1
|
Hu XF, Loan A, Chan HM. Re-thinking the link between exposure to mercury and blood pressure. Arch Toxicol 2025; 99:481-512. [PMID: 39804370 PMCID: PMC11775068 DOI: 10.1007/s00204-024-03919-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/25/2024] [Indexed: 01/30/2025]
Abstract
Hypertension or high blood pressure (BP) is a prevalent and manageable chronic condition which is a significant contributor to the total global disease burden. Environmental chemicals, including mercury (Hg), may contribute to hypertension onset and development. Hg is a global health concern, listed by the World Health Organization (WHO) as a top ten chemical of public health concern. Most people are exposed to some level of Hg, with vulnerable groups, including Indigenous peoples and small-scale gold miners, at a higher risk for exposure. We published a systematic review and meta-analysis in 2018 showing a dose-response relationship between Hg exposure and hypertension. This critical review summarizes the biological effects of Hg (both organic and inorganic form) on the underlying mechanisms that may facilitate the onset and development of hypertension and related health outcomes and updates the association between Hg exposure (total Hg concentrations in hair) and BP outcomes. We also evaluated the weight of evidence using the Bradford Hill criteria. There is a strong dose-response relationship between Hg (both organic and inorganic) exposure and BP in animal studies and convincing evidence that Hg contributes to hypertension by causing structural and functional changes, vascular reactivity, vasoconstriction, atherosclerosis, dyslipidemia, and thrombosis. The underlying mechanisms are vast and include impairments in antioxidant defense mechanisms, increased ROS production, endothelial dysfunction, and alteration of the renin-angiotensin system. We found additional 16 recent epidemiological studies that have reported the relationship between Hg exposure and hypertension in the last 5 years. Strong evidence from epidemiological studies shows a positive association between Hg exposure and the risk of hypertension and elevated BP. The association is mixed at lower exposure levels but suggests that Hg can affect BP even at low doses when co-exposed with other metals. Further research is needed to develop robust conversion factors among different biomarkers and standardized measures of Hg exposure. Regulatory agencies should consider adopting a 2 µg/g hair Hg level as a cut-off for public health regulation, especially for adults older than child-bearing age.
Collapse
Affiliation(s)
- Xue Feng Hu
- Chemical and Environmental Toxicology Program, Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, K1N 6N5, Canada
| | - Allison Loan
- Chemical and Environmental Toxicology Program, Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, K1N 6N5, Canada
| | - Hing Man Chan
- Chemical and Environmental Toxicology Program, Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
2
|
Nakamura R, Iwai T, Takanezawa Y, Shirahata T, Konishi N, Ohshiro Y, Uraguchi S, Tanabe M, Kobayashi Y, Sakamoto K, Nakahara T, Yamamoto M, Kiyono M. Oleanolic acid-3-glucoside, a synthetic oleanane-type saponin, ameliorates methylmercury-induced dysfunction of synaptic transmission in mice. Toxicology 2024; 506:153867. [PMID: 38906242 DOI: 10.1016/j.tox.2024.153867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Methylmercury (MeHg) is widely distributed in nature and is known to cause neurotoxic effects. This study aimed to examine the anti-MeHg activity of oleanolic acid-3-glucoside (OA3Glu), a synthetic oleanane-type saponin derivative, by evaluating its effects on motor function, pathology, and electrophysiological properties in a mouse model of MeHg poisoning. Mice were orally administered 2 or 4 mg·kg-1·d-1 MeHg with or without 100 µg·kg-1·d-1 OA3Glu 5x/week for four weeks. Motor function was evaluated using beam-walking and dynamic weight-bearing (DWB) tests. High-dose MeHg exposure significantly increased the frequency of stepping off the hind leg while crossing the beam in the beam-walking test, and increased weight on forelegs when moving freely in the DWB test. OA3Glu treatment alleviated motor abnormality caused by high-dose MeHg exposure in both motor function tests. Additionally, OA3Glu treatment reduced the number of contracted Purkinje cells frequently observed in the cerebellum of MeHg-treated groups, although cerebrum histology was similar in all experimental groups. The synaptic potential amplitude in the cerebellum decreased as MeHg exposure increased, which was restored by OA3Glu treatment. Even in the cerebrum, where the effects of MeHg were not observed, the amplitude of the field potential was suppressed with increasing MeHg exposure but was restored with OA3Glu treatment. Taken together, the study findings suggest that OA3Glu improves neurotransmission and movement disorders associated with MeHg exposure via protection of Purkinje cells in the cerebellum while ameliorating pre/post-synaptic deficits in the cerebral cortex in which no changes were observed at the tissue level, potentially providing a treatment to mitigate MeHg toxicity.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Takashi Iwai
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yasukazu Takanezawa
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tatsuya Shirahata
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Naruki Konishi
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yuka Ohshiro
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shimpei Uraguchi
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Mitsuo Tanabe
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshinori Kobayashi
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kenji Sakamoto
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan; Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Tsutomu Nakahara
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Megumi Yamamoto
- Department of Environment and Public Health, National Institute for Minamata Disease, 4058-18, Hama, Minamata, Kumamoto 867-0008, Japan
| | - Masako Kiyono
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
3
|
He F, Liu R. Mechanistic insights into phenanthrene-triggered oxidative stress-associated neurotoxicity, genotoxicity, and behavioral disturbances toward the brandling worm (Eisenia fetida) brain: The need for an ecotoxicological evaluation. JOURNAL OF HAZARDOUS MATERIALS 2023; 450:131072. [PMID: 36857826 DOI: 10.1016/j.jhazmat.2023.131072] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
In this study, earthworm (Eisenia fetida) brain was chosen as targeted receptors to probe the mechanisms of oxidative stress-related neurotoxicity, genotoxicity, and behavioral disturbances triggered by PHE. Results showed that PHE stress can initiate significant amounts of ROS, thus triggering oxidative stress in E. fetida brain. These effects were accompanied by a significant increase of damage to macromolecules DNA and lipids, resulting in severe oxidative effects. PHE exposure can induce AChE inhibition by ROS-induced injury and the accumulation of excess ACh at the nicotinic post-synaptic membrane, thus inducing aggravated neurological dysfunction and neurotoxicity of E. fetida through an oxidative stress pathway. Moreover, the burrowing behavior of earthworms was disturbed by oxidative stress-induced neurotoxicity after exposure to PHE. Furthermore, the abnormal mRNA expression profiles of oxidative stress- and neurotoxicity-related genes in worm brain were induced by PHE stress. The IBR results suggested that E. fetida brain was suffered more serious damage caused by PHE under higher doses and long-term exposure. Taken together, PHE exposure can trigger oxidative stress-mediated neurotoxicity and genotoxicity in worm brain and behavioral disorder through ROS-induced damage. This study is of great significance to evaluate the harmful effects of PHE and its mechanisms on soil ecological health.
Collapse
Affiliation(s)
- Falin He
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China
| | - Rutao Liu
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, Shandong Province, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China.
| |
Collapse
|
4
|
He F, Liu R, Tian G, Qi Y, Wang T. Ecotoxicological evaluation of oxidative stress-mediated neurotoxic effects, genetic toxicity, behavioral disorders, and the corresponding mechanisms induced by fluorene-contaminated soil targeted to earthworm (Eisenia fetida) brain. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 871:162014. [PMID: 36740067 DOI: 10.1016/j.scitotenv.2023.162014] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
Fluorene is a commonly identified PAH pollutant in soil and exhibits various worrisome hazardous effects to soil organisms. Currently, the toxicity profiles of fluorene on earthworm brain are rare, and the mechanisms and their corresponding pathways involved in fluorene-triggered neurotoxicity, genotoxicity, and behavior changes have not been reported hitherto. Herein, earthworm (Eisenia fetida) brain was chosen as targeted receptor to explore the neurotoxic effects, genetic toxicity, behavioral disorders, and related mechanisms caused by fluorene-induced oxidative stress pathways. The results showed excess fluorene initiated the release of excessive quantities of ROS in earthworm brain, which have caused oxidative stress and accompanied by serious oxidative effects, including LPO (lipid peroxidation) and DNA injury. To minimize the damage effects, the antioxidant defense mechanisms (antioxidant enzymes and non-enzymatic antioxidants) were activated, and entailed a decrease of the antioxidant capacity in E. fetida brain, which, in turn, causes further ROS-induced ROS release. Exposure of fluorene induced the abnormal mRNA expression of genes relevant to oxidative stress (e.g., GST, SOD, CAT, GPx, MT, and Hsp70) and neurotoxicity (e.g., H02, C04, D06, and E08) in E. fetida brain. Specifically, fluorene can bind directly to AChE, destroying the conformation of this protein, and even affecting its physiological functions. This occurrence caused the inhibition of AChE activity and excess ACh accumulation at the nicotinic post-synaptic membrane, finally triggering neurotoxicity by activation of pathways related to oxidative stress. Moreover, the avoidance responses and burrowing behavior were obviously disturbed by oxidative stress-induced neurotoxicity after exposure to fluorene. The results form IBR suggested more severe poisoning effects to E. fetida brain initiated by high-dose and long-term exposure of fluorene. Among, oxidative stress injury and genotoxic potential are more sensitive endpoint than others. Collectively, fluorene stress can provoke potential neurotoxicity, genotoxicity, and behavioral disturbances targeted to E. fetida brain through the ROS-mediated pathways involving oxidative stress. These findings are of great significance to estimate the detrimental effects of fluorene and the corresponding mechanisms on soil eco-safety.
Collapse
Affiliation(s)
- Falin He
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China
| | - Rutao Liu
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China.
| | - Guang Tian
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China
| | - Yuntao Qi
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China
| | - Tingting Wang
- School of Environmental Science and Engineering, Shandong University, China-America CRC for Environment & Health, 72# Jimo Binhai Road, Qingdao, Shandong 266237, PR China
| |
Collapse
|
5
|
Bose R, Spulber S, Ceccatelli S. The Threat Posed by Environmental Contaminants on Neurodevelopment: What Can We Learn from Neural Stem Cells? Int J Mol Sci 2023; 24:4338. [PMID: 36901772 PMCID: PMC10002364 DOI: 10.3390/ijms24054338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Exposure to chemicals may pose a greater risk to vulnerable groups, including pregnant women, fetuses, and children, that may lead to diseases linked to the toxicants' target organs. Among chemical contaminants, methylmercury (MeHg), present in aquatic food, is one of the most harmful to the developing nervous system depending on time and level of exposure. Moreover, certain man-made PFAS, such as PFOS and PFOA, used in commercial and industrial products including liquid repellants for paper, packaging, textile, leather, and carpets, are developmental neurotoxicants. There is vast knowledge about the detrimental neurotoxic effects induced by high levels of exposure to these chemicals. Less is known about the consequences that low-level exposures may have on neurodevelopment, although an increasing number of studies link neurotoxic chemical exposures to neurodevelopmental disorders. Still, the mechanisms of toxicity are not identified. Here we review in vitro mechanistic studies using neural stem cells (NSCs) from rodents and humans to dissect the cellular and molecular processes changed by exposure to environmentally relevant levels of MeHg or PFOS/PFOA. All studies show that even low concentrations dysregulate critical neurodevelopmental steps supporting the idea that neurotoxic chemicals may play a role in the onset of neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
6
|
Azevedo LF, Karpova N, Rocha BA, Barbosa Junior F, Gobe GC, Hornos Carneiro MF. Evidence on Neurotoxicity after Intrauterine and Childhood Exposure to Organomercurials. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1070. [PMID: 36673825 PMCID: PMC9858833 DOI: 10.3390/ijerph20021070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
Although the molecular mechanisms underlying methylmercury toxicity are not entirely understood, the observed neurotoxicity in early-life is attributed to the covalent binding of methylmercury to sulfhydryl (thiol) groups of proteins and other molecules being able to affect protein post-translational modifications from numerous molecular pathways, such as glutamate signaling, heat-shock chaperones and the antioxidant glutaredoxin/glutathione system. However, for other organomercurials such as ethylmercury or thimerosal, there is not much information available. Therefore, this review critically discusses current knowledge about organomercurials neurotoxicity-both methylmercury and ethylmercury-following intrauterine and childhood exposure, as well as the prospects and future needs for research in this area. Contrasting with the amount of epidemiological evidence available for methylmercury, there are only a few in vivo studies reporting neurotoxic outcomes and mechanisms of toxicity for ethylmercury or thimerosal. There is also a lack of studies on mechanistic approaches to better investigate the pathways involved in the potential neurotoxicity caused by both organomercurials. More impactful follow-up studies, especially following intrauterine and childhood exposure to ethylmercury, are necessary. Childhood vaccination is critically important for controlling infectious diseases; however, the safety of mercury-containing thimerosal and, notably, its effectiveness as preservative in vaccines are still under debate regarding its potential dose-response effects to the central nervous system.
Collapse
Affiliation(s)
- Lara Ferreira Azevedo
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Nina Karpova
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Bruno Alves Rocha
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Fernando Barbosa Junior
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Glenda Carolyn Gobe
- Kidney Disease Research Group, School of Medicine, Translational Research Institute, University of Queensland, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Maria Fernanda Hornos Carneiro
- Department of Pharmacy, Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
7
|
Alterations in UPR Signaling by Methylmercury Trigger Neuronal Cell Death in the Mouse Brain. Int J Mol Sci 2022; 23:ijms232315412. [PMID: 36499738 PMCID: PMC9738736 DOI: 10.3390/ijms232315412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures specific areas of the brain. MeHg is known to induce oxidative and endoplasmic reticulum (ER) stress. The unfolded protein response (UPR) pathway has a dual nature in that it regulates and protects cells from an overload of improperly folded proteins in the ER, whereas excessively stressed cells are eliminated by apoptosis. Oxidative stress/ER stress induced by methylmercury exposure may tilt the UPR toward apoptosis, but there is little in vivo evidence of a direct link to actual neuronal cell death. Here, by using the ER stress-activated indicator (ERAI) system, we investigated the time course signaling alterations of UPR in vivo in the most affected areas, the somatosensory cortex and striatum. In the ERAI-Venus transgenic mice exposed to MeHg (30 or 50 ppm in drinking water), the ERAI signal, which indicates the activation of the cytoprotective pathway of the UPR, was only transiently enhanced, whereas the apoptotic pathway of the UPR was persistently enhanced. Furthermore, detailed analysis following the time course showed that MeHg-induced apoptosis is strongly associated with alterations in UPR signaling. Our results suggest that UPR modulation could be a therapeutic target for treating neuropathy.
Collapse
|
8
|
Ferroptosis as a mechanism of non-ferrous metal toxicity. Arch Toxicol 2022; 96:2391-2417. [PMID: 35727353 DOI: 10.1007/s00204-022-03317-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Ferroptosis is a recently discovered form of regulated cell death, implicated in multiple pathologies. Given that the toxicity elicited by some metals is linked to alterations in iron metabolism and induction of oxidative stress and lipid peroxidation, ferroptosis might be involved in such toxicity. Although direct evidence is insufficient, certain pioneering studies have demonstrated a crosstalk between metal toxicity and ferroptosis. Specifically, the mechanisms underlying metal-induced ferroptosis include induction of ferritinophagy, increased DMT-1 and TfR cellular iron uptake, mitochondrial dysfunction and mitochondrial reactive oxygen species (mitoROS) generation, inhibition of Xc-system and glutathione peroxidase 4 (GPX4) activity, altogether resulting in oxidative stress and lipid peroxidation. In addition, there is direct evidence of the role of ferroptosis in the toxicity of arsenic, cadmium, zinc, manganese, copper, and aluminum exposure. In contrast, findings on the impact of cobalt and nickel on ferroptosis are scant and nearly lacking altogether for mercury and especially lead. Other gaps in the field include limited studies on the role of metal speciation in ferroptosis and the critical cellular targets. Although further detailed studies are required, it seems reasonable to propose even at this early stage that ferroptosis may play a significant role in metal toxicity, and its modulation may be considered as a potential therapeutic tool for the amelioration of metal toxicity.
Collapse
|
9
|
Amaral AU, Wajner M. Pathophysiology of maple syrup urine disease: Focus on the neurotoxic role of the accumulated branched-chain amino acids and branched-chain α-keto acids. Neurochem Int 2022; 157:105360. [DOI: 10.1016/j.neuint.2022.105360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022]
|
10
|
Wang L, Yu L, Ge H, Bu Y, Sun M, Huang D, Wang S. A novel reversible dual-mode probe based on amorphous carbon nanodots for the detection of mercury ion and glutathione. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
11
|
Yang L, Guo H, Kuang Y, Yang H, Zhang X, Tang R, Li D, Li L. Neurotoxicity induced by combined exposure of microcystin-LR and nitrite in male zebrafish (Danio rerio): Effects of oxidant-antioxidant system and neurotransmitter system. Comp Biochem Physiol C Toxicol Pharmacol 2022; 253:109248. [PMID: 34826614 DOI: 10.1016/j.cbpc.2021.109248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 11/15/2022]
Abstract
With the intensification of water eutrophication around the world, cyanobacterial blooms have been becoming a common environmental pollution problem. The levels of microcystin-LR (MC-LR) and nitrite rise sharply during the cyanobacterial bloom period, which may have potential joint toxicity on aquatic organisms. In this study, adult male zebrafish were immersed into different joint solutions of MC-LR (0, 3, 30 μg/L) and nitrite (0, 2, 20 mg/L) for 30 days to explore the neurotoxic effects and underlying mechanisms. The results showed that single factor MC-LR or nitrite caused a concentration-dependent damage in brain ultrastructure and the effects of their joint exposure were much more intense. Downregulated expression of mbp and bdnf associated with myelination of nerve fibers further confirmed that MC-LR and nitrite could damage the structure and function of neuron. The decreases in dopamine content, acetylcholinesterase activity and related gene mRNA levels indicated that MC-LR and nitrite adversely affected the normal function of the dopaminergic and cholinergic systems in zebrafish brain. In addition, the significant increase in malondialdehyde content suggested the occurrence of oxidative stress caused by MC-LR, nitrite and their joint-exposure, which paralleled a significant decrease in antioxidant enzyme‑manganese superoxide dismutase activity and its transcription level. In conclusion, MC-LR + Nitrite joint-exposure has synergistic neurotoxic effects on the structure and neurotransmitter systems of fish brain, and antioxidant capacity disruption caused by these two factors might be one of the underlying synergistic mechanisms. Therefore, there is a risk of being induced neurotoxicity in fish during sustained cyanobacterial bloom events.
Collapse
Affiliation(s)
- Liping Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Honghui Guo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yu Kuang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Hui Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xi Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China; Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, PR China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, PR China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, PR China
| | - Rong Tang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China; Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, PR China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, PR China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, PR China
| | - Dapeng Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China; Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, PR China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, PR China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, PR China
| | - Li Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China; Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, PR China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, PR China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, PR China.
| |
Collapse
|
12
|
A Novel Diselenide-Probucol-Analogue Protects Against Methylmercury-Induced Toxicity in HT22 Cells by Upregulating Peroxide Detoxification Systems: a Comparison with Diphenyl Diselenide. Neurotox Res 2022; 40:127-139. [PMID: 35043379 DOI: 10.1007/s12640-021-00466-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/24/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental neurotoxicant whose mechanisms of action involve oxidation of endogenous nucleophilic groups (mainly thiols and selenols), depletion of antioxidant defenses, and disruption of neurotransmitter homeostasis. Diphenyl diselenide-(PhSe)2-a model diaryl diselenide, has been reported to display significant protective effects against MeHg-induced neurotoxicity under both in vitro and in vivo experimental conditions. In this study, we compared the protective effects of (PhSe)2 with those of RC513 (4,4'-diselanediylbis(2,6-di-tert-butylphenol), a novel diselenide-probucol-analog) against MeHg-induced toxicity in the neuronal (hippocampal) cell line HT22. Although both (PhSe)2 and RC513 significantly mitigated MeHg- and tert-butylhydroperoxide (t-BuOOH)-cytotoxicity, the probucol analog exhibited superior protective effects, which were observed earlier and at lower concentrations compared to (PhSe)2. RC513 treatment (at either 0.5 µM or 2 µM) significantly increased glutathione peroxidase (GPx) activity, which has been reported to counteract MeHg-toxicity. (PhSe)2 was also able to increase GPx activity, but only at 2 µM. Although both compounds increased the Gpx1 transcripts at 6 h after treatments, only RC513 was able to increase mRNA levels of Prx2, Prx3, Prx5, and Txn2, which are also involved in peroxide detoxification. RC513 (at 2 µM) significantly increased GPx-1 protein expression in HT22 cells, although (PhSe)2 displayed a minor (nonsignificant) effect in this parameter. In agreement, RC513 induced a faster and superior capability to cope with exogenously-added peroxide (t-BuOOH). In summary, when compared to the prototypical organic diaryl diselenide [(PhSe)2], RC513 displayed superior protective properties against MeHg-toxicity in vitro; this was paralleled by a more pronounced upregulation of defenses related to detoxification of peroxides, which are well-known MeHg-derived intermediate oxidant species.
Collapse
|
13
|
Erdemli-Köse SB, Yirün A, Balci-Özyurt A, Erkekoğlu P. Modification of the toxic effects of methylmercury and thimerosal by testosterone and estradiol in SH-SY5Y neuroblastoma cell line. J Appl Toxicol 2021; 42:981-994. [PMID: 34874569 DOI: 10.1002/jat.4269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 11/09/2022]
Abstract
Short-chained alkyl mercury compounds accumulate in particularly in the brain. Exposure to these compounds is associated with various neurotoxic effects. Gender-based differences are observed in neurodevelopmental disorders, and testosterone and estradiol may alter the toxic effect of the compounds. The present study aimed to investigate the toxic effects of methylmercury and thimerosal on SH-SY5Y cells in high testosterone/low estradiol and high estradiol/low testosterone containing cellular environment and estimate whether male and female brains react differently to the toxic effects of methylmercury and thimerosal. Study groups (n = 3) were designed as control: growth medium, thimerosal (T): 1.15-μM thimerosal, methylmercury (M): 2.93-μM methylmercury, high testosterone/low estradiol + thimerosal (TT): 1-μM testosterone + 0.75-μM estradiol + 1.15-μM thimerosal, high estradiol/low testosterone + thimerosal (ET): 0.1-μM testosterone + 7.5-μM estradiol + 1.15-μM thimerosal, high testosterone/low estradiol + methylmercury (TM): 1-μM testosterone + 0.75-μM estradiol + 2.93-μM methylmercury and high estradiol/low testosterone + methylmercury (EM): 0.1-μM testosterone + 7.5-μM estradiol + 2.93-μM methylmercury. While a significant decrease in glutathione levels was observed in M group, it was not seen in EM group. A significant increase in the protein carbonyl levels was detected in T group. A similar increase was observed in the TM and TT groups in which testosterone was dominant. It was determined that methylmercury, but not thimerosal, caused significant DNA damage and in TT group. The results showed that both thimerosal and methylmercury are toxic on SH-SY5Y cells and toxic effects of methylmercury are more severe than thimerosal. It has been determined that testosterone and estradiol alter the toxic effects of thimerosal and methylmercury.
Collapse
Affiliation(s)
- Selinay Başak Erdemli-Köse
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey.,Faculty of Arts and Sciences, Department of Chemistry, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Anıl Yirün
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey.,Department of Pharmaceutical Toxicology, Çukurova University Faculty of Pharmacy, Adana, Turkey
| | - Aylin Balci-Özyurt
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Pınar Erkekoğlu
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey.,Department of Vaccine Technology, Hacettepe University Vaccine Institute, Ankara, Turkey
| |
Collapse
|
14
|
Wei Y, Ni L, Pan J, Li X, Xu B, Deng Y, Yang T, Liu W. The Roles of Oxidative Stress in Regulating Autophagy in Methylmercury-induced Neurotoxicity. Neuroscience 2021; 469:175-190. [PMID: 34174372 DOI: 10.1016/j.neuroscience.2021.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022]
Abstract
Methylmercury (MeHg) is a potential neurotoxin that is highly toxic to the human central nervous system. Although MeHg neurotoxicity has been widely studied, the mechanism of MeHg neurotoxicity has not yet been fully elucidated. Some research evidence suggests that oxidative stress and autophagy are important molecular mechanisms of MeHg-induced neurotoxicity. Researchers have widely accepted that oxidative stress regulates the autophagy pathway. The current study reviews the activation of Nuclear factor-erythroid-2-related factor (Nrf2)-related oxidative stress pathways and autophagy signaling pathways in the case of MeHg neurotoxicity. In addition, autophagy mainly plays a role in the neurotoxicity of MeHg through mTOR-dependent and mTOR-independent autophagy signaling pathways. Finally, the regulation of autophagy by reactive oxygen species (ROS) and Nrf2 in MeHg neurotoxicity was explored in this review, providing a new concept for the study of the neurotoxicity mechanism of MeHg.
Collapse
Affiliation(s)
- Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China.
| |
Collapse
|
15
|
Nogara PA, Madabeni A, Bortoli M, Teixeira Rocha JB, Orian L. Methylmercury Can Facilitate the Formation of Dehydroalanine in Selenoenzymes: Insight from DFT Molecular Modeling. Chem Res Toxicol 2021; 34:1655-1663. [PMID: 34077192 DOI: 10.1021/acs.chemrestox.1c00073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Experimental studies have indicated that electrophilic mercury forms (e.g., methylmercury, MeHg+) can accelerate the breakage of selenocysteine in vitro. Particularly, in 2009, Khan et al. (Environ. Toxicol. Chem. 2009, 28, 1567-1577) proposed a mechanism for the degradation of a free methylmercury selenocysteinate complex that was theoretically supported by Asaduzzaman et al. (Inorg. Chem. 2010, 50, 2366-2372). However, little is known about the fate of methylmercury selenocysteinate complexes embedded in an enzyme, especially in conditions of oxidative stress in which methylmercury target enzymes operate. Here, an accurate computational study on molecular models (level of theory: COSMO-ZORA-BLYP-D3(BJ)/TZ2P) was carried out to investigate the formation of dehydroalanine (Dha) in selenoenzymes, which irreversibly impairs their function. Methylselenocysteine as well as methylcysteine and methyltellurocysteine were included to gain insight on the peculiar behavior of selenium. Dha forms in a two-step process, i.e., the oxidation of the chalcogen nucleus followed by a syn-elimination leading to the alkene and the chalcogenic acid. The effect of an excess of hydrogen peroxide, which may lead to the formation of chalcogenones before the elimination, and of MeHg+, a severe toxicant targeting selenoproteins, which leads to the formation of methylmercury selenocysteinate, are also studied with the aim of assessing whether these pathological conditions facilitate the formation of Dha. Indeed, elimination occurs after chalcogen oxidation and MeHg+ facilitates the process. These results indicate a possible mechanism of toxicity of MeHg+ in selenoproteins.
Collapse
Affiliation(s)
- Pablo Andrei Nogara
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil.,Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Andrea Madabeni
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Marco Bortoli
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - João Batista Teixeira Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
16
|
Branco V, Aschner M, Carvalho C. Neurotoxicity of mercury: an old issue with contemporary significance. ADVANCES IN NEUROTOXICOLOGY 2021; 5:239-262. [PMID: 34263092 PMCID: PMC8276940 DOI: 10.1016/bs.ant.2021.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mercury exerts a variety of toxic effects, depending on the specific compound and route of exposure. However, neurotoxicity in virtue of its consequence to health causes the greatest concern for toxicologists. This is particularly true regarding fetal development, where neurotoxic effects are much more severe than in adults, and the toxicity threshold is lower. Here, we review the major concepts regarding the neurotoxicity of mercury compounds (mercury vapor; methylmercury and ethylmercury), from exposure routes to toxicokinetic particularities leading to brain deposition and the development of neurotoxic effects. Albeit research on the neurotoxicity of mercury compounds has significantly advanced from the second half of the twentieth century onwards, several grey areas regarding the mechanism of toxicity still exist. Thus, we emphasize research advances during the last two decades concerning the molecular interactions of mercury which cause neurotoxic effects. Highlights include the disruption of glutamate signaling and excitotoxicity resulting from exposure to mercury and the interaction with redox active residues such as cysteines and selenocysteines which are the premise accounting for the disruption of redox homeostasis caused by mercurials. We also address how immunotoxic effects at the CNS, namely microglia and astrocyte activation modulate developmental neurotoxicity, a major topic in contemporary research.
Collapse
Affiliation(s)
- Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, USA
| | - Cristina Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
17
|
Hiraoka H, Nomura R, Takasugi N, Akai R, Iwawaki T, Kumagai Y, Fujimura M, Uehara T. Spatiotemporal analysis of the UPR transition induced by methylmercury in the mouse brain. Arch Toxicol 2021; 95:1241-1250. [PMID: 33454823 DOI: 10.1007/s00204-021-02982-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/06/2021] [Indexed: 01/11/2023]
Abstract
Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures a specific area of the brain. MeHg-mediated neurotoxicity is believed to be caused by oxidative stress and endoplasmic reticulum (ER) stress but the mechanism by which those stresses lead to neuronal loss is unclear. Here, by utilizing the ER stress-activated indicator (ERAI) system, we investigated the signaling alterations in the unfolded protein response (UPR) prior to neuronal apoptosis in the mouse brain. In ERAI transgenic mice exposed to MeHg (25 mg/kg, S.C.), the ERAI signal, which indicates activation of the cytoprotective pathway of the UPR, was detected in the brain. Interestingly, detailed ex vivo analysis showed that the ERAI signal was localized predominantly in neurons. Time course analysis of MeHg exposure (30 ppm in drinking water) showed that whereas the ERAI signal was gradually attenuated at the late phase after increasing at the early phase, activation of the apoptotic pathway of the UPR was enhanced in proportion to the exposure time. These results suggest that MeHg induces not only ER stress but also neuronal cell death via a UPR shift. UPR modulation could be a therapeutic target for treating neuropathy caused by electrophiles similar to MeHg.
Collapse
Affiliation(s)
- Hideki Hiraoka
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Ryosuke Nomura
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Ryoko Akai
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Masatake Fujimura
- Department of Basic Medical Science, National Institute for Minamata Disease, Kumamoto, 867-0008, Japan
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan.
| |
Collapse
|
18
|
Kasamatsu S. Persulfide-Dependent Regulation of Electrophilic Redox Signaling in Neural Cells. Antioxid Redox Signal 2020; 33:1320-1331. [PMID: 32536194 DOI: 10.1089/ars.2020.8130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Redox homeostasis is precisely modulated by intricate systems that regulate production, elimination, and metabolism of electrophilic substances (electrophiles) in the nervous system. Since the first report of the endogenous production of reactive persulfide species in cells, such as cysteine persulfides (CysSSH), these reactive species have been a topic of extreme interest in the field of redox biology; persulfides/polysulfides possess unique chemical properties and are involved in multiple cellular functions. Recent Advances: Electrophilic signaling is mainly regulated by endogenous electrophiles that are generated from reactive oxygen species, nitric oxide, and their derivatives during stress responses, as well as by exogenous electrophiles, including compounds in foods and environmental pollutants, such as methylmercury (MeHg). Among diverse electrophiles that are endogenously generated, 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) possesses unique redox properties, of which the biosynthetic pathway, signaling mechanism, and metabolism in cells have been elucidated. Critical Issues: Persulfides, such as CysSSH, that are endogenously produced are critically involved in 8-nitro-cGMP metabolism. Exposure of neurons to the exogenous neurotoxicant, MeHg, causes severe neurodegeneration via disruption of persulfide-dependent 8-nitro-cGMP metabolism. Future Directions: Accumulating evidence indicates that persulfides are involved in various cellular functions under physiological and pathological conditions. These new aspects of redox biology related to persulfides may be frontiers of cell research, medical and clinical investigations of neurodegenerative diseases, as well as other fields. 8-Nitro-cGMP-mediated signaling and its persulfide-dependent metabolism in cells could, therefore, be potential targets for drug development, which may lead to the discovery of new therapeutic agents for many diseases, including neurodegenerative diseases.
Collapse
Affiliation(s)
- Shingo Kasamatsu
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka, Japan
| |
Collapse
|
19
|
Zieniewska I, Maciejczyk M, Zalewska A. The Effect of Selected Dental Materials Used in Conservative Dentistry, Endodontics, Surgery, and Orthodontics as Well as during the Periodontal Treatment on the Redox Balance in the Oral Cavity. Int J Mol Sci 2020; 21:ijms21249684. [PMID: 33353105 PMCID: PMC7767252 DOI: 10.3390/ijms21249684] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/31/2022] Open
Abstract
Oxidative stress (OS) is a redox homeostasis disorder that results in oxidation of cell components and thus disturbs cell metabolism. OS is induced by numerous internal as well as external factors. According to recent studies, dental treatment may also be one of them. The aim of our work was to assess the effect of dental treatment on the redox balance of the oral cavity. We reviewed literature available in PubMed, Medline, and Scopus databases, including the results from 2010 to 2020. Publications were searched according to the keywords: oxidative stress and dental monomers; oxidative stress and amalgam; oxidative stress and periodontitis, oxidative stress and braces, oxidative stress and titanium; oxidative stress and dental implants, oxidative stress and endodontics treatment, oxidative stress and dental treatment; and oxidative stress and dental composite. It was found that dental treatment with the use of composites, amalgams, glass-ionomers, materials for root canal filling/rinsing, orthodontic braces (made of various metal alloys), titanium implants, or whitening agents can disturb oral redox homeostasis by affecting the antioxidant barrier and increasing oxidative damage to salivary proteins, lipids, and DNA. Abnormal saliva secretion/composition was also observed in dental patients in the course of OS. It is suggested that the addition of antioxidants to dental materials or antioxidant therapy applied during dental treatment could protect the patient against harmful effects of OS in the oral cavity.
Collapse
Affiliation(s)
- Izabela Zieniewska
- Doctoral Studies, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-274 Bialystok, Poland
- Correspondence: (I.Z.); (A.Z.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 15-022 Bialystok, Poland;
| | - Anna Zalewska
- Experimental Dentistry Laboratory, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-274 Bialystok, Poland
- Correspondence: (I.Z.); (A.Z.)
| |
Collapse
|
20
|
Wajner M, Vargas CR, Amaral AU. Disruption of mitochondrial functions and oxidative stress contribute to neurologic dysfunction in organic acidurias. Arch Biochem Biophys 2020; 696:108646. [PMID: 33098870 DOI: 10.1016/j.abb.2020.108646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/08/2023]
Abstract
Organic acidurias (OADs) are inherited disorders of amino acid metabolism biochemically characterized by accumulation of short-chain carboxylic acids in tissues and biological fluids of the affected patients and clinically by predominant neurological manifestations. Some of these disorders are amenable to treatment, which significantly decreases mortality and morbidity, but it is still ineffective to prevent long-term neurologic and systemic complications. Although pathogenesis of OADs is still poorly established, recent human and animal data, such as lactic acidosis, mitochondrial morphological alterations, decreased activities of respiratory chain complexes and altered parameters of oxidative stress, found in tissues from patients and from genetic mice models with these diseases indicate that disruption of critical mitochondrial functions and oxidative stress play an important role in their pathophysiology. Furthermore, organic acids that accumulate in the most prevalent OADs were shown to compromise bioenergetics, by decreasing ATP synthesis, mitochondrial membrane potential, reducing equivalent content and calcium retention capacity, besides inducing mitochondrial swelling, reactive oxygen and nitrogen species generation and apoptosis. It is therefore presumed that secondary mitochondrial dysfunction and oxidative stress caused by major metabolites accumulating in OADs contribute to tissue damage in these pathologies.
Collapse
Affiliation(s)
- Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Ciências Biológicas, Universidade Regional Integrada do Alto Uruguai e das Missões, Erechim, RS, Brazil
| |
Collapse
|
21
|
Ferrer B, Prince LM, Tinkov AA, Santamaria A, Bowman AB, Aschner M. Chronic exposure to methylmercury disrupts ghrelin actions in C57BL/6J mice. Food Chem Toxicol 2020; 147:111918. [PMID: 33301842 DOI: 10.1016/j.fct.2020.111918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/06/2023]
Abstract
Methylmercury (MeHg) is a neurotoxic pollutant widely present in the environment. Initial symptoms of MeHg may include loss of body weight. However, the mechanisms by which MeHg induces body weight changes have yet to be fully elucidated. Body weight is regulated by multiple mechanisms. Whereas multiple peripheral peptides lead to food intake cessation, ghrelin is the only recognized peripheral hormone that stimulates food intake. It exerts its action on Neuropeptide Y/Agouti-related peptide neurons in the hypothalamus. To test if MeHg affects ghrelin signaling C57BL/6J mice (males and females) were exposed to 5 ppm MeHg via drinking water during a month. On days 15 and 30 of MeHg exposure ghrelin was administered intraperitoneally and changes in body weight and food intake were recorded. In addition, changes in ghrelin-induced signaling pathways in hypothalamus were also analyzed. Here, we show that in males, MeHg enhanced ghrelin-induced body weight gain by activating the AMP-activated Kinase (AMPK)/Uncoupled protein 2 (UCP2) signaling pathway. In contrast, in females, MeHg inhibited ghrelin-induced mTOR signaling activation and decreased Npy mRNA expression, thus mitigating the ghrelin-induced weight gain. Combined, our novel results demonstrate, for the first time, that MeHg disrupts the physiological functions of ghrelin differently in males and females.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia.
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico.
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Michael Aschner
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA; IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia.
| |
Collapse
|
22
|
Fujimura M, Usuki F. Methylmercury-Mediated Oxidative Stress and Activation of the Cellular Protective System. Antioxidants (Basel) 2020; 9:antiox9101004. [PMID: 33081221 PMCID: PMC7602710 DOI: 10.3390/antiox9101004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Methylmercury (MeHg) is a well-known neurotoxicant that causes severe intoxication in humans. In Japan, it is referred to as Minamata disease, which involves two characteristic clinical forms: fetal type and adult type depending on the exposed age. In addition to MeHg burden level, individual susceptibility to MeHg plays a role in the manifestation of MeHg toxicity. Research progress has pointed out the importance of oxidative stress in the pathogenesis of MeHg toxicity. MeHg has a high affinity for selenohydryl groups, sulfhydryl groups, and selenides. It has been clarified that such affinity characteristics cause the impairment of antioxidant enzymes and proteins, resulting in the disruption of antioxidant systems. Furthermore, MeHg-induced intracellular selenium deficiency due to the greater affinity of MeHg for selenohydryl groups and selenides leads to failure in the recoding of a UGA codon for selenocysteine and results in the degradation of antioxidant selenoenzyme mRNA by nonsense-mediated mRNA decay. The defect of antioxidant selenoenzyme replenishment exacerbates MeHg-mediated oxidative stress. On the other hand, it has also been revealed that MeHg can directly activate the antioxidant Keap1/Nrf2 signaling pathway. This review summarizes the incidence of MeHg-mediated oxidative stress from the viewpoint of the individual intracellular redox system interactions and the MeHg-mediated aforementioned intracellular events. In addition, the mechanisms of cellular stress pathways and neuronal cell death triggered by MeHg-mediated oxidative stress and direct interactions of MeHg with reactive residues of proteins are mentioned.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto 867-0008, Japan;
| | - Fusako Usuki
- Division of Neuroimmunology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan
- Correspondence: ; Tel.: +81-99-275-6246; Fax: +81-99-275-5942
| |
Collapse
|
23
|
Liu T, Gao Q, Yang B, Yin C, Chang J, Qian H, Xing G, Wang S, Li F, Zhang Y, Chen D, Cai J, Shi H, Aschner M, Appiah-Kubi K, He D, Lu R. Differential susceptibility of PC12 and BRL cells and the regulatory role of HIF-1α signaling pathway in response to acute methylmercury exposure under normoxia. Toxicol Lett 2020; 331:82-91. [PMID: 32461003 PMCID: PMC7366344 DOI: 10.1016/j.toxlet.2020.05.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022]
Abstract
Hypoxia-inducible factor 1 (HIF-1) is a critical nuclear transcription factor for adaptation to hypoxia; its regulatable subunit, HIF-1α, is a cytoprotective regulatory factor. We examined the effects of methylmercury (MeHg) in rat adrenal pheochromocytoma (PC12) cells and the rat hepatocyte cell line BRL. MeHg treatment led to time- and concentration-dependent toxicity in both lines with statistically significant cytotoxic effects at 5 μM and 10 μM in PC12 and BRL, respectively, at 0.5 h. HIF-1α protein levels were significantly decreased at 2.5 (PC12) and 5 (BRL) μM MeHg. Furthermore, MeHg reduced the protein levels of HIF-1α and its target genes (glucose transporter-1, vascular endothelial growth factor-A and erythropoietin). Overexpression of HIF-1α significantly attenuated MeHg-induced toxicity in both cell types. Notably, cobalt chloride, a pharmacological inducer of HIF-1α, significantly attenuated MeHg-induced toxicity in BRL but not PC12. In both cell lines, an inhibitor of prolyl hydroxylase, 3, 4-dihydroxybenzoic acid, and the proteasome inhibitor carbobenzoxy-L-leucyl-L-leucyl-L-leucinal(MG132), antagonized MeHg toxicity, while 2-methoxyestradiol, a HIF-1α inhibitor, significantly increased it. These data establish that: (a) neuron-like PC12 cells are more sensitive to MeHg than non-neuronal BRL cells; (b) HIF-1α plays a similar role in MeHg-induced toxicity in both cell lines; and (c) upregulation of HIF-1α offers general cytoprotection against MeHg toxicity in PC12 and BRL cell lines.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Qianqian Gao
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Changsheng Yin
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jie Chang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hai Qian
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yubin Zhang
- Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai 200032, China
| | - Da Chen
- School of Environment, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jiyang Cai
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Lindsay, Oklahoma City, OK 73104, USA
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kwaku Appiah-Kubi
- Department of Applied Biology, C. K. Tedam University of Technology and Applied Sciences, Navrongo, UK-0215-5321, Ghana
| | - Dawei He
- Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215130, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215130, China.
| |
Collapse
|
24
|
Malcon LMC, Wearick-Silva LE, Zaparte A, Orso R, Luft C, Tractenberg SG, Donadio MVF, de Oliveira JR, Grassi-Oliveira R. Maternal separation induces long-term oxidative stress alterations and increases anxiety-like behavior of male Balb/cJ mice. Exp Brain Res 2020; 238:2097-2107. [PMID: 32656651 DOI: 10.1007/s00221-020-05859-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/20/2020] [Indexed: 01/23/2023]
Abstract
Early life stress (ELS) exposure is a well-known risk factor for the development of psychiatric conditions, including anxiety disorder. Preclinical studies show that maternal separation (MS), a classical model of ELS, causes hypothalamic-pituitary-adrenal (HPA) axis alterations, a key contributor to the stress response modulation. Given that HPA axis activation has been shown to induce oxidative stress, it is possible to hypothesize that oxidative stress mediates the relationship between chronic ELS exposure and the development of several disorders. Here, we investigate the effects of MS in the oxidative status [plasma and brain reduced glutathione, catalase and thiobarbituric acid reactive substances (TBARS)], metabolism (glucose, triglycerides and cholesterol) and anxiety-like behaviors in adult Balb/cJ mice. In short, we found that MS increased anxiety-like behaviors in the open field, light/dark test but not in the elevated-plus maze. Animals also presented increased circulating cholesterol, increased TBARS in the plasma and decreased catalase in the hippocampus. Our findings suggest that MS induces long-term alterations in oxidative stress and increased anxiety-like behaviors.
Collapse
Affiliation(s)
- Luiza Martins Costa Malcon
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Luis Eduardo Wearick-Silva
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Aline Zaparte
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Rodrigo Orso
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Carolina Luft
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Saulo Gantes Tractenberg
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil.,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Márcio Vinicius Fagundes Donadio
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Avenida Ipiranga, 6681, prédio 11, sala 936-Partenon, Porto Alegre, RS, 90619-900, Brazil. .,Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil.
| |
Collapse
|
25
|
Alterations in the Antioxidant Enzyme Activities in the Neurodevelopmental Rat Model of Schizophrenia Induced by Glutathione Deficiency during Early Postnatal Life. Antioxidants (Basel) 2020; 9:antiox9060538. [PMID: 32575563 PMCID: PMC7346228 DOI: 10.3390/antiox9060538] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 01/24/2023] Open
Abstract
The aim of the present study was to assess the effects of l-buthionine-(S,R)-sulfoximine (BSO), a glutathione (GSH) synthesis inhibitor, and GBR 12909, a dopamine reuptake inhibitor, administered alone or in combination to Sprague-Dawley rats during early postnatal development (p5-p16), on the levels of reactive oxygen species (ROS), lipid peroxidation (LP) and the activities of antioxidant enzymes: superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and glutathione disulfide reductase (GR) in peripheral tissues (liver, kidney) and selected brain structures (prefrontal cortex, PFC; hippocampus, HIP; and striatum, STR) of 16-day-old rats. The studied parameters were analyzed with reference to the content of GSH and sulfur amino acids, methionine (Met) and cysteine (Cys) described in our previous study. This analysis showed that treatment with a BSO + GBR 12909 combination caused significant decreases in the lipid peroxidation levels in the PFC and HIP, in spite of there being no changes in ROS. The reduction of lipid peroxidation indicates a weakening of the oxidative power of the cells, and a shift in balance in favor of reducing processes. Such changes in cellular redox signaling in the PFC and HIP during early postnatal development may result in functional changes in adulthood.
Collapse
|
26
|
THONGSAW A, SANANMUANG R, UDNAN Y, AMPIAH-BONNEY RJ, CHAIYASITH WC. Immobilized Activated Carbon as Sorbent in Solid Phase Extraction with Cold Vapor Atomic Absorption Spectrometry for the Preconcentration and Determination of Mercury Species in Water and Freshwater Fish Samples. ANAL SCI 2019; 35:1195-1202. [DOI: 10.2116/analsci.19p164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Arnon THONGSAW
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| | - Ratana SANANMUANG
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| | - Yuthapong UDNAN
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| | | | - Wipharat Chuachuad CHAIYASITH
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| |
Collapse
|
27
|
Karri R, Chalana A, Kumar B, Jayadev SK, Roy G. Exploiting the κ 2 -Fashioned Coordination of [Se 2 ]-Donor Ligand L 3 Se for Facile Hg-C Bond Cleavage of Mercury Alkyls and Cytoprotection against Methylmercury-Induced Toxicity. Chemistry 2019; 25:12810-12819. [PMID: 31298434 DOI: 10.1002/chem.201902578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/09/2019] [Indexed: 11/10/2022]
Abstract
The Hg-C bond of MeHgCl, a ubiquitous environmental toxicant, is notoriously inert and exceedingly difficult to cleave. The cleavage of the Hg-C bond of MeHgCl at low temperature, therefore, is of significant importance for human health. Among various bis(imidazole)-2-selones Ln Se (n=1-4, or 6), the three-spacer L3 Se shows extraordinarily high reactivity in the degradation of various mercury alkyls including MeHgCl because of its unique ability to coordinate through κ2 -fashion, in which both the Se atoms simultaneously attack the Hg center of mercury alkyls for facile Hg-C bond cleavage. It has the highest softness (σ) parameter and the lowest HOMO(Ln Se)-LUMO(MeHgX) energy gap and, thus, L3 Se is the most reactive among Ln Se towards MeHgX (X=Cl or I). L3 Se is highly efficient, more than L1 Se, in restoring the activity of antioxidant enzyme glutathione reductase (GR) that is completely inhibited by MeHgCl; 80 % GR activity is recovered by L3 Se relative to 50 % by L1 Se. It shows an excellent cytoprotective effect in liver cells against MeHgCl-induced oxidative stress by protecting vital antioxidant enzymes from inhibition caused by MeHgCl and, thus, does not allow to increase the intracellular reactive oxygen species (ROS) levels. Furthermore, it protects the mitochondrial membrane potential (ΔΨm ) from perturbation by MeHgCl. Major Hg-responsive genes analyses demonstrate that L3 Se plays a significant role in MeHg+ detoxification in liver cells.
Collapse
Affiliation(s)
- Ramesh Karri
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Ashish Chalana
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Binayak Kumar
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Sri Krishna Jayadev
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Gouriprasanna Roy
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| |
Collapse
|
28
|
Takahashi T, Shimohata T. Vascular Dysfunction Induced by Mercury Exposure. Int J Mol Sci 2019; 20:E2435. [PMID: 31100949 PMCID: PMC6566353 DOI: 10.3390/ijms20102435] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/10/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
Methylmercury (MeHg) causes severe damage to the central nervous system, and there is increasing evidence of the association between MeHg exposure and vascular dysfunction, hemorrhage, and edema in the brain, but not in other organs of patients with acute MeHg intoxication. These observations suggest that MeHg possibly causes blood-brain barrier (BBB) damage. MeHg penetrates the BBB into the brain parenchyma via active transport systems, mainly the l-type amino acid transporter 1, on endothelial cell membranes. Recently, exposure to mercury has significantly increased. Numerous reports suggest that long-term low-level MeHg exposure can impair endothelial function and increase the risks of cardiovascular disease. The most widely reported mechanism of MeHg toxicity is oxidative stress and related pathways, such as neuroinflammation. BBB dysfunction has been suggested by both in vitro and in vivo models of MeHg intoxication. Therapy targeted at both maintaining the BBB and suppressing oxidative stress may represent a promising therapeutic strategy for MeHg intoxication. This paper reviews studies on the relationship between MeHg exposure and vascular dysfunction, with a special emphasis on the BBB.
Collapse
Affiliation(s)
- Tetsuya Takahashi
- Department of Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata 950-2085, Japan.
| | - Takayoshi Shimohata
- Department of Neurology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.
| |
Collapse
|
29
|
Takasugi N, Hiraoka H, Nakahara K, Akiyama S, Fujikawa K, Nomura R, Furuichi M, Uehara T. The Emerging Role of Electrophiles as a Key Regulator for Endoplasmic Reticulum (ER) Stress. Int J Mol Sci 2019; 20:E1783. [PMID: 30974903 PMCID: PMC6480251 DOI: 10.3390/ijms20071783] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/28/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
The unfolded protein response (UPR) is activated by the accumulation of misfolded proteins in the endoplasmic reticulum (ER), which is called ER stress. ER stress sensors PERK, IRE1, and ATF6 play a central role in the initiation and regulation of the UPR; they inhibit novel protein synthesis and upregulate ER chaperones, such as protein disulfide isomerase, to remove unfolded proteins. However, when recovery from ER stress is difficult, the UPR pathway is activated to eliminate unhealthy cells. This signaling transition is the key event of many human diseases. However, the precise mechanisms are largely unknown. Intriguingly, reactive electrophilic species (RES), which exist in the environment or are produced through cellular metabolism, have been identified as a key player of this transition. In this review, we focused on the function of representative RES: nitric oxide (NO) as a gaseous RES, 4-hydroxynonenal (HNE) as a lipid RES, and methylmercury (MeHg) as an environmental organic compound RES, to outline the relationship between ER stress and RES. Modulation by RES might be a target for the development of next-generation therapy for ER stress-associated diseases.
Collapse
Affiliation(s)
- Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Hideki Hiraoka
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Kengo Nakahara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Shiori Akiyama
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Kana Fujikawa
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Ryosuke Nomura
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Moeka Furuichi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan.
| |
Collapse
|
30
|
Nogara PA, Oliveira CS, Schmitz GL, Piquini PC, Farina M, Aschner M, Rocha JBT. Methylmercury's chemistry: From the environment to the mammalian brain. Biochim Biophys Acta Gen Subj 2019; 1863:129284. [PMID: 30659885 DOI: 10.1016/j.bbagen.2019.01.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/14/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Methylmercury is a neurotoxicant that is found in fish and rice. MeHg's toxicity is mediated by blockage of -SH and -SeH groups of proteins. However, the identification of MeHg's targets is elusive. Here we focus on the chemistry of MeHg in the abiotic and biotic environment. The toxicological chemistry of MeHg is complex in metazoans, but at the atomic level it can be explained by exchange reactions of MeHg bound to -S(e)H with another free -S(e)H group (R1S(e)-HgMe + R2-S(e)H ↔ R1S(e)H + R2-S(e)-HgMe). This reaction was first studied by professor Rabenstein and here it is referred as the "Rabenstein's Reaction". The absorption, distribution, and excretion of MeHg in the environment and in the body of animals will be dictated by Rabenstein's reactions. The affinity of MeHg by thiol and selenol groups and the exchange of MeHg by Rabenstein's Reaction (which is a diffusion controlled reaction) dictates MeHg's neurotoxicity. However, it is important to emphasize that the MeHg exchange reaction velocity with different types of thiol- and selenol-containing proteins will also depend on protein-specific structural and thermodynamical factors. New experimental approaches and detailed studies about the Rabenstein's reaction between MeHg with low molecular mass thiol (LMM-SH) molecules (cysteine, GSH, acetyl-CoA, lipoate, homocysteine) with abundant high molecular mass thiol (HMM-SH) molecules (albumin, hemoglobin) and HMM-SeH (GPxs, Selenoprotein P, TrxR1-3) are needed. The study of MeHg migration from -S(e)-Hg- bonds to free -S(e)H groups (Rabenstein's Reaction) in pure chemical systems and neural cells (with special emphasis to the LMM-SH and HMM-S(e)H molecules cited above) will be critical to developing realistic constants to be used in silico models that will predict the distribution of MeHg in humans.
Collapse
Affiliation(s)
- Pablo A Nogara
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Cláudia S Oliveira
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela L Schmitz
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Paulo C Piquini
- Departamento de Física, CCNE, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
31
|
Farina M, Aschner M. Glutathione antioxidant system and methylmercury-induced neurotoxicity: An intriguing interplay. Biochim Biophys Acta Gen Subj 2019; 1863:129285. [PMID: 30659883 DOI: 10.1016/j.bbagen.2019.01.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/09/2018] [Accepted: 01/09/2019] [Indexed: 01/20/2023]
Abstract
Methylmercury (MeHg) is a toxic chemical compound naturally produced mainly in the aquatic environment through the methylation of inorganic mercury catalyzed by aquatic microorganisms. MeHg is biomagnified in the aquatic food chain and, consequently, piscivorous fish at the top of the food chain possess huge amounts of MeHg (at the ppm level). Some populations that have fish as main protein's source can be exposed to exceedingly high levels of MeHg and develop signs of toxicity. MeHg is toxic to several organs, but the central nervous system (CNS) represents a preferential target, especially during development (prenatal and early postnatal periods). Though the biochemical events involved in MeHg-(neuro)toxicity are not yet entirely comprehended, a vast literature indicates that its pro-oxidative properties explain, at least partially, several of its neurotoxic effects. As result of its electrophilicity, MeHg interacts with (and oxidize) nucleophilic groups, such as thiols and selenols, present in proteins or low-molecular weight molecules. It is noteworthy that such interactions modify the redox state of these groups and, therefore, lead to oxidative stress and impaired function of several molecules, culminating in neurotoxicity. Among these molecules, glutathione (GSH; a major thiol antioxidant) and thiol- or selenol-containing enzymes belonging to the GSH antioxidant system represent key molecular targets involved in MeHg-neurotoxicity. In this review, we firstly present a general overview concerning the neurotoxicity of MeHg. Then, we present fundamental aspects of the GSH-antioxidant system, as well as the effects of MeHg on this system.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, USA
| |
Collapse
|
32
|
Espitia-Pérez P, Albino SM, da Rosa HT, Silveira AK, Espitia-Pérez L, Brango H, Moraes DP, Hermann PRS, Mingori M, Barreto F, Kunzler A, Gelain DP, Schnorr CE, Moreira JCF. Effects of methylmercury and retinol palmitate co-administration in rats during pregnancy and breastfeeding: Metabolic and redox parameters in dams and their offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 162:603-615. [PMID: 30031321 DOI: 10.1016/j.ecoenv.2018.06.093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 06/08/2023]
Abstract
Ubiquitous low-dose methylmercury (MeHg) exposure through an increased fish consumption represents a global public health problem, especially among pregnant women. A plethora of micronutrients presented in fish affects MeHg uptake/distribution, but limited data is available. Vitamin A (VitA), another fish micronutrient is used in nutritional supplementation, especially during pregnancy. However, there is no information about the health effects arising from their combined exposure. Therefore, the present study aimed to examine the effects of both MeHg and retinyl palmitate administered on pregnant and lactating rats in metabolic and redox parameters from dams and their offspring. Thirty Wistar female rats were orally supplemented with MeHg (0,5 mg/kg/day) and retinyl palmitate (7500 µg RAE/kg/day) via gavage, either individually or in combination from the gestational day 0 to weaning. For dams (150 days old) and their offspring (31 days old), glycogen accumulation (hepatic and cardiac) and retinoid contents (plasma and liver) were analyzed. Hg deposition in liver tissue was quantified. Redox parameters (liver, kidney, and heart) were evaluated for both animals. Cytogenetic damage was analyzed with micronucleus test. Our results showed no general toxic or metabolic alterations in dams and their offspring by MeHg-VitA co-administration during pregnancy and lactation. However, increased lipoperoxidation in maternal liver and a disrupted pro-oxidant response in the heart of male pups was encountered, with apparently no particular effects in the antioxidant response in female offspring. GST activity in dam kidney was altered leading to possible redox disruption of this tissue with no alterations in offspring. Finally, the genomic damage was exacerbated in both male and female pups. In conclusion, low-dose MeHg exposure and retinyl palmitate supplementation during gestation and lactation produced a potentiated pro-oxidant effect, which was tissue-specific. Although this is a pre-clinical approach, we recommend precaution for pregnant women regarding food consumption, and we encourage more epidemiological studies to assess possible modulations effects of MeHg-VitA co-administration at safe or inadvertently used doses in humans, which may be related to specific pathologies in mothers and their children.
Collapse
Affiliation(s)
- Pedro Espitia-Pérez
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Suelen Marin Albino
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Helen Tais da Rosa
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Alexandre Kleber Silveira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Lyda Espitia-Pérez
- Facultad de Ciencias de la Salud, Laboratorio de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Calle 38 Carrera 1W, Barrio Juan XXIII, Montería, Córdoba, Colombia
| | - Hugo Brango
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, Brazil
| | - Diogo Pompéu Moraes
- Instituto de Química, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves 9500, CEP 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Paolla Rissi Silva Hermann
- Instituto de Química, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves 9500, CEP 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Moara Mingori
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fabiano Barreto
- Laboratório de Análise de Resíduos de Pesticidas e Medicamentos Veterinários (RPM), Laboratório Nacional Agropecuário RS, Estrada da Ponta Grossa 3036, CEP: 91780-580 Porto Alegre, Rio Grande do Sul, Brazil
| | - Alice Kunzler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Eduardo Schnorr
- Departamento de Civil y Ambiental, Programa de Ingeniería Ambiental, Universidad de la Costa, Calle 58 #55- 66, Barranquilla, Atlántico, Colombia
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, Anexo Depto. Bioquímica, Lab 32, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
33
|
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
|
34
|
Sumathi T, Jacob S, Gopalakrishnan R. Methylmercury exposure develops atherosclerotic risk factors in the aorta and programmed cell death in the cerebellum: ameliorative action of Celastrus paniculatus ethanolic extract in male Wistar rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:30212-30223. [PMID: 30155631 DOI: 10.1007/s11356-018-3031-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/21/2018] [Indexed: 06/08/2023]
Abstract
Methylmercury (MeHg) is a bioaccumulative global environmental contaminant present in fishes and seafood. MeHg is the methylated form of mercury emitted from diverse anthropogenic and natural sources. MeHg is accumulated in the aquatic environment and eventually reaches human system via food chain by biomagnification. We have reported previously that the neurotoxic effect of MeHg in rat cerebellum is mitigated by the administration of an ayurvedic medicinal plant, Celastrus paniculatus ethanolic extract. The present study has focussed to further explore the mechanism of action of Celastrus paniculatus against MeHg-induced neurotoxicity in the cerebellum. We have also inspected the effect of Celastrus paniculatus (CP) against MeHg-induced atherosclerotic risk factors like alterations in antioxidant levels, aortic lipid profile, and aortic histology by MeHg in the largest vasculature, aorta, which are the initiating factors of cardiovascular diseases. Male Wistar rats were divided as (i) control, (ii) MeHg (5 mg/kg b.w.), (iii) MeHg + CP (200 mg/kg b.w.), and (iv) CP alone (200 mg/kg b.w.). All were given orally for 21 days. In cerebellum Celastrus paniculatus, there were increased mitochondrial electron transport chain (p < 0.05) activity, reduced cytochrome c release (p < 0.05), and caspase 3 mRNA expression (p < 0.05). In the aorta, MeHg-induced oxidative stress, lipid profile changes, and endothelial denudation were ameliorated by Celastrus paniculatus. Hence, we conclude that Celastrus paniculatus protects against MeHg toxicity by inhibiting mitochondrial cytochrome c/caspase 3 apoptotic pathway in the cerebellum and reducing the development of atherosclerotic risk factors in the aorta.
Collapse
Affiliation(s)
- Thangarajan Sumathi
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600113, India.
| | - Sherin Jacob
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600113, India
| | - Rahul Gopalakrishnan
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600113, India
| |
Collapse
|
35
|
Jacob S, Thangarajan S. Fisetin impedes developmental methylmercury neurotoxicity via downregulating apoptotic signalling pathway and upregulating Rho GTPase signalling pathway in hippocampus of F 1 generation rats. Int J Dev Neurosci 2018; 69:88-96. [PMID: 30009881 DOI: 10.1016/j.ijdevneu.2018.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/14/2018] [Accepted: 07/08/2018] [Indexed: 01/14/2023] Open
Abstract
Methyl mercury is a teratogenic and neurodevelopmental toxicant in the environment. MeHg affects several biological pathways critical for brain development. The present study validated the effect of Fisetin on developmental MeHg exposure induced alterations in mitochondrial apoptotic pathway and Rho GTPase mRNA expressions in hippocampus of F1 generation rats. Pregnant Wistar rats were grouped as Group I : administered with vehicle control, Group II: MeHg (1.5 mg/kg b.w), Group III: MeHg + Fisetin (10 mg/kg b.w), Group IV: MeHg + Fisetin (30 mg/kg b.w), Group V: MeHg + Fisetin (50 mg/kg b.w), Group VI: MeHg + Fisetin (70 mg/kg b.w), Group VII: Fisetin (30 mg/kg b.w) alone. Fisetin reduced mercury accumulation in offspring brain. In hippocampus, Fisetin preserved mitochondrial total thiol status, glutathione antioxidant system, mitochondrial metabolic integrity and respiratory chain activity. Fisetin ameliorated apoptotic signals by preventing Cytochrome c release, down regulating ERK 1/2 and Caspase 3 gene expression. Fisetin also upregulated mRNA expressions of RhoA/Rac1/Cdc42 in hippocampus. Predominant effect of Fisetin was to reduce mercury accumulation in offspring brain there by diminishing the toxic effect of MeHg. Hence we showed that, gestational intake of Fisetin (30 mg/kg b.w.) impedes developmental MeHg neurotoxicity by regulating mitochondrial apoptotic and Rho GTPase signalling molecules and by reducing the mercury accumulation in hippocampus of F1 generation rats.
Collapse
Affiliation(s)
- Sherin Jacob
- Dr.ALMPG IBMS, University of Madras, Taramani Campus, Chennai, 600113, Tamil Nadu, India
| | - Sumathi Thangarajan
- Dr.ALMPG IBMS, University of Madras, Taramani Campus, Chennai, 600113, Tamil Nadu, India.
| |
Collapse
|
36
|
Garza-Lombó C, Posadas Y, Quintanar L, Gonsebatt ME, Franco R. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid Redox Signal 2018; 28:1669-1703. [PMID: 29402131 PMCID: PMC5962337 DOI: 10.1089/ars.2017.7272] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Essential metals such as copper, iron, manganese, and zinc play a role as cofactors in the activity of a wide range of processes involved in cellular homeostasis and survival, as well as during organ and tissue development. Throughout our life span, humans are also exposed to xenobiotic metals from natural and anthropogenic sources, including aluminum, arsenic, cadmium, lead, and mercury. It is well recognized that alterations in the homeostasis of essential metals and an increased environmental/occupational exposure to xenobiotic metals are linked to several neurological disorders, including neurodegeneration and neurodevelopmental alterations. Recent Advances: The redox activity of essential metals is key for neuronal homeostasis and brain function. Alterations in redox homeostasis and signaling are central to the pathological consequences of dysfunctional metal ion homeostasis and increased exposure to xenobiotic metals. Both redox-active and redox-inactive metals trigger oxidative stress and damage in the central nervous system, and the exact mechanisms involved are starting to become delineated. CRITICAL ISSUES In this review, we aim to appraise the role of essential metals in determining the redox balance in the brain and the mechanisms by which alterations in the homeostasis of essential metals and exposure to xenobiotic metals disturb the cellular redox balance and signaling. We focus on recent literature regarding their transport, metabolism, and mechanisms of toxicity in neural systems. FUTURE DIRECTIONS Delineating the specific mechanisms by which metals alter redox homeostasis is key to understand the pathological processes that convey chronic neuronal dysfunction in neurodegenerative and neurodevelopmental disorders. Antioxid. Redox Signal. 28, 1669-1703.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska.,2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Yanahi Posadas
- 3 Departamentos de Farmacología y de, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México .,4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - Liliana Quintanar
- 4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - María E Gonsebatt
- 2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Rodrigo Franco
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska
| |
Collapse
|
37
|
Prince LM, Rand MD. Methylmercury exposure causes a persistent inhibition of myogenin expression and C2C12 myoblast differentiation. Toxicology 2018; 393:113-122. [PMID: 29104120 PMCID: PMC5757876 DOI: 10.1016/j.tox.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxicant, best known for its selective targeting of the developing nervous system. MeHg exposure has been shown to cause motor deficits such as impaired gait and coordination, muscle weakness, and muscle atrophy, which have been associated with disruption of motor neurons. However, recent studies have suggested that muscle may also be a target of MeHg toxicity, both in the context of developmental myogenic events and of low-level chronic exposures affecting muscle wasting in aging. We therefore investigated the effects of MeHg on myotube formation, using the C2C12 mouse myoblast model. We found that MeHg inhibits both differentiation and fusion, in a concentration-dependent manner. Furthermore, MeHg specifically and persistently inhibits myogenin (MyoG), a transcription factor involved in myocyte differentiation, within the first six hours of exposure. MeHg-induced reduction in MyoG expression is contemporaneous with a reduction of a number of factors involved in mitochondrial biogenesis and mtDNA transcription and translation, which may implicate a role for mitochondria in mediating MeHg-induced change in the differentiation program. Unexpectedly, inhibition of myoblast differentiation with MeHg parallels inhibition of Notch receptor signaling. Our research establishes muscle cell differentiation as a target for MeHg toxicity, which may contribute to the underlying etiology of motor deficits with MeHg toxicity.
Collapse
Affiliation(s)
- Lisa M Prince
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| |
Collapse
|
38
|
Ihara H, Kasamatsu S, Kitamura A, Nishimura A, Tsutsuki H, Ida T, Ishizaki K, Toyama T, Yoshida E, Abdul Hamid H, Jung M, Matsunaga T, Fujii S, Sawa T, Nishida M, Kumagai Y, Akaike T. Exposure to Electrophiles Impairs Reactive Persulfide-Dependent Redox Signaling in Neuronal Cells. Chem Res Toxicol 2017; 30:1673-1684. [PMID: 28837763 DOI: 10.1021/acs.chemrestox.7b00120] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Electrophiles such as methylmercury (MeHg) affect cellular functions by covalent modification with endogenous thiols. Reactive persulfide species were recently reported to mediate antioxidant responses and redox signaling because of their strong nucleophilicity. In this study, we used MeHg as an environmental electrophile and found that exposure of cells to the exogenous electrophile elevated intracellular concentrations of the endogenous electrophilic molecule 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), accompanied by depletion of reactive persulfide species and 8-SH-cGMP which is a metabolite of 8-nitro-cGMP. Exposure to MeHg also induced S-guanylation and activation of H-Ras followed by injury to cerebellar granule neurons. The electrophile-induced activation of redox signaling and the consequent cell damage were attenuated by pretreatment with a reactive persulfide species donor. In conclusion, exogenous electrophiles such as MeHg with strong electrophilicity impair the redox signaling regulatory mechanism, particularly of intracellular reactive persulfide species and therefore lead to cellular pathogenesis. Our results suggest that reactive persulfide species may be potential therapeutic targets for attenuating cell injury by electrophiles.
Collapse
Affiliation(s)
- Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University , Osaka 599-8531, Japan
| | - Shingo Kasamatsu
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University , Osaka 599-8531, Japan.,Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| | - Atsushi Kitamura
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University , Osaka 599-8531, Japan
| | - Akira Nishimura
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| | - Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University , Kumamoto 860-8556, Japan
| | - Tomoaki Ida
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| | - Kento Ishizaki
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University , Osaka 599-8531, Japan
| | - Takashi Toyama
- Environmental Biology Section, Faculty of Medicine, University of Tsukuba , Tsukuba, Ibaraki 305-8575, Japan
| | - Eiko Yoshida
- Environmental Biology Section, Faculty of Medicine, University of Tsukuba , Tsukuba, Ibaraki 305-8575, Japan
| | - Hisyam Abdul Hamid
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan.,Department of Pharmaceutical Pharmacology and Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA Puncak Alam Campus , 42300 Puncak Alam, Selangor, Malaysia
| | - Minkyung Jung
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| | - Shigemoto Fujii
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University , Kumamoto 860-8556, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences , Aichi 444-8787, Japan
| | - Yoshito Kumagai
- Environmental Biology Section, Faculty of Medicine, University of Tsukuba , Tsukuba, Ibaraki 305-8575, Japan
| | - Takaaki Akaike
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine , Sendai 980-8575, Japan
| |
Collapse
|
39
|
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]
|
40
|
Jacob S, Thangarajan S. Effect of Gestational Intake of Fisetin (3,3',4',7-Tetrahydroxyflavone) on Developmental Methyl Mercury Neurotoxicity in F 1 Generation Rats. Biol Trace Elem Res 2017; 177:297-315. [PMID: 27815688 DOI: 10.1007/s12011-016-0886-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/24/2016] [Indexed: 12/16/2022]
Abstract
Methyl mercury (MeHg) is a developmental neurotoxin that causes irreversible cognitive damage in offspring of gestationally exposed mothers. Currently, no preventive drugs are established against MeHg developmental neurotoxicity. The neuroprotective effect of gestational administration of a flavanoid against in utero toxicity of MeHg is not explored much. Hence, the present study validated the effect of a bioactive flavanoid, fisetin, on MeHg developmental neurotoxicity outcomes in rat offspring at postnatal weaning age. Pregnant Wistar rats were simultaneously given MeHg (1.5 mg/kg b.w.) and two doses of fisetin (10 and 50 mg/kg b.w. in two separate groups) orally from gestational day (GD) 5 till parturition. Accordingly, after parturition, on postnatal day (PND) 24, weaning F1 generation rats were studied for motor and cognitive behavioural changes. Biochemical and histopathological changes were also studied in the cerebral cortex, cerebellum and hippocampus on PND 25. Administration of fisetin during pregnancy prevented behavioural impairment due to transplacental MeHg exposure in weaning rats. Fisetin decreased the levels of oxidative stress markers, increased enzymatic and non-enzymatic antioxidant levels and increased the activity of membrane-bound ATPases and cholinergic function in F1 generation rats. In light microscopic studies, fisetin treatment protected the specific offspring brain regions from significant morphological aberrations. Between the two doses of fisetin studied, 10 mg/kg b.w. was found to be more satisfactory and effective than 50 mg/kg b.w. The present study shows that intake of fisetin during pregnancy in rats ameliorated in utero MeHg exposure-induced neurotoxicity outcomes in postnatal weaning F1 generation rats.
Collapse
Affiliation(s)
- Sherin Jacob
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600 113, India
| | - Sumathi Thangarajan
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600 113, India.
| |
Collapse
|
41
|
Lovato FL, Teixeira da Rocha JB, Dalla Corte CL. Diphenyl Diselenide Protects against Methylmercury-Induced Toxicity in Saccharomyces cerevisiae via the Yap1 Transcription Factor. Chem Res Toxicol 2017; 30:1134-1144. [DOI: 10.1021/acs.chemrestox.6b00449] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Fabricio Luís Lovato
- Departamento
de Bioquímica e Biologia Molecular, Programa de Pós-graduação
em Ciências Biológicas: Bioquímica Toxicológica,
Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Cep 97105-900 Santa Maria, RS, Brazil
| | - João Batista Teixeira da Rocha
- Departamento
de Bioquímica e Biologia Molecular, Programa de Pós-graduação
em Ciências Biológicas: Bioquímica Toxicológica,
Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Cep 97105-900 Santa Maria, RS, Brazil
| | - Cristiane Lenz Dalla Corte
- Departamento
de Bioquímica e Biologia Molecular, Programa de Pós-graduação
em Ciências Biológicas: Bioquímica Toxicológica,
Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Cep 97105-900 Santa Maria, RS, Brazil
- Universidade Federal do Pampa, Campus
Caçapava do Sul, Av. Pedro Anunciação, 111, Vila
Batista, 96570-000 Caçapava do Sul, RS, Brazil
| |
Collapse
|
42
|
α-Ketoadipic Acid and α-Aminoadipic Acid Cause Disturbance of Glutamatergic Neurotransmission and Induction of Oxidative Stress In Vitro in Brain of Adolescent Rats. Neurotox Res 2017; 32:276-290. [DOI: 10.1007/s12640-017-9735-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 12/21/2022]
|
43
|
Meinerz DF, Branco V, Aschner M, Carvalho C, Rocha JBT. Diphenyl diselenide protects against methylmercury-induced inhibition of thioredoxin reductase and glutathione peroxidase in human neuroblastoma cells: a comparison with ebselen. J Appl Toxicol 2017; 37:1073-1081. [DOI: 10.1002/jat.3458] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Daiane F. Meinerz
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas; Universidade Federal de Santa Maria; 97105-900 Santa Maria RS Brazil
| | - Vasco Branco
- Research Institute for Medicines (iMed.ULisboa); Faculty of Pharmacy, Universidade de Lisboa; Av. Prof. Gama Pinto 1649-003 Lisbon Portugal
| | - Michael Aschner
- Department of Molecular Pharmacology; Albert Einstein College of Medicine; Forchheimer 209, 1300 Morris Park Avenue Bronx NY 10461 USA
| | - Cristina Carvalho
- Research Institute for Medicines (iMed.ULisboa); Faculty of Pharmacy, Universidade de Lisboa; Av. Prof. Gama Pinto 1649-003 Lisbon Portugal
| | - João Batista T. Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas; Universidade Federal de Santa Maria; 97105-900 Santa Maria RS Brazil
| |
Collapse
|
44
|
Wyatt LH, Luz AL, Cao X, Maurer LL, Blawas AM, Aballay A, Pan WKY, Meyer JN. Effects of methyl and inorganic mercury exposure on genome homeostasis and mitochondrial function in Caenorhabditis elegans. DNA Repair (Amst) 2017; 52:31-48. [PMID: 28242054 PMCID: PMC5394729 DOI: 10.1016/j.dnarep.2017.02.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 12/05/2016] [Accepted: 02/06/2017] [Indexed: 12/21/2022]
Abstract
Mercury toxicity mechanisms have the potential to induce DNA damage and disrupt cellular processes, like mitochondrial function. Proper mitochondrial function is important for cellular bioenergetics and immune signaling and function. Reported impacts of mercury on the nuclear genome (nDNA) are conflicting and inconclusive, and mitochondrial DNA (mtDNA) impacts are relatively unknown. In this study, we assessed genotoxic (mtDNA and nDNA), metabolic, and innate immune impacts of inorganic and organic mercury exposure in Caenorhabditis elegans. Genotoxic outcomes measured included DNA damage, DNA damage repair (nucleotide excision repair, NER; base excision repair, BER), and genomic copy number following MeHg and HgCl2 exposure alone and in combination with known DNA damage-inducing agents ultraviolet C radiation (UVC) and hydrogen peroxide (H2O2), which cause bulky DNA lesions and oxidative DNA damage, respectively. Following exposure to both MeHg and HgCl2, low-level DNA damage (∼0.25 lesions/10kb mtDNA and nDNA) was observed. Unexpectedly, a higher MeHg concentration reduced damage in both genomes compared to controls. However, this observation was likely the result of developmental delay. In co-exposure treatments, both mercury compounds increased initial DNA damage (mtDNA and nDNA) in combination with H2O2 exposure, but had no impact in combination with UVC exposure. Mercury exposure both increased and decreased DNA damage removal via BER. DNA repair after H2O2 exposure in mercury-exposed nematodes resulted in damage levels lower than measured in controls. Impacts to NER were not detected. mtDNA copy number was significantly decreased in the MeHg-UVC and MeHg-H2O2 co-exposure treatments. Mercury exposure had metabolic impacts (steady-state ATP levels) that differed between the compounds; HgCl2 exposure decreased these levels, while MeHg slightly increased levels or had no impact. Both mercury species reduced mRNA levels for immune signaling-related genes, but had mild or no effects on survival on pathogenic bacteria. Overall, mercury exposure disrupted mitochondrial endpoints in a mercury-compound dependent fashion.
Collapse
Affiliation(s)
- Lauren H Wyatt
- Nicholas School of the Environment, Duke University, Durham, NC, United States.
| | - Anthony L Luz
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | - Xiou Cao
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| | - Laura L Maurer
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | - Ashley M Blawas
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | - Alejandro Aballay
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| | - William K Y Pan
- Nicholas School of the Environment, Duke University, Durham, NC, United States; Duke Global Health Institute, Duke University, Durham, NC, United States
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, NC, United States.
| |
Collapse
|
45
|
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
|
46
|
Cecatto C, Amaral AU, da Silva JC, Wajner A, Godoy KDS, Ribeiro RT, Gonçalves ADM, Vargas CR, Wajner M. Mevalonolactone disrupts mitochondrial functions and induces permeability transition pore opening in rat brain mitochondria: Implications for the pathogenesis of mevalonic aciduria. Neurochem Int 2017; 108:133-145. [PMID: 28284974 DOI: 10.1016/j.neuint.2017.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/07/2017] [Accepted: 03/06/2017] [Indexed: 12/13/2022]
Abstract
Mevalonic aciduria (MVA) is caused by severe deficiency of mevalonic kinase activity leading to tissue accumulation and high urinary excretion of mevalonic acid (MA) and mevalonolactone (ML). Patients usually present severe neurologic symptoms whose pathophysiology is poorly known. Here, we tested the hypothesis that the major accumulating metabolites are toxic by investigating the in vitro effects of MA and ML on important mitochondrial functions in rat brain and liver mitochondria. ML, but not MA, markedly decreased mitochondrial membrane potential (ΔΨm), NAD(P)H content and the capacity to retain Ca2+ in the brain, besides inducing mitochondrial swelling. These biochemical alterations were totally prevented by the classical inhibitors of mitochondrial permeability transition (MPT) cyclosporine A and ADP, as well as by ruthenium red in Ca2+-loaded mitochondria, indicating the involvement of MPT and an important role for mitochondrial Ca2+ in these effects. ML also induced lipid peroxidation and markedly inhibited aconitase activity, an enzyme that is highly susceptible to free radical attack, in brain mitochondrial fractions, indicating that lipid and protein oxidative damage may underlie some of ML-induced deleterious effects including MTP induction. In contrast, ML and MA did not compromise oxidative phosphorylation in the brain and all mitochondrial functions evaluated in the liver, evidencing a selective toxicity of ML towards the central nervous system. Our present study provides for the first time evidence that ML impairs essential brain mitochondrial functions with the involvement of MPT pore opening. It is therefore presumed that disturbance of brain mitochondrial homeostasis possibly contributes to the neurologic symptoms in MVA.
Collapse
Affiliation(s)
- Cristiane Cecatto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Janaína Camacho da Silva
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alessandro Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Kálita Dos Santos Godoy
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rafael Teixeira Ribeiro
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Aline de Mello Gonçalves
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carmen Regla Vargas
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
47
|
Karri V, Schuhmacher M, Kumar V. Heavy metals (Pb, Cd, As and MeHg) as risk factors for cognitive dysfunction: A general review of metal mixture mechanism in brain. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 48:203-213. [PMID: 27816841 DOI: 10.1016/j.etap.2016.09.016] [Citation(s) in RCA: 302] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/21/2016] [Accepted: 09/24/2016] [Indexed: 05/22/2023]
Abstract
Human exposure to toxic heavy metals is a global challenge. Concurrent exposure of heavy metals, such as lead (Pb), cadmium (Cd), arsenic (As) and methylmercury (MeHg) are particularly important due to their long lasting effects on the brain. The exact toxicological mechanisms invoked by exposure to mixtures of the metals Pb, Cd, As and MeHg are still unclear, however they share many common pathways for causing cognitive dysfunction. The combination of metals may produce additive/synergetic effects due to their common binding affinity with NMDA receptor (Pb, As, MeHg), Na+ - K+ ATP-ase pump (Cd, MeHg), biological Ca+2 (Pb, Cd, MeHg), Glu neurotransmitter (Pb, MeHg), which can lead to imbalance between the pro-oxidant elements (ROS) and the antioxidants (reducing elements). In this process, ROS dominates the antioxidants factors such as GPx, GS, GSH, MT-III, Catalase, SOD, BDNF, and CERB, and finally leads to cognitive dysfunction. The present review illustrates an account of the current knowledge about the individual metal induced cognitive dysfunction mechanisms and analyse common Mode of Actions (MOAs) of quaternary metal mixture (Pb, Cd, As, MeHg). This review aims to help advancement in mixture toxicology and development of next generation predictive model (such as PBPK/PD) combining both kinetic and dynamic interactions of metals.
Collapse
Affiliation(s)
- Venkatanaidu Karri
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Marta Schuhmacher
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Vikas Kumar
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain.
| |
Collapse
|
48
|
Ishihara Y, Tsuji M, Kawamoto T, Yamazaki T. Involvement of reactive oxygen species derived from mitochondria in neuronal injury elicited by methylmercury. J Clin Biochem Nutr 2016; 59:182-190. [PMID: 27895385 PMCID: PMC5110935 DOI: 10.3164/jcbn.16-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/04/2016] [Indexed: 01/02/2023] Open
Abstract
Methylmercury induces oxidative stress and subsequent neuronal injury. However, the mechanism by which methylmercury elicits reactive oxygen species (ROS) production remains under debate. In this study, we investigated the involvement of mitochondrial ROS in methylmercury-induced neuronal cell injury using human neuroblastoma SH-SY5Y-derived ρ0 cells, which have a deletion of mitochondrial DNA and thus decreased respiratory activity. SH-SY5Y cells were cultured for 60 days in the presence of ethidium bromide to produce ρ0 cells. Our ρ0 cells showed decreases in the cytochrome c oxidase expression and activity as well as oxygen consumption compared with original SH-SY5Y cells. Methylmercury at a concentration of 1 µM induced cell death with oxidative stress in original SH-SY5Y cells, but not ρ0 cells, indicating that ρ0 cells are resistant to methylmercury-induced oxidative stress. ρ0 cells also showed tolerance against hydrogen peroxide and superoxide anion, suggesting that ρ0 cells are resistant to total ROS. These data indicate that mitochondrial ROS are clearly involved in oxidative stress and subsequent cell death induced by methylmercury. Considering that the dominant mechanism of ROS generation elicited by methylmercury is due to direct antioxidant enzyme inhibition, mitochondria might play a role in amplifying ROS in methylmercury-induced neurotoxicity.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima 739-8521, Japan
| | - Mayumi Tsuji
- Department of Environmental Health, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka 807-8555, Japan
| | - Toshihiro Kawamoto
- Department of Environmental Health, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka 807-8555, Japan
| | - Takeshi Yamazaki
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima 739-8521, Japan
| |
Collapse
|
49
|
Maternal methylmercury from a wild-caught walleye diet induces developmental abnormalities in zebrafish. Reprod Toxicol 2016; 65:272-282. [DOI: 10.1016/j.reprotox.2016.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/03/2016] [Accepted: 08/16/2016] [Indexed: 01/20/2023]
|
50
|
Sumathi T, Christinal J. Neuroprotective Effect of Portulaca oleraceae Ethanolic Extract Ameliorates Methylmercury Induced Cognitive Dysfunction and Oxidative Stress in Cerebellum and Cortex of Rat Brain. Biol Trace Elem Res 2016; 172:155-165. [PMID: 26563420 DOI: 10.1007/s12011-015-0546-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/15/2015] [Indexed: 12/30/2022]
Abstract
Methylmercury (MeHg) is highly toxic, and its principal target tissue in human is the nervous system, which has made MeHg intoxication a public health concern for many decades. Portulaca oleraceae (purslane), a member of the Portulacaceae family, is widespread as a weed and has been ranked the eighth most common plant in the world. In this study, we sought for potential beneficial effects of Portulaca oleracea ethanolic extract (POEE) against the neurotoxicity induced by MeHg in cerebellum and cortex of rats. Male Wistar rats were administered with MeHg orally at a dose of 5 mg/kg b.w. for 21 days. Experimental rats were given MeHg and also administered with POEE (4 mg/kg, orally) 1 h prior to the administration of MeHg for 21 days. After MeHg exposure, we determine the mercury concentration by atomic absorption spectroscopy (AAS); mercury content was observed high in MeHg-induced group. POEE reduced the mercury content. We also observed that the activities of catalase, superoxide dismutase, glutathione peroxidase, and the level of glutathione were reduced. The levels of glutathione reductase and thiobarbituric acid reactive substance were found to be increased. The above biochemical changes were found to be reversed with POEE. Behavioral changes like decrease tail flick response, longer immobility time, and decreased motor activity were noted down during MeHg exposure. POEE pretreatment offered protection from these behavioral changes. MeHg intoxication also caused histopathological changes in cerebellum and cortex, which was found to be normalized by treatment with POEE. The present results indicate that POEE has protective effect against MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Thangarajan Sumathi
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600 113, India.
| | - Johnson Christinal
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600 113, India
| |
Collapse
|