposure to one dose of MeHg during the perinatal period acutely induces apoptotic cell death, which results in later deficits in hippocampal structure and function.
Collapse
Affiliation(s)
- Anthony Falluel-Morel
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Katie Sokolowski
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Helene M. Sisti
- Department of Psychology, Rutgers University, Piscataway, New Jersey, USA
| | - Xiaofeng Zhou
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Tracey J. Shors
- Department of Psychology, Rutgers University, Piscataway, New Jersey, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Department of Pediatrics; Member of the Cancer Institute of New Jersey, New Jersey, USA
| |
Collapse
|
44
|
Hagele TJ, Mazerik JN, Gregory A, Kaufman B, Magalang U, Kuppusamy ML, Marsh CB, Kuppusamy P, Parinandi NL. Mercury activates vascular endothelial cell phospholipase D through thiols and oxidative stress. Int J Toxicol 2007; 26:57-69. [PMID: 17365148 DOI: 10.1080/10915810601120509] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Currently, mercury has been identified as a risk factor of cardiovascular diseases among humans. Here, the authors tested the hypothesis that mercury modulates the activity of the endothelial lipid signaling enzyme, phospholipase D (PLD), which is an important player in the endothelial cell (EC) barrier functions. Monolayers of bovine pulmonary artery ECs (BPAECs) in culture, following labeling of membrane phospholipids with [32P]orthophosphate, were exposed to mercuric chloride (inorganic form), methylmercury chloride (environmental form), and thimerosal (pharmaceutical form), and the formation of phosphatidylbutanol as an index of PLD activity was determined by thin-layer chromatography and liquid scintillation counting. All three forms of mercury significantly activated PLD in BPAECs in a dose-dependent (0 to 50 microM) and time-dependent (0 to 60 min) fashion. Metal chelators significantly attenuated mercury-induced PLD activation, suggesting that cellular mercury-ligand interaction(s) is required for the enzyme activation and that chelators are suitable blockers for mercury-induced PLD activation. Sulfhydryl (thiol-protective) agents and antioxidants also significantly attenuated the mercury-induced PLD activation in BPAECs. Enhanced reactive oxygen species generation, as an index of oxidative stress, was observed in BPAECs treated with methylmercury that was attenuated by antioxidants. All the three different forms of mercury significantly induced the decrease of levels of total cellular thiols. For the first time, this study revealed that mercury induced the activation of PLD in the vascular ECs wherein cellular thiols and oxidative stress acted as signal mediators for the enzyme activation. The results underscore the importance of PLD signaling in mercury-induced endothelial dysfunctions ultimately leading to cardiovascular diseases.
Collapse
Affiliation(s)
- Thomas J Hagele
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Lipidomics and Lipid Signaling Laboratory, The Ohio State University College of Medicine, Columbus, Ohio 43210, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chang JY. Methylmercury causes glial IL-6 release. Neurosci Lett 2007; 416:217-20. [PMID: 17368937 DOI: 10.1016/j.neulet.2007.01.076] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Revised: 12/26/2006] [Accepted: 01/08/2007] [Indexed: 11/25/2022]
Abstract
Methylmercury (MeHg) is an environmental toxin that causes severe neurological complications in humans and experimental animals. MeHg caused IL-6 release from the rat C6 glioma cells, the human U251HF glioma cells and the human retina pigment epithelial (ARPE-19) cells. These results plus those we reported earlier using mouse N9 microglia cells indicate that IL-6 induction may be a general property of MeHg among various glial cell types across species. MeHg caused a concentration-dependent increase of cellular oxidation with a maximal level reached by approximately 10 microM MeHg, which was similar to that caused by 30 microM H2O2 or t-butyl hydroperoxide (tBH). The ability of MeHg to induce IL-6 release was not affected by exogenously added H2O2 or t-butyl hydroperoxide. Furthermore, IL-6 release was not accompanied by other cytokine release. Given the reports by others that IL-6 could modulate neuronal survival, glia may affect MeHg neurotoxicity by their IL-6 release when exposed to this neurotoxin.
Collapse
Affiliation(s)
- Jason Y Chang
- Department of Neurobiology and Developmental Sciences, Slot 510, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA.
| |
Collapse
|
46
|
Burke K, Cheng Y, Li B, Petrov A, Joshi P, Berman R, Reuhl KR, DiCicco-Bloom E. Methylmercury elicits rapid inhibition of cell proliferation in the developing brain and decreases cell cycle regulator, cyclin E. Neurotoxicology 2006; 27:970-81. [PMID: 17056119 PMCID: PMC2013736 DOI: 10.1016/j.neuro.2006.09.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 08/21/2006] [Accepted: 09/08/2006] [Indexed: 01/12/2023]
Abstract
The developing brain is highly sensitive to methylmercury (MeHg). Still, the initial changes in cell proliferation that may contribute to long-term MeHg effects are largely undefined. Our previous studies with growth factors indicate that acute alterations of the G1/S-phase transition can permanently affect cell numbers and organ size. Therefore, we determined whether an environmental toxicant could also impact brain development with rapid (6-7h) effects on DNA synthesis and cell cycle machinery in neuronal precursors. In vivo studies in newborn rat hippocampus and cerebellum, two regions of postnatal neurogenesis, were followed by in vitro analysis of two precursor models, cortical and cerebellar cells, focusing on the proteins that regulate the G1/S transition. In postnatal day 7 (P7) pups, a single subcutaneous injection of MeHg (3microg/g) acutely (7h) decreased DNA synthesis in the hippocampus by 40% and produced long-term (2 weeks) reductions in total cell number, estimated by DNA quantification. Surprisingly, cerebellar granule cells were resistant to MeHg effects in vivo at comparable tissue concentrations, suggesting region-specific differences in precursor populations. In vitro, MeHg altered proliferation and cell viability, with DNA synthesis selectively inhibited at an early timepoint (6h) corresponding to our in vivo observations. Considering that G1/S regulators are targets of exogenous signals, we used a well-defined cortical cell model to examine MeHg effects on relevant cyclin-dependent kinases (CDK) and CDK inhibitors. At 6h, MeHg decreased by 75% levels of cyclin E, a cell cycle regulator with roles in proliferation and apoptosis, without altering p57, p27, or CDK2 nor levels of activated caspase 3. In aggregate, our observations identify the G1/S transition as an early target of MeHg toxicity and raise the possibility that cyclin E degradation contributes to both decreased proliferation and eventual cell death.
Collapse
Affiliation(s)
- Kelly Burke
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Yinghong Cheng
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Baogang Li
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Alex Petrov
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Pushkar Joshi
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Robert Berman
- Department of Neurological Surgery, University of California at Davis
| | | | - Emanuel DiCicco-Bloom
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
- Department of Pediatrics; Member of the Cancer Institute of New Jersey
| |
Collapse
|