1
|
Althobaiti NA. Heavy metals exposure and Alzheimer's disease: Underlying mechanisms and advancing therapeutic approaches. Behav Brain Res 2025; 476:115212. [PMID: 39187176 DOI: 10.1016/j.bbr.2024.115212] [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: 05/27/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Heavy metals such as lead, cadmium, mercury, and arsenic are prevalent in the environment due to both natural and anthropogenic sources, leading to significant public health concerns. These heavy metals are known to cause damage to the nervous system, potentially leading to a range of neurological conditions including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and attention-deficit hyperactivity disorder (ADHD). The present study examines the complex relationship between heavy metal exposure and AD, focusing on the underlying mechanisms of toxicity and potential therapeutic approaches. This review article highlights how these metals can impair brain function through mechanisms such as oxidative stress, inflammation, and neurotransmitter disruption, ultimately contributing to neurodegenerative diseases like AD. It also addresses the challenges in diagnosing heavy metal-induced cognitive impairments and emphasizes the need for further research to explore effective treatment strategies and preventive measures against heavy metal exposure.
Collapse
Affiliation(s)
- Norah A Althobaiti
- Biology Department, College of Science and Humanities, Shaqra University, Saudi Arabia.
| |
Collapse
|
2
|
Zhu Z, Wang Y, Wang Y, Fu M, Luo X, Wang G, Zhang J, Yang X, Shan W, Li C, Liu T. The association of mixed multi-metal exposure with sleep duration and self-reported sleep disorder: A subgroup analysis from the National Health and Nutrition Examination Survey (NHANES). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124798. [PMID: 39197640 DOI: 10.1016/j.envpol.2024.124798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
Sleep disorders significantly affect sleep duration and constitute a major public health issue. However, the relationship between metal exposure and sleep is not fully elucidated. This study utilized publicly available data from the National Health and Nutrition Examination Survey (NHANES) to measure blood concentrations of seven metals-copper (Cu), zinc (Zn), selenium (Se), manganese (Mn), mercury (Hg), cadmium (Cd), and lead (Pb)-in a cohort of 4263 American adults. The relationship between metal exposure and self-reported sleep duration and sleep disorder risk was analyzed using single exposure models like logistic and linear regression and mixedexposure models such as weighted quantile sum (WQS) regression and Bayesian kernel machine regression (BKMR). The results indicated an absence of statistically significant findings in the single exposure model. In contrast, the mixed exposure model revealed a positive correlation between selenium levels and the risk of sleep disorders across the entire population. A "U-shaped" association was identified between copper levels and the risk of sleep disorders in males, females, and individuals aged 60 and above. Moreover, a positive trend was observed between manganese levels and the risk of sleep disorders in individuals aged 60 and above. Additionally, elevated concentrations of metal mixtures were significantly associated with reduced sleep duration among females. Sensitivity analyses corroborated these findings. In conclusion, within the context of metal mixtures, selenium may be a risk factor for sleep disorders in the general population. Manganese may be a unique risk factor in older adults. Copper levels have a "U" shaped link to sleep disorder risk in specific population subgroups. Finally, the accumulation of blood metal mixtures in females, mainly due to lead and mercury, may reduce sleep duration. Further research is necessary to validate these findings.
Collapse
Affiliation(s)
- Zifan Zhu
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China; School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Yongjun Wang
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China.
| | - Yuanlong Wang
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, 528451, China.
| | - Maoling Fu
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xinxin Luo
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, 528451, China.
| | - Guojun Wang
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China.
| | - Jian Zhang
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China.
| | - Xiujuan Yang
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China; State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Wei Shan
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Cunxue Li
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China.
| | - Tiebang Liu
- Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, 518118, China; School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
3
|
Kang B, Wang J, Guo S, Yang L. Mercury-induced toxicity: Mechanisms, molecular pathways, and gene regulation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173577. [PMID: 38852866 DOI: 10.1016/j.scitotenv.2024.173577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/01/2024] [Accepted: 05/25/2024] [Indexed: 06/11/2024]
Abstract
Mercury is a well-known neurotoxicant for humans and wildlife. The epidemic of mercury poisoning in Japan has clearly demonstrated that chronic exposure to methylmercury (MeHg) results in serious neurological damage to the cerebral and cerebellar cortex, leading to the dysfunction of the central nervous system (CNS), especially in infants exposed to MeHg in utero. The occurrences of poisoning have caused a wide public concern regarding the health risk emanating from MeHg exposure; particularly those eating large amounts of fish may experience the low-level and long-term exposure. There is growing evidence that MeHg at environmentally relevant concentrations can affect the health of biota in the ecosystem. Although extensive in vivo and in vitro studies have demonstrated that the disruption of redox homeostasis and microtube assembly is mainly responsible for mercurial toxicity leading to adverse health outcomes, it is still unclear whether we could quantitively determine the occurrence of interaction between mercurial and thiols and/or selenols groups of proteins linked directly to outcomes, especially at very low levels of exposure. Furthermore, intracellular calcium homeostasis, cytoskeleton, mitochondrial function, oxidative stress, neurotransmitter release, and DNA methylation may be the targets of mercury compounds; however, the primary targets associated with the adverse outcomes remain to be elucidated. Considering these knowledge gaps, in this article, we conducted a comprehensive review of mercurial toxicity, focusing mainly on the mechanism, and genes/proteins expression. We speculated that comprehensive analyses of transcriptomics, proteomics, and metabolomics could enhance interpretation of "omics" profiles, which may reveal specific biomarkers obviously correlated with specific pathways that mediate selective neurotoxicity.
Collapse
Affiliation(s)
- Bolun Kang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Jinghan Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
| | - Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012 Beijing, China.
| |
Collapse
|
4
|
Porru S, Esplugues A, Llop S, Delgado-Saborit JM. The effects of heavy metal exposure on brain and gut microbiota: A systematic review of animal studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 348:123732. [PMID: 38462196 DOI: 10.1016/j.envpol.2024.123732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/12/2024] [Accepted: 03/05/2024] [Indexed: 03/12/2024]
Abstract
The gut-brain axis is a crucial interface between the central nervous system and the gut microbiota. Recent evidence shows that exposure to environmental contaminants, such as heavy metals, can cause dysbiosis in gut microbiota, which may affect the gut-brain communication, impacting aspects of brain function and behavior. This systematic review of the literature aims to evaluate whether deleterious effects on brain function due to heavy metal exposure could be mediated by changes in the gut microbiota profile. Animal studies involving exposure to heavy metals and a comparison with a control group that evaluated neuropsychological outcomes and/or molecular outcomes along with the analysis of microbiota composition were reviewed. The authors independently assessed studies for inclusion, extracted data and assessed risk of bias using the protocol of Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) for preclinical studies. A search in 3 databases yielded 16 eligible studies focused on lead (n = 10), cadmium (n = 1), mercury (n = 3), manganese (n = 1), and combined exposure of lead and manganese (n = 1). The animal species were rats (n = 7), mice (n = 4), zebrafish (n = 3), carp (n = 1) and fruit fly (n = 1). Heavy metals were found to adversely affect cognitive function, behavior, and neuronal morphology. Moreover, heavy metal exposure was associated with changes in the abundance of specific bacterial phyla, such as Firmicutes and Proteobacteria, which play crucial roles in gut health. In some studies, these alterations were correlated with learning and memory impairments and mood disorders. The interplay of heavy metals, gut microbiota, and brain suggests that heavy metals can induce direct brain alterations and indirect effects through the microbiota, contributing to neurotoxicity and the development of neuropsychological disorders. However, the small number of papers under review makes it difficult to draw definitive conclusions. Further research is warranted to unravel the underlying mechanisms and evaluate the translational implications for human health.
Collapse
Affiliation(s)
- Simona Porru
- Department of Medicine, Faculty of Health Sciences. Universitat Jaume I, Avenida de Vicent Sos Baynat s/n, 12071, Castellón de la Plana, Spain
| | - Ana Esplugues
- Faculty of Nursing and Podiatry, Universitat de València, C/Menendez Pelayo S/n, 46010, València, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de Valencia, Av. Catalunya 21, 46020, València, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Av. Monforte de Lemos, 3-5. Pabellón 11, 28029, Madrid, Spain
| | - Sabrina Llop
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de Valencia, Av. Catalunya 21, 46020, València, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Av. Monforte de Lemos, 3-5. Pabellón 11, 28029, Madrid, Spain
| | - Juana María Delgado-Saborit
- Department of Medicine, Faculty of Health Sciences. Universitat Jaume I, Avenida de Vicent Sos Baynat s/n, 12071, Castellón de la Plana, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de Valencia, Av. Catalunya 21, 46020, València, Spain.
| |
Collapse
|
5
|
Leal-Nazaré CG, Arrifano GP, Lopes-Araújo A, Santos-Sacramento L, Barthelemy JL, Soares-Silva I, Crespo-Lopez ME, Augusto-Oliveira M. Methylmercury neurotoxicity: Beyond the neurocentric view. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 920:170939. [PMID: 38365040 DOI: 10.1016/j.scitotenv.2024.170939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Mercury is a highly toxic metal widely used in human activities worldwide, therefore considered a global public health problem. Many cases of mercury intoxication have occurred in history and represent a huge challenge nowadays. Of particular importance is its methylated form, methylmercury (MeHg). This mercurial species induces damage to several organs in the human body, especially to the central nervous system. Neurological impairments such as executive, memory, motor and visual deficits are associated with MeHg neurotoxicity. Molecular mechanisms involved in MeHg-induced neurotoxicity include excitotoxicity due to glutamatergic imbalance, disturbance in calcium homeostasis and oxidative balance, failure in synaptic support, and inflammatory response. Although neurons are largely affected by MeHg intoxication, they only represent half of the brain cells. Glial cells represent roughly 50 % of the brain cells and are key elements in the functioning of the central nervous system. Particularly, astrocytes and microglia are deeply involved in MeHg-induced neurotoxicity, resulting in distinct neurological outcomes depending on the context. In this review, we discuss the main findings on astroglial and microglial involvement as mediators of neuroprotective and neurotoxic responses to MeHg intoxication. The literature shows that these responses depend on chemical and morphophysiological features, thus, we present some insights for future investigations, considering the particularities of the context, including time and dose of exposure, brain region, and species of study.
Collapse
Affiliation(s)
- Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Amanda Lopes-Araújo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Jean Ludger Barthelemy
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Isabela Soares-Silva
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| | - Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil.
| |
Collapse
|
6
|
Xu Y, Nie J, Lu C, Hu C, Chen Y, Ma Y, Huang Y, Lu L. Effects and mechanisms of bisphenols exposure on neurodegenerative diseases risk: A systemic review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170670. [PMID: 38325473 DOI: 10.1016/j.scitotenv.2024.170670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Environmental bisphenols (BPs) pose a global threat to human health because of their extensive use as additives in plastic products. BP residues are increasing in various environmental media (i.e., water, soil, and indoor dust) and biological and human samples (i.e., serum and brain). Both epidemiological and animal studies have determined an association between exposure to BPs and an increased risk of neurodegenerative diseases (e.g., Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis), including cognitive abnormalities and behavioral disturbances. Hence, understanding the biological responses to different BPs is essential for prevention, and treatment. This study provides an overview of the underlying pathogenic molecular mechanisms as a valuable basis for understanding neurodegenerative disease responses to BPs, including accumulation of misfolded proteins, reduction of tyrosine hydroxylase and dopamine, abnormal hormone signaling, neuronal death, oxidative stress, calcium homeostasis, and inflammation. These findings provide new insights into the neurotoxic potential of BPs and ultimately contribute to a comprehensive health risk evaluation.
Collapse
Affiliation(s)
- Yeqing Xu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jun Nie
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chenghao Lu
- College of Mathematics and Computer Science, Zhejiang A & F University, Hangzhou 311300, China
| | - Chao Hu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yunlu Chen
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ying Ma
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuru Huang
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Liping Lu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
7
|
Mallamaci R, Barbarossa A, Carocci A, Meleleo D. Evaluation of the Potential Protective Effect of Ellagic Acid against Heavy Metal (Cadmium, Mercury, and Lead) Toxicity in SH-SY5Y Neuroblastoma Cells. Foods 2024; 13:419. [PMID: 38338554 PMCID: PMC10855963 DOI: 10.3390/foods13030419] [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: 12/22/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Ellagic acid (EA), a polyphenolic constituent of plant origin, has been thoroughly investigated for its hypothesised pharmacological properties among which antioxidant and neuroprotective activities are included. The present study was designed to explore whether EA could attenuate heavy metal (cadmium, mercury, and lead)-induced neurotoxicity in SH-SY5Y cells, which were utilized as a model system for brain cells. MTT and LDH assays were performed to examine the viability of the SH-SY5Y cells after exposure to Cd, Hg, and Pb (either individually or in combination with EA) as well as the effects of necrotic cell death, respectively. Furthermore, 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA), a cell-based assay, was performed to determine whether EA could protect SH-SY5Y from heavy metal-induced oxidative stress. Results allowed us to assess the capability of EA to enhance the number of viable SH-SY5Y cells after exposure to heavy metal toxicity. Pre-treatment with EA showed a considerable, concentration-dependent, cytoprotective effect, particularly against Cd2+-induced toxicity. This effect was confirmed through the reduction of LDH release after the simultaneous cell treatment with Cd2+ and EA compared with Cd2+-treated cells. Furthermore, a significant, concentration-dependent decrease in reactive oxygen species (ROS) production, induced by H2O2 or heavy metals, was observed in the same model. Overall, the obtained results provide further insight into the protective role of EA against heavy metal-induced neurotoxicity and oxidative stress, thus indicating the potential beneficial effects of the consumption of EA-rich foods. However, to confirm its effects, well-designed human randomized controlled trials are needed to fill the existing gap between experimental and clinical research.
Collapse
Affiliation(s)
- Rosanna Mallamaci
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Alexia Barbarossa
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Alessia Carocci
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Daniela Meleleo
- Department of Science of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
8
|
Barbosa NV, Aschner M, Tinkov AA, Farina M, da Rocha JBT. Should ebselen be considered for the treatment of mercury intoxication? A minireview. Toxicol Mech Methods 2024; 34:1-12. [PMID: 37731353 PMCID: PMC10841883 DOI: 10.1080/15376516.2023.2258958] [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: 04/13/2023] [Accepted: 09/09/2023] [Indexed: 09/22/2023]
Abstract
Mercury is a ubiquitous environmental contaminant and can be found in inorganic (Hg0, Hg+ and Hg2+) and organic forms (chiefly CH3Hg+ or MeHg+). The main route of human, mammals and bird exposure occurs via predatory fish ingestion. Occupational exposure to Hg0 (and Hg2+) can also occur; furthermore, in gold mining areas the exposure to inorganic Hg can also be high. The toxicity of electrophilic forms of Hg (E+Hg) is mediated by disruption of thiol (-SH)- or selenol (-SeH)-containing proteins. The therapeutic approaches to treat methylmercury (MeHg+), Hg0 and Hg2+ are limited. Here we discuss the potential use of ebselen as a potential therapeutic agent to lower the body burden of Hg in man. Ebselen is a safe drug for humans and has been tested in clinical trials (for instance, brain ischemia, noise-induce hearing loss, diabetes complications, bipolar disorders) at doses varying from 400 to 3600 mg per day. Two clinical trials with ebselen in moderate and severe COVID are also approved. Ebselen can be metabolized to an intermediate with -SeH (selenol) functional group, which has a greater affinity to electrophilic Hg (E+Hg) forms than the available thiol-containing therapeutic agents. Accordingly, as observed in vitro and rodent models in vivo, Ebselen exhibited protective effects against MeHg+, indicating its potential as a therapeutic agent to treat MeHg+ overexposure. The combined use of ebselen with thiol-containing molecules (e.g. N-acetylcysteine and enaramide)) is also commented, because they can have synergistic protective effects against MeHg+.
Collapse
Affiliation(s)
- Nilda V. Barbosa
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexey A. Tinkov
- Yaroslavl State University, Yaroslavl, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - João Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
- Departamento de Bioquímica, Instituto Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
9
|
Bottini CLJ, MacDougall-Shackleton SA. Methylmercury effects on avian brains. Neurotoxicology 2023; 96:140-153. [PMID: 37059311 DOI: 10.1016/j.neuro.2023.04.004] [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: 01/22/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Methylmercury (MeHg) is a concerning contaminant due to its ubiquity and harmful effects on organisms. Although birds are important models in the neurobiology of vocal learning and adult neuroplasticity, the neurotoxic effects of MeHg are less understood in birds than mammals. We surveyed the literature on MeHg effects on biochemical changes in the avian brain. Publication rates of papers related to neurology and/or birds and/or MeHg increased with time and can be linked with historical events, regulations, and increased understanding of MeHg cycling in the environment. However, publications on MeHg effects on the avian brain remain relatively low across time. The neural effects measured to evaluate MeHg neurotoxicity in birds changed with time and researcher interest. The measures most consistently affected by MeHg exposure in birds were markers of oxidative stress. NMDA, acetylcholinesterase, and Purkinje cells also seem sensitive to some extent. MeHg exposure has the potential to affect most neurotransmitter systems but more studies are needed for validation in birds. We also review the main mechanisms of MeHg-induced neurotoxicity in mammals and compare it to what is known in birds. The literature on MeHg effects on the avian brain is limited, preventing full construction of an adverse outcome pathway. We identify research gaps for taxonomic groups such as songbirds, and age- and life-stage groups such as immature fledgling stage and adult non-reproductive life stage. In addition, results are often inconsistent between experimental and field studies. We conclude that future neurotoxicological studies of MeHg impacts on birds need to better connect the numerous aspects of exposure from molecular physiological effects to behavioural outcomes that would be ecologically or biologically relevant for birds, especially under challenging conditions.
Collapse
Affiliation(s)
- Claire L J Bottini
- University of Western Ontario, Department of Biology, 1151 Richmond St., London Ontario, N6A 5B7; Advanced Facility for Avian Research, University of Western Ontario, London, Ontario, Canada.
| | - Scott A MacDougall-Shackleton
- Advanced Facility for Avian Research, University of Western Ontario, London, Ontario, Canada; University of Western Ontario, Department of Psychology, 1151 Richmond St., London Ontario, N6A 5C2
| |
Collapse
|
10
|
Abdelhamid MS, El Bohi KM, Sherif MH, Abdelhamid MS, Abdel-Daim MM, Elewa YHA, Metwally MMM, Albadrani GM, Najda A, El Abdel-Hamid S, Abu-Zeid EH. Apitoxin alleviates methyl mercury-induced peripheral neurotoxicity in male rats by regulating dorsal root ganglia neuronal degeneration and oxidative stress. Biomed Pharmacother 2023; 161:114521. [PMID: 36921536 DOI: 10.1016/j.biopha.2023.114521] [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: 01/06/2023] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Methylmercury (MeHg) toxicity is associated with extensive neuronal degeneration of dorsal root ganglia (DRG). This study aimed to assess the ameliorative effect of bee venom (BV) on methyl mercury chloride (MeHgCl)-induced peripheral neurotoxicity using DRGs in rats. Forty-eight adult male Sprague Dawley rats were allocated into four equal groups: G I: control (gavaged MilliQ water 1 ml/rat), G II: subcutaneously injected with BV (0.5 mg/kg b.wt), G III: gavaged MeHgCl (6.7 mg/kg b.wt), and G IV: received MeHgCl+BV. Dosing was done five times/week for 2 weeks. Ataxic behavior and visual impairments were significantly increased, whereas the movement behavior and motility gait were suppressed in the MeHgCl group. MeHgCl significantly decreased total antioxidant capacity (TAC) in DRG and significantly decreased the serum levels of glutathione (GSH), catalase (CAT), and superoxide dismutase (SOD). Tumor necrosis factor-alpha (TNF-α) and interleukin 1β (IL-1β) levels were significantly elevated, whereas interleukin 10 (IL-10) levels were significantly decreased in the MeHgCl group compared with the control group. DRGs of the MeHgCl-exposed rats showed pyknotic shrunken neurons with perineural vacuolations, demyelination of nerve axons, and proliferation of the satellite cells. MeHgCl significantly induced a higher positive index ratio of Iba-1, SOX10, neurofilament, pan-neuron, and vimentin immunostaining in the DRG. BV administration significantly mitigated the MeHgCl-induced alterations in oxidative stress-related indices. BV modified the immunostaining of Iba-1, SOX10, neurofilament, pan-neuron, and vimentin-positive index ratio in the DRG of the MeHgCl group. Our findings acknowledged that BV could enhance in vivo neuroprotective effects against MeHgCl-induced DRGs damage in male rats.
Collapse
Affiliation(s)
- Moustafa S Abdelhamid
- Biochemistry division, Chemistry Department, Faculty of Science, Zagazig University, 44511, Egypt
| | - Khlood M El Bohi
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt
| | - Mohamed H Sherif
- Biochemistry division, Chemistry Department, Faculty of Science, Zagazig University, 44511, Egypt
| | - Manar S Abdelhamid
- Biochemistry division, Chemistry Department, Faculty of Science, Zagazig University, 44511, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Yaser H A Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt; Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, B.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Agnieszka Najda
- Department of Vegetable Crops and Medicinal Plants University of Life Sciences in Lublin, 50 A Doświadczalna Street, 20-280 Lublin, Poland.
| | - Shereen El Abdel-Hamid
- Department of Behavior and Management of Animal, Poultry and Aquatics, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt
| | - Ehsan H Abu-Zeid
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt.
| |
Collapse
|
11
|
Liu BY, Chen IW, Chen PW, Chen TY, Hwang DF. Risk assessment of methylmercury and species identification in shark meats ingested by Taiwan children. Food Control 2023. [DOI: 10.1016/j.foodcont.2022.109461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
12
|
Ke T, Santamaria A, Barbosa F, Rocha JBT, Skalny AV, Tinkov AA, Bowman AB, Aschner M. Developmental Methylmercury Exposure Induced and Age-Dependent Glutamatergic Neurotoxicity in Caenorhabditis elegans. Neurochem Res 2023; 48:920-928. [PMID: 36385214 DOI: 10.1007/s11064-022-03816-5] [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: 05/07/2022] [Revised: 10/12/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Developmental methylmercury (MeHg) exposures cause latent neurotoxic effects in adults; however, the mechanisms underlying the latent neurotoxicity are not fully understood. In the current study, we used C. elegans as an animal model to investigate the latent neurotoxic effects of developmental MeHg exposures on glutamatergic neurons. The young larvae stage 1 worms were exposed to MeHg (0.05 ~ 5 µM) for 48 h. The morphological and behavioral endpoints of glutamatergic neurons were compared when worms reached to adult stages including the young adult stage (day 1 adult) and the old adult stage (day 10 adult). Here, we showed that C. elegans glutamatergic neurons were morphologically intact following low or medium MeHg exposures (0.05 ~ 0.5 µM). The morphological damage of glutamatergic neurons appeared to be pronounced in day 10 adults developmentally exposed to 5 µM MeHg. Behavioral assays also showed an age-dependent latent effect of MeHg. In the nose touch response assay, only day 10 adult worms exhibited a functional decline following prior 5 µM MeHg exposure. Moreover, the disruption of NaCl memory appeared only in day 1 adults following MeHg exposures but not in day 10 adults. The expression of C. elegans homologs of mammalian vesicular glutamate transporter (eat-4) was repressed in day 1 adults, while the glutamate receptor homolog (glr-1) was upregulated in day 10 adults with 5 µM MeHg. In the comparison of age-dependent changes in the insulin-like pathway (daf-2/age-1/daf-16) following MeHg exposures, we showed that the daf-2/age-1/daf-16 pathway was mobilized in day 1 adults but repressed in day 10 adults. Collectively, our data supports a conclusion that MeHg-induced glutamatergic neurotoxicity exhibits an age-dependent pattern, possibly related to the prominent changes in age-dependent modulation in the glutamatergic neurotransmission and metabolic pathways.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico
| | - Fernando Barbosa
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-900, Brazil
| | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, 97105900, Brazil
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Yaroslavl State University, Yaroslavl, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907-2051, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- , Forchheimer Building, Room 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
13
|
Puty B, Bittencourt LO, Plaça JR, de Oliveira EHC, Lima RR. Astrocyte-Like Cells Transcriptome Changes After Exposure to a Low and Non-cytotoxic MeHg Concentration. Biol Trace Elem Res 2023; 201:1151-1162. [PMID: 35378667 DOI: 10.1007/s12011-022-03225-3] [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: 02/12/2022] [Accepted: 03/29/2022] [Indexed: 02/07/2023]
Abstract
The central nervous system is the main target of MeHg toxicity and glial cells are the first line of defense; however, their true role remains unclear. This study aimed to identify the global map of human glial-like (U87) cells transcriptome after exposure to a non-toxic and non-lethal MeHg concentration and to investigate the related molecular changes. U87 cells were exposed upon 0.1, 0.5, and 1 µM MeHg for 4 and 24 h. Although no changes were observed in the percentage of viable cells, the metabolic viability was significantly decreased after exposure to 1 µM MeHg for 24 h; thus, the non-toxic concentration of 0.1 µM MeHg was chosen to perform microarray analysis. Significant changes in U87 cells transcriptome were observed only after 24 h. The expression of 392 genes was down regulated while 431 genes were up-regulated. Gene ontology showed alterations in biological processes (75%), cellular components (21%), and molecular functions (4%). The main pathways showed by KEGG and Reactome were cell cycle regulation and Rho GTPase signaling. The complex mechanism of U87 cells response against MeHg exposure indicates that even a low and non-toxic concentration is able to alter the gene expression profile.
Collapse
Affiliation(s)
- Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
- Laboratory of Tissue Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
| | - Jéssica Rodrigues Plaça
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | | | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil.
| |
Collapse
|
14
|
Zhang X, Hao H, Ma K, Pang H, Li X, Tian T, Hou S, Ning X, Wu H, Hou Q, Li M, Sun Y, Song X, Jin M. The role and mechanism of unfolded protein response signaling pathway in methylmercury-induced apoptosis of mouse spermatocytes germ cell-2 cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:472-482. [PMID: 36330985 DOI: 10.1002/tox.23684] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/09/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
The study aimed to explore the role and mechanism of unfolded protein response (UPR) in methylmercury (MeHg)-induced Mouse Spermatocytes (GC-2spd[ts]) apoptosis. Methods such as MTT, flow cytometry, and Western Blot were used to evaluate the cell viability, membrane potential (MMP), reactive oxygen species (ROS), calcium ion (Ca2+ ), rate of cell apoptosis, and the expression of apoptosis-related and UPR-related protein. The results showed that with the increase of MeHg concentration, cell viability and MMP decreased, ROS, Ca2+ , rate of cell apoptosis, and the expression of apoptosis-related protein and UPR-related protein increased. To further explore the effect of ROS-induced oxidative damage on it, the ROS inhibitor N-acetyl-L-cysteine (NAC) was used. The effects of MeHg on germ cell (GC-2) cells were partially inhibited after NAC pretreatment. Our present study proved that MeHg might induce cell apoptosis by activating the UPR signaling pathway in GC-2 cells and affect normal reproductive function.
Collapse
Affiliation(s)
- Xiayu Zhang
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Huifang Hao
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Kai Ma
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Huan Pang
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Xinyue Li
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Tiantian Tian
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Shanshan Hou
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Xiaofan Ning
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Hao Wu
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Qiaohong Hou
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Meng Li
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Yunxiang Sun
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Xiuling Song
- School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Minghua Jin
- School of Public Health, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
15
|
Nguyen HD. Interactions between heavy metals and sleep duration among pre-and postmenopausal women: A current approach to molecular mechanisms involved. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120607. [PMID: 36347409 DOI: 10.1016/j.envpol.2022.120607] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
The effects of heavy metals (cadmium, lead, and mercury) and their mixtures on sleep duration in pre-and postmenopausal women, particularly the molecular mechanisms, remain unknown. Here, we assessed the interaction between heavy metals and sleep duration among pre-and postmenopausal women (n = 1134). Furthermore, molecular mechanisms related to sleep disorders induced by studied heavy metals were further analyzed to support the previous findings. We found that serum lead levels were positively related to weekend and weekday sleep duration in premenopausal women. There were interactions between serum lead and mercury and menopausal status for sleep duration. Serum lead and mercury levels were shown to be inversely related to sleep duration in postmenopausal women. Despite the lack of statistically significant associations between mixed heavy metals and sleep duration, there were increasing trends in premenopausal women's sleeping patterns and decreasing trends in postmenopausal women's sleeping patterns. In silico analysis, IL1B, hsa-21-5p, hsa-887-3p, hsa-877-3p, and NR4A1 were identified as the most relevant genes, miRNAs, and transcription factors linked with sleep disorders induced by combined heavy metals (cadmium, lead, and mercury). Furthermore, "type 1 melanocortin receptor binding," "endocrine hormone secretion," "interleukin-1 receptor antagonist activity," "altered melanocortin system," and "sleep wake disorders" were identified as the predominant molecular mechanisms involved in the pathophysiology of sleep disorders induced by the studied heavy metals. Cut off point values and miRNA sponge templates for heavy metal exposure levels relevant to sleep disorders in pre- and postmenopausal women were reported. Future research is needed to verify our findings and provide insight into the molecular processes of long-term mixed heavy metal exposure in various populations, such as children and the elderly.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, jeonnam, 57922, Republic of Korea.
| |
Collapse
|
16
|
Maria B, Maria MC, Antonio B, Simona M, Rosaria A, Andrea S, Giulia M, Marianna DC, Mario S. Chemical and biochemical responses to sub-lethal doses of mercury and cadmium in gilthead seabream (Sparus aurata). CHEMOSPHERE 2022; 307:135822. [PMID: 35963385 DOI: 10.1016/j.chemosphere.2022.135822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Specimens of Sparus aurata were exposed to sub-lethal concentrations of Hg and Cd for 25 days and the levels of both metals were investigated in organs and tissues. Bioaccumulation of Hg decreased as follow: gills > kidney > liver > skin > muscle, while the order of Cd bioaccumulation was: liver > kidney > gills > skin > muscle. Immediately after exposure, both metals showed the highest bioaccumulation in gills and skin indicating that these organs are reliable targets for biomonitoring studies after short term exposure. Metals introduction caused a significant time-dependent concentrations increase in kidney and liver, while in the muscle a significant increase of Hg was recorded only at the end of the experimentation. The effects of exposure were also investigated, at biochemical level, in the liver, which represents the main target of xenobiotics biotransformation and metabolism in fish. Exposed fishes exhibited a reduction of total lipid level, a decrease of polyunsaturated fatty acids (PUFA), together with a MDA increase. This suggests a direct effect of contaminants on oxidative stress induction that, through the MDA increase, altered the membrane fatty acids composition decreasing the PUFA content. As it regards molecular markers related to oxidative stress and lipid metanolism, a significant increase of Nrf2, Hif-1α and Ampk and a decrease of Fas were observed after exposure to both metals, while an Nf-kB increase was recorded in specimens exposed to Hg, docuemnting a correlation with oxidative stress and consequent metabolism adaptation. Finally, these results suggest the possibility to adopt these biomarkers to explore fish metabolic responses to environmental pollution.
Collapse
Affiliation(s)
- Bonsignore Maria
- National Research Council of Italy - Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), via del Mare, 3 Campobello di Mazara, Italy
| | - Messina Concetta Maria
- National Research Council of Italy - Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), via del Mare, 3 Campobello di Mazara, Italy; University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Bellante Antonio
- National Research Council of Italy - Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), via del Mare, 3 Campobello di Mazara, Italy.
| | - Manuguerra Simona
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Arena Rosaria
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Santulli Andrea
- University of Palermo, Dept. of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Maricchiolo Giulia
- National Research Council of Italy, Institute of Biological Resources and Marine Biotechnologies (IRBIM-CNR), Spianata S. Raineri 86, 98122, Messina, Italy
| | - Del Core Marianna
- National Research Council of Italy - Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), via del Mare, 3 Campobello di Mazara, Italy
| | - Sprovieri Mario
- National Research Council of Italy - Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), via del Mare, 3 Campobello di Mazara, Italy
| |
Collapse
|
17
|
Bjørklund G, Antonyak H, Polishchuk A, Semenova Y, Lesiv M, Lysiuk R, Peana M. Effect of methylmercury on fetal neurobehavioral development: an overview of the possible mechanisms of toxicity and the neuroprotective effect of phytochemicals. Arch Toxicol 2022; 96:3175-3199. [PMID: 36063174 DOI: 10.1007/s00204-022-03366-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022]
Abstract
Methylmercury (MeHg) is a global environmental pollutant with neurotoxic effects. Exposure to MeHg via consumption of seafood and fish can severely impact fetal neurobehavioral development even when MeHg levels in maternal blood are as low as about 5 μg/L, which the mother tolerates well. Persistent motor dysfunctions and cognitive deficits may result from trans-placental exposure. The present review summarizes current knowledge on the mechanisms of MeHg toxicity during the period of nervous system development. Although cerebellar Purkinje cells are MeHg targets, the actions of MeHg on thiol components in the neuronal cytoskeleton as well as on mitochondrial enzymes and induction of disturbances of glutamate signaling can impair extra-cerebellar functions, also at levels well tolerated by adult individuals. Numerous herbal substances possess neuroprotective effects, predominantly represented by natural polyphenolic molecules that might be utilized to develop natural drugs to alleviate neurotoxicity symptoms caused by MeHg or other Hg compounds.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610, Mo i Rana, Norway.
| | | | | | | | - Marta Lesiv
- Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Roman Lysiuk
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Massimiliano Peana
- Department of Chemical, Physics, Mathematics and Natural Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
18
|
Reactive oxygen species, the trident of Neptune in the hands of hecate; role in different diseases, signaling pathways, and detection methods. Arch Biochem Biophys 2022; 728:109357. [DOI: 10.1016/j.abb.2022.109357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 12/22/2022]
|
19
|
Lozano M, Yousefi P, Broberg K, Soler-Blasco R, Miyashita C, Pesce G, Kim WJ, Rahman M, Bakulski KM, Haug LS, Ikeda-Araki A, Huel G, Park J, Relton C, Vrijheid M, Rifas-Shiman S, Oken E, Dou JF, Kishi R, Gutzkow KB, Annesi-Maesano I, Won S, Hivert MF, Fallin MD, Vafeiadi M, Ballester F, Bustamante M, Llop S. DNA methylation changes associated with prenatal mercury exposure: A meta-analysis of prospective cohort studies from PACE consortium. ENVIRONMENTAL RESEARCH 2022; 204:112093. [PMID: 34562483 PMCID: PMC10879652 DOI: 10.1016/j.envres.2021.112093] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 06/13/2023]
Abstract
Mercury (Hg) is a ubiquitous heavy metal that originates from both natural and anthropogenic sources and is transformed in the environment to its most toxicant form, methylmercury (MeHg). Recent studies suggest that MeHg exposure can alter epigenetic modifications during embryogenesis. In this study, we examined associations between prenatal MeHg exposure and levels of cord blood DNA methylation (DNAm) by meta-analysis in up to seven independent studies (n = 1462) as well as persistence of those relationships in blood from 7 to 8 year-old children (n = 794). In cord blood, we found limited evidence of differential DNAm at cg24184221 in MED31 (β = 2.28 × 10-4, p-value = 5.87 × 10-5) in relation to prenatal MeHg exposure. In child blood, we identified differential DNAm at cg15288800 (β = 0.004, p-value = 4.97 × 10-5), also located in MED31. This repeated link to MED31, a gene involved in lipid metabolism and RNA Polymerase II transcription function, may suggest a DNAm perturbation related to MeHg exposure that persists into early childhood. Further, we found evidence for association between prenatal MeHg exposure and child blood DNAm levels at two additional CpGs: cg12204245 (β = 0.002, p-value = 4.81 × 10-7) in GRK1 and cg02212000 (β = -0.001, p-value = 8.13 × 10-7) in GGH. Prenatal MeHg exposure was associated with DNAm modifications that may influence health outcomes, such as cognitive or anthropometric development, in different populations.
Collapse
Affiliation(s)
- Manuel Lozano
- Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine Department, Universitat de València, Valencia, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain.
| | - Paul Yousefi
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Karin Broberg
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Raquel Soler-Blasco
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
| | - Chihiro Miyashita
- Center for Environmental and Health Sciences, Hokkaido University, Hokkaido, Japan
| | - Giancarlo Pesce
- INSERM UMR1018, Université Paris-Saclay, UVSQ, Centre for Epidemiology and Public Health (CESP), Villejuif, France
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University, Chuncheon, South Korea
| | - Mohammad Rahman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, United States
| | - Kelly M Bakulski
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Line S Haug
- Norwegian Institute of Public Health, Division of Climate and Environment, Oslo, Norway
| | - Atsuko Ikeda-Araki
- Center for Environmental and Health Sciences, Hokkaido University, Hokkaido, Japan; Faculty of Health Sciences, Hokkaido University, Hokkaido, Japan
| | - Guy Huel
- INSERM UMR1018, Université Paris-Saclay, UVSQ, Centre for Epidemiology and Public Health (CESP), Villejuif, France
| | - Jaehyun Park
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Caroline Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Martine Vrijheid
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain; ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Sheryl Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, United States
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, United States
| | - John F Dou
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Reiko Kishi
- Center for Environmental and Health Sciences, Hokkaido University, Hokkaido, Japan
| | - Kristine B Gutzkow
- Norwegian Institute of Public Health, Division of Climate and Environment, Oslo, Norway
| | - Isabella Annesi-Maesano
- INSERM UMR1302, Montpellier University, Insitut Desbrest d'Épidémiologie et de Santé Publique (IDESP), Montpellier, France
| | - Sungho Won
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, South Korea; Department of Public Health Sciences, Seoul National University, Seoul, South Korea
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, United States; Diabetes Unit, Massachusetts General Hospital, Boston, MA, United States
| | - M Daniele Fallin
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Marina Vafeiadi
- Department of Social Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Ferran Ballester
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain; School of Nursing, Universitat de València, Valencia, Spain
| | - Mariona Bustamante
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain; ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Sabrina Llop
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
20
|
Pereira PS, Costa AR, de Oliveira TJS, Oliveira CVB, de Lima MDCA, de Oliveira JF, Kim B, Coutinho HDM, Duarte AE, Kamdem JP, da Silva TG. Neurolocomotor Behavior and Oxidative Stress Markers of Thiazole and Thiazolidinedione Derivatives against Nauphoeta cinerea. Antioxidants (Basel) 2022; 11:antiox11020420. [PMID: 35204302 PMCID: PMC8869355 DOI: 10.3390/antiox11020420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
Thiazolidine compounds NJ20 {(E)-2-(2-(5-bromo-2-methoxybenzylidene)hydrazinyl)-4-(4-nitrophenyl)thiazole} and NW05 [(2-(benzo (d) (1,3) dioxol-4-ylmethylene)-N-(4-bromophenyl)-thiosemicarbazone] potentiated the effect of norfloxacin in resistant bacteria; however, there are no reports on their effects on Nauphoeta cinerea in the literature. The objective of this work was to evaluate the behavioral effects and oxidative markers of NW05 and NJ20 in lobster cockroach N. cinerea. To evaluate the behavioral study, a video tracking software was used to evaluate the locomotor points and the exploratory profile of cockroaches in the horizontal and vertical regions of a new environment. The total concentration of thiol and reduced glutathione (GSH), substances reactive to thiobarbituric acid (TBARS), free iron (II) content and mitochondrial viability were determined. The antioxidant potential was evaluated by the DPPH method. Both substances induced changes in the behavior of cockroaches, showing a significant reduction in the total distance covered and in the speed. In the cell viability test (MTT), there was a significant reduction for NJ20 (1 mM). NJ20 caused a significant increase in total levels of thiol and non-protein thiol (NPSH), although it also slightly increased the content of malondialdehyde (MDA). Both compounds (NW05 and NJ20) caused a significant reduction in the content of free iron at a concentration of 10 mM. In conclusion, the compound NJ20 caused moderate neurotoxicity (1 mM), but had good antioxidant action, while NW05 did not show toxicity or significant antioxidant activity in the model organism tested. It is desirable to carry out complementary tests related to the antioxidant prospection of these same compounds, evaluating them at different concentrations.
Collapse
Affiliation(s)
- Pedro Silvino Pereira
- Department of Antibiotics, Federal University of Pernambuco (UFPE), Prof. Artur de Sa Avenue, University City, Recife 54740-520, PE, Brazil; (P.S.P.); (M.d.C.A.d.L.); (T.G.d.S.)
| | - Adrielle Rodrigues Costa
- Department of Biological Sciences, Regional University of Cariri (URCA), 1161 Cel. Antonio Luiz Avenue, Pimenta, Crato 63105-000, CE, Brazil; (A.R.C.); (T.J.S.d.O.); (C.V.B.O.); (A.E.D.); (J.P.K.)
| | - Thalyta Julyanne Silva de Oliveira
- Department of Biological Sciences, Regional University of Cariri (URCA), 1161 Cel. Antonio Luiz Avenue, Pimenta, Crato 63105-000, CE, Brazil; (A.R.C.); (T.J.S.d.O.); (C.V.B.O.); (A.E.D.); (J.P.K.)
| | - Carlos Vinícius Barros Oliveira
- Department of Biological Sciences, Regional University of Cariri (URCA), 1161 Cel. Antonio Luiz Avenue, Pimenta, Crato 63105-000, CE, Brazil; (A.R.C.); (T.J.S.d.O.); (C.V.B.O.); (A.E.D.); (J.P.K.)
| | - Maria do Carmo Alves de Lima
- Department of Antibiotics, Federal University of Pernambuco (UFPE), Prof. Artur de Sa Avenue, University City, Recife 54740-520, PE, Brazil; (P.S.P.); (M.d.C.A.d.L.); (T.G.d.S.)
| | - Jamerson Ferreira de Oliveira
- Institute of Health Sciences, Auroras Campus, University of International Integration of Afro-Brazilian Lusophony (UNILAB), 3 Abolition Avenue, Downtown, Redenção 62790-000, CE, Brazil;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (B.K.); (H.D.M.C.)
| | - Henrique D. M. Coutinho
- Department of Biological Chemistry, Regional University of Cariri (URCA), 1161 Cel. Antonio Luiz Avenue, Pimenta, Crato 63105-000, CE, Brazil
- Correspondence: (B.K.); (H.D.M.C.)
| | - Antonia Eliene Duarte
- Department of Biological Sciences, Regional University of Cariri (URCA), 1161 Cel. Antonio Luiz Avenue, Pimenta, Crato 63105-000, CE, Brazil; (A.R.C.); (T.J.S.d.O.); (C.V.B.O.); (A.E.D.); (J.P.K.)
| | - Jean Paul Kamdem
- Department of Biological Sciences, Regional University of Cariri (URCA), 1161 Cel. Antonio Luiz Avenue, Pimenta, Crato 63105-000, CE, Brazil; (A.R.C.); (T.J.S.d.O.); (C.V.B.O.); (A.E.D.); (J.P.K.)
| | - Teresinha Gonçalves da Silva
- Department of Antibiotics, Federal University of Pernambuco (UFPE), Prof. Artur de Sa Avenue, University City, Recife 54740-520, PE, Brazil; (P.S.P.); (M.d.C.A.d.L.); (T.G.d.S.)
| |
Collapse
|
21
|
Ferrer B, Suresh H, Tinkov AA, Santamaria A, Rocha JB, Skalny AV, Bowman AB, Aschner M. Ghrelin attenuates methylmercury-induced oxidative stress in neuronal cells. Mol Neurobiol 2022; 59:2098-2115. [PMID: 35040042 DOI: 10.1007/s12035-022-02726-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022]
Abstract
Methylmercury (MeHg) is a global pollutant, which can cause damage to the central nervous system at both high-acute and chronic-low exposures, especially in vulnerable populations, such as children and pregnant women. Nowadays, acute-high poisoning is rare. However, chronic exposure to low MeHg concentrations via fish consumption remains a health concern. Current therapeutic strategies for MeHg poisoning are based on the use of chelators. However, these therapies have limited efficacy. Ghrelin is a gut hormone with an important role in regulating physiologic processes. It has been reported that ghrelin plays a protective role against the toxicity of several xenobiotics. Here, we explored the role of ghrelin as a putative protector against MeHg-induced oxidative stress. Our data show that ghrelin was able to ameliorate MeHg-induced reactive oxygen species (ROS) production in primary neuronal hypothalamic and hippocampal cultures. An analogous effect was observed in mouse hypothalamic neuronal GT 1-7 cells. Using this model, our novel findings show that antioxidant protection of ghrelin against MeHg is mediated by glutathione upregulation and induction of the NRF2/NQO1 pathway.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| | - Harshini Suresh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia.,Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, Russia
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular Y Nanotecnología, Instituto Nacional de Neurología Y Neurocirugía, 14269, Mexico City, Mexico
| | - João Batista Rocha
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Anatoly V Skalny
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia.,Department of Bioelementology, KG Razumovsky Moscow State University of Technologies and Management, Moscow, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA. .,Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
22
|
Pan J, Li X, Wei Y, Ni L, Xu B, Deng Y, Yang T, Liu W. Advances on the Influence of Methylmercury Exposure during Neurodevelopment. Chem Res Toxicol 2022; 35:43-58. [PMID: 34989572 DOI: 10.1021/acs.chemrestox.1c00255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mercury (Hg) is a toxic heavy-metal element, which can be enriched in fauna and flora and transformed into methylmercury (MeHg). MeHg is a widely distributed environmental pollutant that may be harmful to fish-eating populations through enrichment of aquatic food chains. The central nervous system is a primary target of MeHg. Embryos and infants are more sensitive to MeHg, and exposure to MeHg during gestational feeding can significantly impair the homeostasis of offspring, leading to long-term neurodevelopmental defects. At present, MeHg-induced neurodevelopmental toxicity has become a hotspot in the field of neurotoxicology, but its mechanisms are not fully understood. Some evidence point to oxidative damage, excitotoxicity, calcium ion imbalance, mitochondrial dysfunction, epigenetic changes, and other molecular mechanisms that play important roles in MeHg-induced neurodevelopmental toxicity. In this review, advances in the study of neurodevelopmental toxicity of MeHg exposure during pregnancy and the molecular mechanisms of related pathways are summarized, in order to provide more scientific basis for the study of neurodevelopmental toxicity of MeHg.
Collapse
Affiliation(s)
- Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| |
Collapse
|
23
|
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
|
24
|
Contributing to Understand the Crosstalk between Brain and Periphery in Methylmercury Intoxication: Neurotoxicity and Extracellular Vesicles. Int J Mol Sci 2021; 22:ijms221910855. [PMID: 34639196 PMCID: PMC8509412 DOI: 10.3390/ijms221910855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
Human exposure to methylmercury (MeHg) is currently high in regions such as the Amazon. Understanding the molecular changes associated with MeHg-induced neurotoxicity and the crosstalk with the periphery is essential to support early diagnoses. This work aimed to evaluate cellular and molecular changes associated with behavioral alterations in MeHg acute exposure and the possible changes in extracellular vesicles (EVs) number and S100β content. Adults male Wistar rats were orally treated with 5 mg/kg for four days. Behavioral performance, molecular and histological changes in the cerebellum, and plasma EVs were assessed. MeHg-intoxicated animals performed significantly worse in behavioral tests. MeHg increased the number of GFAP+ cells and GFAP and S100β mRNA expression in the cerebellum but no change in NeuN+ or IBA-1+ cells number was detected. The number of exosomes isolated from plasma were decreased by the metal. S100B mRNA was detected in circulating plasma EVs cargo in MeHg exposure. Though preliminary, our results suggest astrocytic reactivity is displaying a protective role once there was no neuronal death. Interestingly, the reduction in exosomes number could be a new mechanism associated with MeHg-induced neurotoxicity and plasma EVs could represent a source of future biomarkers in MeHg intoxication.
Collapse
|
25
|
He B, Wang Y, Li S, Zhao Y, Ma X, Wang W, Li X, Zhang Y. A cross-sectional survey of preschool children: Exploring heavy metal exposure, neurotransmitters, and neurobehavioural relationships and mediation effects. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112391. [PMID: 34090107 DOI: 10.1016/j.ecoenv.2021.112391] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Exposure to heavy metals has been considered harmful and can cause cognitive deficits in preschool children. OBJECTIVE To investigate the possible mediation effect of neurotransmitters on the relationship of heavy metal exposure with neurobehaviour. METHODS The levels of blood heavy metals and neurotransmitters, along with the neurobehavioural scores, were determined in preschool children. Multiple linear regression was used to assess the relationship between heavy metals, neurotransmitters, and neurobehavioural scores. Furthermore, the mediating role of neurotransmitters was investigated. RESULTS An interquartile range (IQR) increase in lead (6.10 μg/L) was associated with a decrease of 8.52%, 30.06%, and 20.10% for Glutamic acid (Glu), Glycine (Gly), and gamma-aminobutyric acid (GABA), respectively. An IQR increase in arsenic (19.37 μg/L) was associated with an increase of 6.32% and 2.09% for Gly and GABA, respectively. Further, an IQR increase in zinc (15.58 μg/L) was associated with an increase of 1.44% for Ser, whereas the IQR increase was associated with a decrease of 2.14%, 2.24%, and 1.89% for Glu, Gly, and GABA, respectively. An IQR increase in selenium (38.75 μg/L) was associated with an increase of 1.88% for GABA. Moreover, both Glu and Gly decreased by 2.87% for an IQR increase in manganese (16.92 μg/L). An IQR increase in mercury (15.22 μg/L) was associated with a decrease of 2.43% for Ser, but the IQR increase was associated with an increase of 4.99% and 3.09% for Gly and GABA, respectively. It was found that Glu and Serine (Ser) have a significant linear relationship with conduct score and impulsivity-hyperactivity index, and that there was a significant linear relationship between Ser and the learning disability index. GABA and conduct score and attention-deficit hyperactivity disorder (ADHD) index have a significant linear relationship. There is a significant linear relationship between Gly and conduct, anxiety, ADHD, and impulsivity-hyperactivity index. The results of the mediating effect analysis indicated that Ser, Glu, Gly, and GABA have a specific mediating effect between blood heavy metals and neurobehaviour. CONCLUSION We showed the mediating effect of neurotransmitters. The current study may provide valuable information regarding the prevention and management of metal-related neurological disorders in preschool children.
Collapse
Affiliation(s)
- Bin He
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China; The Laboratory Animal Center, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Yan Wang
- Special Medical Service Teaching and Research Section, Tactical Medical Service Department, Army Medical University NCO School, Shijia Zhuang 050051, China
| | - Shuang Li
- The Laboratory Animal Center, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Yuwei Zhao
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Xiaolong Ma
- The Laboratory Animal Center, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Weicheng Wang
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Xinran Li
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China; The Laboratory Animal Center, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China.
| |
Collapse
|
26
|
Zhang J, Li X, Shen L, Khan NU, Zhang X, Chen L, Zhao H, Luo P. Trace elements in children with autism spectrum disorder: A meta-analysis based on case-control studies. J Trace Elem Med Biol 2021; 67:126782. [PMID: 34049201 DOI: 10.1016/j.jtemb.2021.126782] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/30/2021] [Accepted: 05/10/2021] [Indexed: 12/25/2022]
Abstract
Autism spectrum disorder (ASD) is a common childhood neurodevelopmental disorder that may be related to trace elements. However, reports on the relationship between them are still inconsistent. In this article, we conducted a meta-analysis on this issue. We searched the PubMed, EMBASE, and Cochrane databases as of November 15, 2019. A random-effects model was used, and subgroups of studies were analyzed using samples of different measurements. Twenty-two original articles were identified (18 trace elements, including a total of 1014 children with ASD and 999 healthy controls). In autistic children, the overall levels of barium (Ba), mercury (Hg), lithium (Li), and lead (Pb) were higher. There were significant differences in the levels of copper (Cu) in the hair and serum between autistic children and the control group. The levels of Hg, Li, Pb and selenium (Se) in the hair of autistic children were higher than those of healthy children, while the levels of zinc (Zn) in the blood were lower. Excessive exposure to toxic heavy metals and inadequate intake of essential metal elements may be associated with ASD. Preventing excessive exposure to toxic metals and correcting poor dietary behaviors may be beneficial for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Jun Zhang
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring Control Ministry of Education, Guizhou Medical University, 550025, PR China
| | - Xi Li
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring Control Ministry of Education, Guizhou Medical University, 550025, PR China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, PR China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, PR China.
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518060, PR China
| | - Xiao Zhang
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring Control Ministry of Education, Guizhou Medical University, 550025, PR China
| | - Lulu Chen
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring Control Ministry of Education, Guizhou Medical University, 550025, PR China
| | - Huan Zhao
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring Control Ministry of Education, Guizhou Medical University, 550025, PR China
| | - Peng Luo
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring Control Ministry of Education, Guizhou Medical University, 550025, PR China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China.
| |
Collapse
|
27
|
Ferrer B, Suresh H, Santamaria A, Rocha JB, Bowman AB, Aschner M. The antioxidant role of STAT3 in methylmercury-induced toxicity in mouse hypothalamic neuronal GT1-7 cell line. Free Radic Biol Med 2021; 171:245-259. [PMID: 34010664 PMCID: PMC8217327 DOI: 10.1016/j.freeradbiomed.2021.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/04/2021] [Accepted: 05/13/2021] [Indexed: 12/27/2022]
Abstract
Oxidative stress, impairment of antioxidant defenses, and disruption of calcium homeostasis are associated with the toxicity of methylmercury (MeHg). Yet, the relative contribution and interdependence of these effects and other molecular mechanisms that mediate MeHg-induced neurotoxicity remain uncertain. The signal transducer and activator of transcription 3 (STAT3) is a transcription factor that regulates the expression of anti-apoptotic and cell cycle progression genes. In addition to its role in cell growth and survival, STAT3 regulates redox homeostasis and prevents oxidative stress by the modulation of nuclear genes that encode for electron transport complexes (ETC) and antioxidant enzymes. Here we tested the hypothesis that STAT3 contributes to the orchestration of the antioxidant defense response against MeHg injury. We show that MeHg (>1 μM) exposure induced STAT3 activation within 1 h and beyond in mouse hypothalamic neuronal GT1-7 cells in a concentration-and time-dependent manner. Pharmacological inhibition of STAT3 phosphorylation exacerbated MeHg-induced reactive oxygen species (ROS) production and antioxidant responses. Finally, treatment with the antioxidant Trolox demonstrated that MeHg-induced STAT3 activation is mediated, at least in part, by MeHg-induced ROS generation. Combined, our results demonstrated a role for the STAT3 signaling pathway as an early response to MeHg-induced oxidative stress.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Harshini Suresh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico.
| | - João Batista Rocha
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil.
| | - 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, 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
|
28
|
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: 2.7] [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
|
29
|
Mechanisms of oxidative stress in methylmercury-induced neurodevelopmental toxicity. Neurotoxicology 2021; 85:33-46. [PMID: 33964343 DOI: 10.1016/j.neuro.2021.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022]
Abstract
Methylmercury (MeHg) is a long-lasting organic environmental pollutant that poses a great threat to human health. Ingestion of seafood containing MeHg is the most important way by which it comes into contact with human body, where the central nervous system (CNS) is the primary target of MeHg toxicity. During periods of pre-plus postnatal, in particular, the brain of offspring is vulnerable to specific developmental insults that result in abnormal neurobehavioral development, even without symptoms in mothers. While many studies on neurotoxic effects of MeHg on the developing brain have been conducted, the mechanisms of oxidative stress in MeHg-induced neurodevelopmental toxicity is less clear. Hitherto, no single process can explain the many effects observed in MeHg-induced neurodevelopmental toxicity. This review summarizes the possible mechanisms of oxidative stress in MeHg-induced neurodevelopmental toxicity, highlighting modulation of Nrf2/Keap1/Notch1, PI3K/AKT, and PKC/MAPK molecular pathways as well as some preventive drugs, and thus contributes to the discovery of endogenous and exogenous molecules that can counteract MeHg-induced neurodevelopmental toxicity.
Collapse
|
30
|
Environmentally relevant developmental methylmercury exposures alter neuronal differentiation in a human-induced pluripotent stem cell model. Food Chem Toxicol 2021; 152:112178. [PMID: 33831500 DOI: 10.1016/j.fct.2021.112178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Developmental methylmercury (MeHg) exposure selectively targets the cerebral and cerebellar cortices, as seen by disruption of cytoarchitecture and glutamatergic (GLUergic) neuron hypoplasia. To begin to understand the mechanisms of this loss of GLUergic neurons, we aimed to develop a model of developmental MeHg neurotoxicity in human-induced pluripotent stem cells differentiating into cortical GLUergic neurons. Three dosing paradigms at 0.1 μM and 1.0 μM MeHg, which span different stages of neurodevelopment and reflect toxicologically relevant accumulation levels seen in human studies and mammalian models, were established. With these exposure paradigms, no changes were seen in commonly studied endpoints of MeHg toxicity, including viability, proliferation, and glutathione levels. However, MeHg exposure induced changes in mitochondrial respiration and glycolysis and in markers of neuronal differentiation. Our novel data suggests that GLUergic neuron hypoplasia seen with MeHg toxicity may be due to the partial inhibition of neuronal differentiation, given the increased expression of the early dorsal forebrain marker FOXG1 and corresponding decrease in expression on neuronal markers MAP2 and DCX and the deep layer cortical neuronal marker TBR1. Future studies should examine the persistent and latent functional effects of this MeHg-induced disruption of neuronal differentiation as well as transcriptomic and metabolomic alterations that may mediate MeHg toxicity.
Collapse
|
31
|
Novo JP, Martins B, Raposo RS, Pereira FC, Oriá RB, Malva JO, Fontes-Ribeiro C. Cellular and Molecular Mechanisms Mediating Methylmercury Neurotoxicity and Neuroinflammation. Int J Mol Sci 2021; 22:ijms22063101. [PMID: 33803585 PMCID: PMC8003103 DOI: 10.3390/ijms22063101] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Methylmercury (MeHg) toxicity is a major environmental concern. In the aquatic reservoir, MeHg bioaccumulates along the food chain until it is consumed by riverine populations. There has been much interest in the neurotoxicity of MeHg due to recent environmental disasters. Studies have also addressed the implications of long-term MeHg exposure for humans. The central nervous system is particularly susceptible to the deleterious effects of MeHg, as evidenced by clinical symptoms and histopathological changes in poisoned humans. In vitro and in vivo studies have been crucial in deciphering the molecular mechanisms underlying MeHg-induced neurotoxicity. A collection of cellular and molecular alterations including cytokine release, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate dyshomeostasis, and cell death mechanisms are important consequences of brain cells exposure to MeHg. The purpose of this review is to organize an overview of the mercury cycle and MeHg poisoning events and to summarize data from cellular, animal, and human studies focusing on MeHg effects in neurons and glial cells. This review proposes an up-to-date compendium that will serve as a starting point for further studies and a consultation reference of published studies.
Collapse
Affiliation(s)
- João P. Novo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Beatriz Martins
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Ramon S. Raposo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Experimental Biology Core, University of Fortaleza, Health Sciences, Fortaleza 60110-001, Brazil
| | - Frederico C. Pereira
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil;
| | - João O. Malva
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| | - Carlos Fontes-Ribeiro
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| |
Collapse
|
32
|
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: 0.8] [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
|
33
|
Neuroprotective Effect of Tricyclic Pyridine Alkaloids from Fusarium lateritium SSF2, against Glutamate-Induced Oxidative Stress and Apoptosis in the HT22 Hippocampal Neuronal Cell Line. Antioxidants (Basel) 2020; 9:antiox9111115. [PMID: 33187346 PMCID: PMC7698086 DOI: 10.3390/antiox9111115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/01/2020] [Accepted: 11/06/2020] [Indexed: 12/24/2022] Open
Abstract
Excessive glutamate damages neuronal cells via the accumulation of intracellular reactive oxygen species (ROS), calcium ion (Ca2+) influx, depolarization of mitochondrial membrane potential, and apoptosis, which may result in the development of chronic neurodegenerative diseases. In this study, we evaluated the effects of 4,6′-anhydrooxysporidinone isolated from endophytic fungus Fusarium lateritium SSF2 on glutamate-induced cytotoxicity, accumulation of intracellular ROS, increases in superoxide anion production, Ca2+, depolarization of mitochondrial membrane potential, and apoptotic cell death in hippocampal HT22 cells. 2′,7′-Dichlorofluorescin diacetate (H2DCFDA) staining was used to determine the intracellular reactive oxygen species concentration and dihydroethidine (DHE) staining was used to determine the superoxide radical. Expression of the nuclear factor-erythroid-2–related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) was analyzed by Western blot. Fluo-4 staining was used to determine the intracellular Ca2+ levels. In order to explore mitochondrial membrane potential, tetramethylrhodamine methyl ester (TMRM) staining was used. Apoptotic cell death was evaluated using Annexin-V/propidium iodide (PI) staining and TUNEL staining. Expression of the cytochrome c release and cleaved caspase-9, -3 was analyzed by Western blot. Here, we were able to isolate 4,6′-anhydrooxysporidinone from endophytic fungus, Fusarium lateritium SSF2, which was shown to protect HT22 cells from glutamate-induced cytotoxicity, accumulation of intracellular ROS, increases in superoxide anion production, Ca2+, and depolarization of mitochondrial membrane potential. In addition, 4,6′-anhydrooxysporidinone enhanced the expressions of Nrf2 and HO-1. It also inhibited the apoptotic cell death through the inhibition of cytochrome c release and cleaved caspase-9, -3 in glutamate-treated HT22 cells. Therefore, our results provide ample evidence of the neuroprotective properties of 4,6′-anhydrooxysporidinone.
Collapse
|
34
|
Freire MAM, Lima RR, Nascimento PC, Gomes-Leal W, Pereira A. Effects of methylmercury on the pattern of NADPH diaphorase expression and astrocytic activation in the rat. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 201:110799. [PMID: 32544743 DOI: 10.1016/j.ecoenv.2020.110799] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
Mercury (Hg) is an environmental contaminant that poses great risk to human health. However, it is still widely used in artisanal gold-mining enterprises around the world, especially in developing countries. Methylmercury (MeHg) is produced environmentally by biomethylation of inorganic Hg present in water sediments, leading to its subsequent accumulation in the aquatic food chain. Due to its high metabolic rate, the Central Nervous System (CNS) is one of the main targets of MeHg. In the present study, we investigate the impact of chronic MeHg intoxication on NADPH diaphorase (NADPH-d) activity and astrocyte mobilization in the visual cortex of the rat. After 60 days of MeHg administration by oral gavage (0.04 mg/kg/day), tissue samples containing the visual cortex were submitted to measurements of Hg levels, NADPH-d activity, and GFAP immunohistochemistry for identification of astrocytes. MeHg intoxication was associated with increased Hg deposits and with reduced NADPH-d neuropil reactivity in the visual cortex. A morphometric analysis suggested that NADPH-d-positive neurons were mostly spared from MeHg harmful action and intoxicated animals had astrocytic activation similar to the control group. The decrease in NADPH-d neuropil reactivity may be due to the negative effect of chronic MeHg poisoning on both the synthesis and transport of this enzyme in afferent pathways to the visual cortex. The relative resistance of NADPH-d-reactive neurons to chronic MeHg intoxication may be associated with peculiarities in cell metabolism or to a protective role of nitric oxide, safeguarding those neurons from Hg deleterious effects.
Collapse
Affiliation(s)
- Marco Aurelio M Freire
- Graduate Program in Health and Society, Faculty of Health Sciences, University of State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Priscila Cunha Nascimento
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Walace Gomes-Leal
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Antonio Pereira
- Department of Electrical and Biomedical Engineering, Institute of Technology, Federal University of Pará (UFPA), Belém, PA, Brazil.
| |
Collapse
|
35
|
Effects of foetal and breastfeeding exposure to methylmercury (MeHg) and retinol palmitate (Vitamin A) in rats: Redox parameters and susceptibility to DNA damage in liver. Mutat Res 2020; 858-860:503239. [PMID: 33198929 DOI: 10.1016/j.mrgentox.2020.503239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 08/01/2020] [Accepted: 08/14/2020] [Indexed: 01/01/2023]
Abstract
Methylmercury (MeHg) is known to be a chemical that poses a risk to public health. Exposure to MeHg and vitamin A (VitA) occurs through the ingestion of fish, present in the diet of most pregnant women. The absorption of these elements generates oxidative stress and can generate adaptations for future stressful events. Here, we assessed how exposure to VitA and/or MeHg during the fetal and breastfeeding period modulates the toxicity of MeHg reexposure in adulthood. We focus on redox systems and repairing DNA damage. Male rats (n = 50), were divided into 5 groups. Control received mineral oil; The VitA group received VitA during pregnancy, during breastfeeding and was exposed to MeHg in adulthood; VitA + MeHg received VitA and MeHg during pregnancy and breastfeeding and was exposed to MeHg in adulthood. The single exposure group (SE) was exposed to MeHg only in adulthood; and the MeHg group was pre-exposed to MeHg during pregnancy and breastfeeding and re-exposed to MeHg in adulthood. After treating the animals, we evaluated the redox status and the level of DNA damage in all rats. The results revealed that MeHg significantly decreased the activity of glutathione peroxidase (GPx) and sulfhydryl levels and increased the activity of superoxide dismutase (SOD), glutathione transferase, glutathione and carbonyl in all exposed groups. These results suggest that the second exposure to MeHg directly altered the effects of oxidation and that there were no specific effects associated with exposure during the fetal and breastfeeding periods. In addition, our findings indicate that MDA levels increased in MeHg and SE levels and no differences in MDA levels were observed between the VitA and MeHg + VitA groups. We also observed that animals pretreated exclusively with VitA showed residual damage similar to the control's DNA, while the other groups showed statistically higher levels of damage. In conclusion, low doses of MeHg and VitA during fetal and breastfeeding periods were unable to condition an adaptive response to subsequent exposure to MeHg in adulthood in relation to the observed levels of oxidative damage assessed after exposure.
Collapse
|
36
|
Gunderson JT, Peppriell AE, Vorojeikina D, Rand MD. Tissue-specific Nrf2 signaling protects against methylmercury toxicity in Drosophila neuromuscular development. Arch Toxicol 2020; 94:4007-4022. [PMID: 32816092 DOI: 10.1007/s00204-020-02879-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Methylmercury (MeHg) can elicit cognitive and motor deficits due to its developmental neuro- and myotoxic properties. While previous work has demonstrated that Nrf2 antioxidant signaling protects from MeHg toxicity, in vivo tissue-specific studies are lacking. In Drosophila, MeHg exposure shows greatest developmental toxicity in the pupal stage resulting in failed eclosion (emergence of adults) and an accompanying 'myosphere' phenotype in indirect flight muscles (IFMs). To delineate tissue-specific contributions to MeHg-induced motor deficits, we investigated the potential of Nrf2 signaling in either muscles or neurons to moderate MeHg toxicity. Larva were exposed to various concentrations of MeHg (0-20 µM in food) in combination with genetic modulation of the Nrf2 homolog cap-n-collar C (CncC), or its negative regulator Keap1. Eclosion behavior was evaluated in parallel with the morphology of two muscle groups, the thoracic IFMs and the abdominal dorsal internal oblique muscles (DIOMs). CncC signaling activity was reported with an antioxidant response element construct (ARE-GFP). We observed that DIOMs are distinguished by elevated endogenous ARE-GFP expression, which is only transiently seen in the IFMs. Dose-dependent MeHg reductions in eclosion behavior parallel formation of myospheres in the DIOMs and IFMs, while also increasing ARE-GFP expression in the DIOMs. Modulating CncC signaling via muscle-specific Keap1 knockdown and upregulation gives a rescue and exacerbation, respectively, of MeHg effects on eclosion and myospheres. Interestingly, muscle-specific CncC upregulation and knockdown both induce lethality. In contrast, neuron-specific upregulation of CncC, as well as Keap1 knockdown, rescued MeHg effects on eclosion and myospheres. Our findings indicate that enhanced CncC signaling localized to either muscles or neurons is sufficient to rescue muscle development and neuromuscular function from a MeHg insult. Additionally, there may be distinct roles for CncC signaling in myo-morphogenesis.
Collapse
Affiliation(s)
- Jakob T Gunderson
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ashley E Peppriell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
37
|
Lee JY, Hwang GW, Naganuma A, Satoh M. Methylmercury toxic mechanism related to protein degradation and chemokine transcription. Environ Health Prev Med 2020; 25:30. [PMID: 32680455 PMCID: PMC7469908 DOI: 10.1186/s12199-020-00868-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/29/2020] [Indexed: 01/01/2023] Open
Abstract
Methylmercury is an environmental pollutant that causes neurotoxicity. Recent studies have reported that the ubiquitin-proteasome system is involved in defense against methylmercury toxicity through the degradation of proteins synthesizing the pyruvate. Mitochondrial accumulation of pyruvate can enhance methylmercury toxicity. In addition, methylmercury exposure induces several immune-related chemokines, specifically in the brain, and may cause neurotoxicity. This summary highlights several molecular mechanisms of methylmercury-induced neurotoxicity.
Collapse
Affiliation(s)
- Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| | - Gi-Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, 981-8558, Japan.,Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Akira Naganuma
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Masahiko Satoh
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| |
Collapse
|
38
|
Yang T, Xu Z, Liu W, Xu B, Deng Y. Oxidative stress accelerates synaptic glutamate dyshomeostasis and NMDARs disorder during methylmercury-induced neuronal apoptosis in rat cerebral cortex. ENVIRONMENTAL TOXICOLOGY 2020; 35:683-696. [PMID: 32061141 DOI: 10.1002/tox.22904] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 06/10/2023]
Abstract
Methylmercury (MeHg) is a potent neurotoxin,which leads to a wide range of intracellular effects. The molecular mechanismsassociated to MeHg-induced neurotoxicity have not been fully understood.Oxidative stress, as well as synaptic glutamate (Glu) dyshomeostasis have beenidentified as two critical mechanisms during MeHg-mediated cytotoxicity. Here,we developed a rat model of MeHg poisoning to evaluate its neurotoxic effectsby focusing on cellular oxidative stress and synaptic Glu disruption. Inaddition, we investigated the neuroprotective role of alpha-lipoic acid (α-LA), a natural antioxidant, todeeply explore the underlying interaction between them. Fifty-six rats wererandomly divided into four groups: saline control, MeHg treatment (4 or 12μmol/kg MeHg), and α-LApre-treatment (35 μmol/kg α-LA+12μmol/kg MeHg). Rats exposed to 12 μmol/kg MeHg induced neuronal oxidativestress, with ROS accumulation and cellular antioxidant system impairment. Nrf2 andxCT pathways were activated with MeHg treatment. The enzymatic or non-enzymaticof cellular GSH synthesis were also disrupted by MeHg. On the other hand, the abnormalactivities of GS and PAG disturbed the "Glu-Gln cycle", leading to NMDARsover-activation, Ca2+ overload, and the calpain activation, which acceleratedNMDARs degradation. Meanwhile, the high expressions of phospho-p44/42 MAPK,phospho-p38 MAPK, phospho-CREB, and the high levels of caspase 3 and Bax/Bcl-2 finallyindicated the neuronal apoptosis after MeHg exposure. Pre-treatment with α-LA significantly preventedMeHg-induced neurotoxicity. In conclusion, the oxidative stress and synapticGlu dyshomeostasis contributed to MeHg-induced neuronal apoptosis. Alpha-LAattenuated these toxic effects through mechanisms of anti-oxidation andindirect Glu dyshomeostasis prevention.
Collapse
Affiliation(s)
- Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, People's Republic of China
| |
Collapse
|
39
|
Yang L, Zhang Y, Wang F, Luo Z, Guo S, Strähle U. Toxicity of mercury: Molecular evidence. CHEMOSPHERE 2020; 245:125586. [PMID: 31881386 DOI: 10.1016/j.chemosphere.2019.125586] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/28/2019] [Accepted: 12/08/2019] [Indexed: 05/25/2023]
Abstract
Minamata disease in Japan and the large-scale poisoning by methylmercury (MeHg) in Iraq caused wide public concerns about the risk emanating from mercury for human health. Nowadays, it is widely known that all forms of mercury induce toxic effects in mammals, and increasing evidence supports the concern that environmentally relevant levels of MeHg could impact normal biological functions in wildlife. The information of mechanism involved in mercurial toxicity is growing but knowledge gaps still exist between the adverse effects and mechanisms of action, especially at the molecular level. A body of data obtained from experimental studies on mechanisms of mercurial toxicity in vivo and in vitro points to that disruption of the antioxidant system may play an important role in the mercurial toxic effects. Moreover, the accumulating evidence indicates that signaling transduction, protein or/and enzyme activity, and gene regulation are involving in mediating toxic and adaptive response to mercury exposure. We conducted here a comprehensive review of mercurial toxic effects on wildlife and human, in particular synthesized key findings of molecular pathways involved in mercurial toxicity from the cells to human. We discuss the molecular evidence related mercurial toxicity to the adverse effects, with particular emphasis on the gene regulation. The further studies relying on Omic analysis connected to adverse effects and modes of action of mercury will aid in the evaluation and validation of causative relationship between health outcomes and gene expression.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| | - Yuanyuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Zidie Luo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Uwe Strähle
- Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
40
|
Lin X, Zhao J, Zhang W, He L, Wang L, Chang D, Cui L, Gao Y, Li B, Chen C, Li YF. Acute oral methylmercury exposure perturbs the gut microbiome and alters gut-brain axis related metabolites in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 190:110130. [PMID: 31918252 DOI: 10.1016/j.ecoenv.2019.110130] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/18/2019] [Accepted: 12/22/2019] [Indexed: 06/10/2023]
Abstract
Environmental pollutants like methylmercury (MeHg) can bring devastating neurotoxicity to animals and human beings. Gut microbiota has been found to demethylate MeHg and promote the excretion of Hg through feces. However, the impacts of MeHg on gut microbiota and metabolites related to gut-brain interactions were less studied in mammals. The object of this study was to investigate the impacts of acute MeHg exposure on gut microbiome and metabolites together with its impact on gut integrity and related biological responses in rats. Rats were exposed to MeHg through oral administration and were sacrificed after 24 h 16 S rRNA gene sequencing was used to study the perturbance to gut microbiome and liquid chromatography mass spectrometry (LC-MS) was used for metabolomics profiling. It was found that gut was one of the target tissues of MeHg. MeHg induce the changes of intestinal microbial community structure and induce the regulating neuron activity change of intestinal neurotransmitters and metabolites on intestinal neurotransmitters and metabolites regulating the neuron activity. This was supported by the increased BDNF level. These findings may suggest a potential new mechanism regarding the neurotoxicity of MeHg. The protocols used in this study may also be applied to understand the neurotoxicity of other environmental neurotoxins like Pb, Mn, polychlorinated biphenyls, and pesticides, etc and to screen the neurotoxicity of emerging environmental contaminants.
Collapse
Affiliation(s)
- Xiaoying Lin
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiating Zhao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Zhang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Lina He
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Liming Wang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Dunhu Chang
- School of Environment and Natural Resources, Renmin University of China, Beijing, 100872, China.
| | - Liwei Cui
- State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Beijing Advanced Sciences and Innovation Centre, Chinese Academy of Sciences, Beijing, 101407, China
| | - Yuxi Gao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Bai Li
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunying Chen
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing, 100191, China
| | - Yu-Feng Li
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, Beijing, 100049, China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Chinese Academy of Sciences, Beijing, 100049, China; State Environmental Protection Engineering Centre for Mercury Pollution Prevention and Control, Chinese Academy of Sciences, Beijing, 100049, China; Beijing Metallomics Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
41
|
Rajabian A, Sadeghnia HR, Hosseini A, Mousavi SH, Boroushaki MT. 3-Acetyl-11-keto-β-boswellic acid attenuated oxidative glutamate toxicity in neuron-like cell lines by apoptosis inhibition. J Cell Biochem 2020; 121:1778-1789. [PMID: 31642100 DOI: 10.1002/jcb.29413] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 10/04/2019] [Indexed: 12/18/2022]
Abstract
3-Acetyl-11-keto-β-boswellic acid (AKBA), a pentacyclic triterpenic acid present in gum resin of Boswellia serrata, has been found to possess antioxidant and neuroprotective properties. In this study, we aimed to examine protective properties of AKBA against glutamate-induced neuronal injury. To investigate the effects of AKBA (2.5-10 µM) on glutamate injury in neuron-like cells PC12 and N2a, two treatment regimens (incubation for 2 or 0 hours before glutamate exposure) were used. Then, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method was used to determine viability of the cells. Cellular redox status was evaluated using fluorimetry and comet assays. Annexin V/propidium iodide double staining and Western blot analysis of relative apoptotic proteins were conducted. Based on the results, 24 hours incubation with glutamate (8 mM) increased the cell mortality of PC12 and N2a (P < .001). However, AKBA (2.5-10 µM) enhanced the cell viability in both treatment regimens (P < .001). Also co- and pretreatment with AKBA significantly attenuated lipid peroxidation, reactive oxygen species production, and DNA injury (P < .05 and P < .001). AKBA also restored the activity of cellular superoxide dismutase under glutamate toxicity; this effect was seen to be more significant during the pretreatment regimen (P < .001). Moreover, Western blot analysis indicated that AKBA inhibited glutamate-induced programmed cell death through depressing the elevation of the expression ratio of Bax/Bcl-2 and cleaved-caspase-3 proteins, concentration-dependently. Overall, the present findings suggest the neuroprotective activities of AKBA against glutamate-induced cell injury probably by inhibiting oxidative damage and reducing apoptotic cell death.
Collapse
Affiliation(s)
- Arezoo Rajabian
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azar Hosseini
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hadi Mousavi
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Taher Boroushaki
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
42
|
Aaseth J, Wallace DR, Vejrup K, Alexander J. Methylmercury and developmental neurotoxicity: A global concern. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
43
|
Effects of Gintonin-Enriched Fraction on Methylmercury-Induced Neurotoxicity and Organ Methylmercury Elimination. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17030838. [PMID: 32013120 PMCID: PMC7038146 DOI: 10.3390/ijerph17030838] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/18/2020] [Accepted: 01/24/2020] [Indexed: 01/09/2023]
Abstract
Gintonin is a newly discovered ingredient of ginseng and plays an exogenous ligand for G protein-coupled lysophosphatidic acid receptors. We previously showed that gintonin exhibits diverse effects from neurotransmitter release to improvement of Alzheimer’s disease-related cognitive dysfunctions. However, previous studies did not show whether gintonin has protective effects against environmental heavy metal. We investigated the effects of gintonin-enriched fraction (GEF) on methylmercury (MeHg)-induced neurotoxicity and learning and memory dysfunction and on organ MeHg elimination. Using hippocampal neural progenitor cells (hNPCs) and mice we examined the effects of GEF on MeHg-induced hippocampal NPC neurotoxicity, on formation of reactive oxygen species (ROS), and on in vivo learning and memory functions after acute MeHg exposure. Treatment of GEF to hNPCs attenuated MeHg-induced neurotoxicity with concentration- and time-dependent manner. GEF treatment inhibited MeHg- and ROS inducer-induced ROS formations. Long-term treatment of GEF also improved MeHg-induced learning and memory dysfunctions. Oral administration of GEF decreased the concentrations of MeHg in blood, brain, liver, and kidney. This is the first report that GEF attenuated MeHg-induced in vitro and in vivo neurotoxicities through LPA (lysophosphatidic acids) receptor-independent manner and increased organ MeHg elimination. GEF-mediated neuroprotection might achieve via inhibition of ROS formation and facilitation of MeHg elimination from body.
Collapse
|
44
|
Gagnon-Chauvin A, Bastien K, Saint-Amour D. Environmental toxic agents: The impact of heavy metals and organochlorides on brain development. HANDBOOK OF CLINICAL NEUROLOGY 2020; 173:423-442. [PMID: 32958188 DOI: 10.1016/b978-0-444-64150-2.00030-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Exposure to environmental toxicants can have deleterious effects on the development of physical, cognitive, and mental health. Extensive laboratory and clinical studies have demonstrated how the developing brain is uniquely sensitive to toxic agents. This chapter focuses on the main neurologic impairments linked to prenatal and postnatal exposure to lead, methylmercury, and polychlorinated biphenyls, three legacy environmental contaminants whose neurotoxic effects have been extensively studied with respect to cognitive and behavioral development. The main cognitive, emotion regulation, sensory, and motor impairments in association with these contaminants are briefly reviewed, including the underlying neural mechanisms such as neuropathologic damages, brain neurotransmission, and endocrine system alterations. The use of neuroimaging as a novel tool to better understand how the brain is affected by exposure to environmental contaminants is also discussed.
Collapse
Affiliation(s)
- Avril Gagnon-Chauvin
- Department of Psychology, Université du Québec à Montréal, Montréal, QC, Canada; Research Centre, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Kevin Bastien
- Department of Psychology, Université du Québec à Montréal, Montréal, QC, Canada; Research Centre, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Dave Saint-Amour
- Department of Psychology, Université du Québec à Montréal, Montréal, QC, Canada; Research Centre, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
45
|
Yuan L, Qian L, Qian Y, Liu J, Yang K, Huang Y, Wang C, Li Y, Mu X. Bisphenol F-Induced Neurotoxicity toward Zebrafish Embryos. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:14638-14648. [PMID: 31702913 DOI: 10.1021/acs.est.9b04097] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In this study, the influence of bisphenol F (BPF) toward central nervous system (CNS) was assessed using zebrafish embryos. We found that BPF could induce significant neurotoxicity toward zebrafish embryos, including inhibited locomotion, reduced moving distance, and CNS cell apoptosis at an effective concentration of 0.0005 mg/L. Immunofluorescence assay showed that both microglia and astrocyte in zebrafish brain were significantly activated by BPF, indicating the existence of neuroinflammatory response. Peripheral motor neuron development was significantly inhibited by BPF at 72 hpf. RNA-seq data indicated that neuronal developmental processes and cell apoptosis pathways were significantly affected by BPF exposure, which was consistent with the phenotypic results. Chip-seq assay implied that the transcriptional changes were not mediated by ERα. Additionally, no significant change was found in neurotransmitter levels (5-hydroxytryptamine, dopamine, and acetylcholine) or acetylcholinesterase (Ache) enzyme activity after BPF exposure, indicating that BPF may not affect neurotransmission. In conclusion, BPF could lead to abnormal neural outcomes during zebrafish early life stage through inducing neuroinflammation and CNS cell apoptosis even at environmentally relevant concentration.
Collapse
Affiliation(s)
- Lilai Yuan
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Le Qian
- College of Sciences , China Agricultural University , Beijing 100193 , People's Republic of China
| | - Yu Qian
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Jia Liu
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Ke Yang
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Ying Huang
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Chengju Wang
- College of Sciences , China Agricultural University , Beijing 100193 , People's Republic of China
| | - Yingren Li
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Xiyan Mu
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| |
Collapse
|
46
|
ROS and diseases: role in metabolism and energy supply. Mol Cell Biochem 2019; 467:1-12. [PMID: 31813106 PMCID: PMC7089381 DOI: 10.1007/s11010-019-03667-9] [Citation(s) in RCA: 323] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 11/30/2019] [Indexed: 02/07/2023]
Abstract
Researches dedicated to reactive oxygen species (ROS) had been performed for decades, yet the outcomes remain controversial. With the relentless effort of studies, researchers have explored the role of ROS in biosystem and various diseases. ROS are beneficial for biosystem presenting as signalling molecules and enhancing immunologic defence. However, they also have harmful effects such as causing tissue and organ damages. The results are controversial in studies focusing on ROS and ROS-related diseases by regulating ROS with inhibitors or promotors. These competing results hindered the process for further investigation of the specific mechanisms lying behind. The opinions presented in this review interpret the researches of ROS from a different dimension that might explain the competing results of ROS introduced so far from a broader perspective. This review brings a different thinking to researchers, with the neglected features and potentials of ROS, to relate their works with ROS and to explore the mechanisms between their subject and ROS.
Collapse
|
47
|
Chang J, Yang B, Zhou Y, Yin C, Liu T, Qian H, Xing G, Wang S, Li F, Zhang Y, Chen D, Aschner M, Lu R. Acute Methylmercury Exposure and the Hypoxia-Inducible Factor-1α Signaling Pathway under Normoxic Conditions in the Rat Brain and Astrocytes in Vitro. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:127006. [PMID: 31850806 PMCID: PMC6957278 DOI: 10.1289/ehp5139] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 11/07/2019] [Accepted: 11/18/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND As a ubiquitous environmental pollutant, methylmercury (MeHg) induces toxic effects in the nervous system, one of its main targets. However, the exact mechanisms of its neurotoxicity have not been fully elucidated. Hypoxia-inducible factor- 1 α (HIF- 1 α ), a transcription factor, plays a crucial role in adaptive and cytoprotective responses in cells and is involved in cell survival, proliferation, apoptosis, inflammation, angiogenesis, glucose metabolism, erythropoiesis, and other physiological activities. OBJECTIVES The aim of this study was to explore the role of HIF- 1 α in response to acute MeHg exposure in rat brain and primary cultured astrocytes to improve understanding of the mechanisms of MeHg-induced neurotoxicity and the development of effective neuroprotective strategies. METHODS Primary rat astrocytes were treated with MeHg (0 - 10 μ M ) for 0.5 h . Cell proliferation and cytotoxicity were assessed with a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl diphenyltetrazolium bromide (MTT) assay and a lactate dehydrogenase (LDH) release assay, respectively. Reactive oxygen species (ROS) levels were analyzed to assess the level of oxidative stress using 2',7'-dichlorofluorescin diacetate (DCFH-DA) fluorescence. HIF- 1 α , and its downstream proteins, glucose transporter 1 (GLUT-1), erythropoietin (EPO), and vascular endothelial growth factor A (VEGF-A) were analyzed by means of Western blotting. Real-time PCR was used to detect the expression of HIF- 1 α mRNA. Pretreatment with protein synthesis inhibitor (CHX), proteasome inhibitor (MG132), or proline hydroxylase inhibitor (DHB) were applied to explore the possible mechanisms of HIF- 1 α inhibition by MeHg. To investigate the role of HIF- 1 α in MeHg-induced neurotoxicity, cobalt chloride (CoC l 2 ), 2-methoxyestradiol (2-MeOE2), small interfering RNA (siRNA) transfection and adenovirus overexpression were used. Pretreatment with N-acetyl-L-cysteine (NAC) and vitamin E (Trolox) were used to investigate the putative role of oxidative stress in MeHg-induced alterations in HIF- 1 α levels. The expression of HIF- 1 α and related downstream proteins was detected in adult rat brain exposed to MeHg (0 - 10 mg / kg ) for 0.5 h in vivo. RESULTS MeHg caused lower cell proliferation and higher cytotoxicity in primary rat astrocytes in a time- and concentration-dependent manner. In comparison with the control cells, exposure to 10 μ M MeHg for 0.5 h significantly inhibited the expression of astrocytic HIF- 1 α , and the downstream genes GLUT-1, EPO, and VEGF-A (p < 0.05 ), in the absence of a significant decrease in HIF- 1 α mRNA levels. When protein synthesis was inhibited by CHX, MeHg promoted the degradation rate of HIF- 1 α . MG132 and DHB significantly blocked the MeHg-induced decrease in HIF- 1 α expression (p < 0.05 ). Overexpression of HIF- 1 α significantly attenuated the decline in MeHg-induced cell proliferation, whereas the inhibition of HIF- 1 α significantly increased the decline in cell proliferation (p < 0.05 ). NAC and Trolox, two established antioxidants, reversed the MeHg-induced decline in HIF- 1 α protein levels and the decrease in cell proliferation (p < 0.05 ). MeHg suppressed the expression of HIF- 1 α and related downstream target proteins in adult rat brain. DISCUSSION MeHg induced a significant reduction in HIF- 1 α protein by activating proline hydroxylase (PHD) and the ubiquitin proteasome system (UPS) in primary rat astrocytes. Additionally, ROS scavenging by antioxidants played a neuroprotective role via increasing HIF- 1 α expression in response to MeHg toxicity. Moreover, we established that up-regulation of HIF- 1 α might serve to mitigate the acute toxicity of MeHg in astrocytes, affording a novel therapeutic target for future exploration. https://doi.org/10.1289/EHP5139.
Collapse
Affiliation(s)
- Jie Chang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yun Zhou
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Changsheng Yin
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Tingting Liu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hai Qian
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yubin Zhang
- Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai, China
| | - Da Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China
- Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, China
| |
Collapse
|
48
|
Yang B, Yin C, Zhou Y, Wang Q, Jiang Y, Bai Y, Qian H, Xing G, Wang S, Li F, Feng Y, Zhang Y, Cai J, Aschner M, Lu R. Curcumin protects against methylmercury-induced cytotoxicity in primary rat astrocytes by activating the Nrf2/ARE pathway independently of PKCδ. Toxicology 2019; 425:152248. [PMID: 31330227 PMCID: PMC6710134 DOI: 10.1016/j.tox.2019.152248] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/07/2019] [Accepted: 07/18/2019] [Indexed: 12/11/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxicant that leads to long-lasting neurological deficits in animals and humans. Curcumin, a polyphenol obtained from the rhizome of turmeric, has well-known antioxidant functions. Here, we evaluated curcumin's efficacy in mitigating MeHg-induced cytotoxicity and further investigated the underlying mechanism of this neuroprotection in primary rat astrocytes. Pretreatment with curcumin (2, 5, 10 and 20 μM for 3, 6, 12 or 24 h) protected against MeHg-induced (5 μM for 6 h) cell death in a time and dose-dependent manner. Curcumin (2, 5, 10 or 20 μM) pretreatment for 12 h significantly ameliorated the MeHg-induced astrocyte injury and oxidative stress, as evidenced by morphological alterations, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) generation, and glutathione (GSH) and catalase (CAT) levels. Moreover, curcumin pretreatment increased Nrf2 nuclear translocation and downstream enzyme expression, heme oxygenase-1 (HO-1) and NADPH quinone reductase-1 (NQO1). Knockdown of Nrf2 with siRNA attenuated the protective effect of curcumin against MeHg-induced cell death. However, both the pan-protein kinase C (PKC) inhibitor, Ro 31-8220, and the selective PKCδ inhibitor, rottlerin, failed to suppress the curcumin-activated Nrf2/Antioxidant Response Element(ARE) pathway and attenuate the protection exerted by curcumin. Taken together, these findings confirm that curcumin protects against MeHg-induced neurotoxicity by activating the Nrf2/ARE pathway and this protection is independent of PKCδ activation. More studies are needed to understand the mechanisms of curcumin cytoprotection.
Collapse
Affiliation(s)
- Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Changsheng Yin
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yun Zhou
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Qiang Wang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yuanyue Jiang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yu Bai
- 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
| | - Yun Feng
- Department of Pharmacology, 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
| | - Jiyang Cai
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77550-1106, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - 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 215132, China.
| |
Collapse
|
49
|
Freire MAM, Santana LNS, Bittencourt LO, Nascimento PC, Fernandes RM, Leão LKR, Fernandes LMP, Silva MCF, Amado LL, Gomes-Leal W, Crespo-Lopez ME, Maia CDSF, Lima RR. Methylmercury intoxication and cortical ischemia: Pre-clinical study of their comorbidity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 174:557-565. [PMID: 30865911 DOI: 10.1016/j.ecoenv.2019.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 06/09/2023]
Abstract
Stroke is one of the main causes of human disability worldwide. Ischemic stroke is mostly characterized by metabolic collapse and fast tissue damage, followed by secondary damage in adjacent regions not previously affected. Heavy metals intoxication can be associated with stroke incidence, because of their damaging action in the vascular system. Mercury, in particular, possesses a high tropism by metabolically active regions, such as the brain. In the present study we sought to evaluate whether methylmercury (MeHg) intoxication can aggravate the tissue damage caused by an ischemic stroke induced by microinjections of endothelin-1 (ET-1) into the motor cortex of adult rats. Following MeHg intoxication by gavage (0.04 mg/kg/day) during 60 days, the animals were injected with ET-1 (1 μl, 40 pmol/μl) or vehicle (1 μl). After 7 days, all animals were submitted to behavioral tests and then their brains were processed to biochemical and immunohistochemical analyses. We observed that long-term MeHg intoxication promoted a significant Hg deposits in the motor cortex, with concomitant increase of microglial response, followed by reduction of the neuronal population following ischemia and MeHg intoxication, as well as disturbance in the antioxidant defense mechanisms by misbalance of oxidative biochemistry with increase of both lipid peroxidation and nitrite levels, associated to behavioral deficits. MeHg exposure and cortical ischemia demonstrated that both injuries are able of causing significant neurobehavioural impairments in motor coordination and learning accompanied of an exacerbated microglial activation, oxidative stress and neuronal loss in the motor cortex, indicating that MeHg as a source of metabolic disturbance can act as an important increasing factor of ischemic events in the brain.
Collapse
Affiliation(s)
| | - Luana Nazaré S Santana
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Priscila Cunha Nascimento
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Luana Ketlen R Leão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Luanna Melo P Fernandes
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Marcia Cristina F Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Lílian Lund Amado
- Laboratory of Ecotoxicology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Walace Gomes-Leal
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Cristiane do Socorro F Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil.
| |
Collapse
|
50
|
Insights into the Potential Role of Mercury in Alzheimer's Disease. J Mol Neurosci 2019; 67:511-533. [PMID: 30877448 DOI: 10.1007/s12031-019-01274-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/30/2019] [Indexed: 12/18/2022]
Abstract
Mercury (Hg), which is a non-essential element, is considered a highly toxic pollutant for biological systems even when present at trace levels. Elevated Hg exposure with the growing release of atmospheric pollutant Hg and rising accumulations of mono-methylmercury (highly neurotoxic) in seafood products have increased its toxic potential for humans. This review aims to highlight the potential relationship between Hg exposure and Alzheimer's disease (AD), based on the existing literature in the field. Recent reports have hypothesized that Hg exposure could increase the potential risk of developing AD. Also, AD is known as a complex neurological disorder with increased amounts of both extracellular neuritic plaques and intracellular neurofibrillary tangles, which may also be related to lifestyle and genetic variables. Research reports on AD and relationships between Hg and AD indicate that neurotransmitters such as serotonin, acetylcholine, dopamine, norepinephrine, and glutamate are dysregulated in patients with AD. Many researchers have suggested that AD patients should be evaluated for Hg exposure and toxicity. Some authors suggest further exploration of the Hg concentrations in AD patients. Dysfunctional signaling pathways in AD and Hg exposure appear to be interlinked with some driving factors such as arachidonic acid, homocysteine, dehydroepiandrosterone (DHEA) sulfate, hydrogen peroxide, glucosamine glycans, glutathione, acetyl-L carnitine, melatonin, and HDL. This evidence suggests the need for a better understanding of the relationship between AD and Hg exposure, and potential mechanisms underlying the effects of Hg exposure on regional brain functions. Also, further studies evaluating brain functions are needed to explore the long-term effects of subclinical and untreated Hg toxicity on the brain function of AD patients.
Collapse
|