1
|
Riedel JA, Smolina I, Donat C, Svendheim LH, Farkas J, Hansen BH, Olsvik PA. Into the deep: Exploring the molecular mechanisms of hyperactive behaviour induced by three rare earth elements in early life-stages of the deep-sea scavenging amphipod Tmetonyx cicada (Lysianassidae). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175968. [PMID: 39226952 DOI: 10.1016/j.scitotenv.2024.175968] [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: 06/29/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
With increasing socio-economic importance of the rare earth elements and yttrium (REY), Norway has laid out plans for REY mining, from land-based to deep-sea mining, thereby enhancing REY mobility in the marine ecosystem. Little is known about associated environmental consequences, especially in the deep ocean. We explored the toxicity and modes of action of a light (Nd), medium (Gd) and heavy (Yb) REY-Cl3 at four concentrations (3, 30, 300, and 3000 μg L-1) in the Arcto-boreal deep-sea amphipod Tmetonyx cicada. At the highest concentration, REY solubility was limited and increased with atomic weight (Nd < Gd < Yb). Lethal effects were practically restricted to this treatment, with the lighter elements being more acutely toxic than Yb (from ∼50 % mortality in the Gd-group at dissolved 689-504 μg L-1 to <20 % in the Yb-group at ca. 2000 μg L-1), which could be a function of bioavailability. All three REY induced hyperactivity at the low-medium concentrations. Delving into the transcriptome of T. cicada allowed us to determine a whole array of potential (neurotoxic) mechanisms underlying this behaviour. Gd induced the vastest response, affecting serotonin-synthesis; sphingolipid-synthesis; the renin-angiotensin system; mitochondrial and endoplasmic reticulum functioning (Gd, Nd); and lysosome integrity (Gd, Yb); as well as the expression of hemocyanin, potentially governing REY-uptake (Gd, Yb). While Nd and Yb shared only few pathways, suggesting a link between mode of action and atomic weight/radius, almost all discussed mechanisms imply the disruption of organismal Ca-homeostasis. Despite only fragmental genomic information available for crustaceans to date, our results provide novel insight into the toxicophysiology of REY in marine biota. The neurotoxic/behavioural effects in T. cicada at concentrations with potential environmental relevance warn about the possibility of bottom-up ecological consequences in mining exposed fjords and deep-sea ecosystems, calling for follow-up studies and regulatory measures prior to the onset of REY mining in Norway.
Collapse
Affiliation(s)
- Juliane Annemieke Riedel
- Faculty of Biosciences and Aquaculture, Nord University, Universitetsalléen 11, 8026 Bodø, Norway.
| | - Irina Smolina
- Faculty of Biosciences and Aquaculture, Nord University, Universitetsalléen 11, 8026 Bodø, Norway
| | - Coline Donat
- IUT de Saint Étienne, Université Jean Monnet, 28 Av. Léon Jouhaux, 42100 Saint-Étienne, France
| | | | - Julia Farkas
- Department of Climate and Environment, SINTEF Ocean, Brattørkaia 17C, 7010 Trondheim, Norway
| | - Bjørn Henrik Hansen
- Department of Climate and Environment, SINTEF Ocean, Brattørkaia 17C, 7010 Trondheim, Norway
| | - Pål Asgeir Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Universitetsalléen 11, 8026 Bodø, Norway
| |
Collapse
|
2
|
Silva MG, Martinez CG, Cavalcanti de Albuquerque JP, Gouvêa AL, Freire MM, Lauthartte LC, Mignaco J, Bastos WR, de Mattos EC, Galina A, Kurtenbach E. Mitochondrial Dysfunction Plays a Relevant Role in Heart Toxicity Caused by MeHg. TOXICS 2024; 12:712. [PMID: 39453132 PMCID: PMC11511492 DOI: 10.3390/toxics12100712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 10/26/2024]
Abstract
The effects of methylmercury (MeHg) on exposed populations are a public health problem. In contrast to widely studied neurological damage, few cardiovascular changes have been described. Our group evaluated the cardiotoxicity of a cumulative dose of 70 mg.kg-1 fractioned over a 14-day exposure period in mice (MeHg70 group). The effects of MeHg on proteins relevant to cardiac mitochondrial function were also investigated. The results obtained showed a reduction in oxygen consumption in the two settings. In cardiac tissue samples in oxygraphy studies, this reduction was related to a lower efficiency of complexes II and V, which belong to the oxidative phosphorylation system. In vivo, mice in the MeHg70 group presented lower oxygen consumption and running tolerance, as shown by ergometric analyses. Cardiac stress was evident in the MeHg70 group, as indicated by a marked increase in the level of the mRNA encoding atrial natriuretic peptide. Electrocardiogram studies revealed a lower heart rate at rest in the animals from the MeHg70 group, as well as prolonged left ventricular depolarisation and repolarisation. Through echocardiographic analysis, reductions in the left ventricular ejection fraction and left ventricular wall thickness of approximately 10% and 20%, respectively, were detected. These results indicate that the oral intake of MeHg can decrease cardiac function and oxidative metabolism. This finding highlights the importance of monitoring MeHg levels in humans and animals in contaminated areas, as well as periodically carrying out cardiac function tests.
Collapse
Affiliation(s)
- Marcia Gracindo Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (A.L.G.); (E.K.)
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Camila Guerra Martinez
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (A.L.G.); (E.K.)
| | | | - André Luiz Gouvêa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (A.L.G.); (E.K.)
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Monica Maria Freire
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Leidiane Caroline Lauthartte
- Laboratório de Biogeoquímica Ambiental Wolfgang C. Pfeiffer, Universidade Federal de Rondônia, Porto Velho 76801-974, RO, Brazil
| | - Julio Mignaco
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Wanderley Rodrigues Bastos
- Laboratório de Biogeoquímica Ambiental Wolfgang C. Pfeiffer, Universidade Federal de Rondônia, Porto Velho 76801-974, RO, Brazil
| | | | - Antonio Galina
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Eleonora Kurtenbach
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (A.L.G.); (E.K.)
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
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
|
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
|
5
|
Li B, Jin X, Chan HM. Effects of low doses of methylmercury (MeHg) exposure on definitive endoderm cell differentiation in human embryonic stem cells. Arch Toxicol 2023; 97:2625-2641. [PMID: 37612375 PMCID: PMC10475006 DOI: 10.1007/s00204-023-03580-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 08/25/2023]
Abstract
Fetal development is one of the most sensitive windows to methylmercury (MeHg) toxicity. Laboratory and epidemiological studies have shown a dose-response relationship between fetal MeHg exposure and neuro performance in different life stages from infants to adults. In addition, MeHg exposure has been reported to be associated with disorders in endoderm-derived organs, such as morphological changes in liver cells and pancreatic cell dysfunctions. However, the mechanisms of the effects of MeHg on non-neuronal organs or systems, especially during the early development of endoderm-derived organs, remain unclear. Here we determined the effects of low concentrations of MeHg exposure during the differentiation of definitive endoderm (DE) cells from human embryonic stem cells (hESCs). hESCs were exposed to MeHg (0, 10, 100, and 200 nM) that covers the range of Hg concentrations typically found in human maternal blood during DE cell induction. Transcriptomic analysis showed that sub-lethal doses of MeHg exposure could alter global gene expression patterns during hESC to DE cell differentiation, leading to increased expression of endodermal genes/proteins and the over-promotion of endodermal fate, mainly through disrupting calcium homeostasis and generating ROS. Bioinformatic analysis results suggested that MeHg exerts its developmental toxicity mainly by disrupting ribosome biogenesis during early cell lineage differentiation. This disruption could lead to aberrant growth or dysfunctions of the developing endoderm-derived organs, and it may be the underlying mechanism for the observed congenital diseases later in life. Based on the results, we proposed an adverse outcome pathway for the effects of MeHg exposure during human embryonic stem cells to definitive endoderm differentiation.
Collapse
Affiliation(s)
- Bai Li
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, K1N 6N5, Canada
| | - Xiaolei Jin
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, 251 Sir Frederick Banting Driveway, Ottawa, ON, K1A 0K9, Canada.
| | - Hing Man Chan
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
6
|
Yang D, Chen M, Yang S, Deng F, Guo X. Connexin hemichannels and pannexin channels in toxicity: Recent advances and mechanistic insights. Toxicology 2023; 488:153488. [PMID: 36918108 DOI: 10.1016/j.tox.2023.153488] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023]
Abstract
Connexin hemichannels and pannexin channels are two types of transmembrane channels that allow autocrine/paracrine signalling through the exchange of ions and molecules between the intra- and extracellular compartments. However, owing to the poor selectivity of permeable ions and metabolites, the massive opening of these plasma membrane channels can lead to an excessive influx of toxic substances and an outflux of essential metabolites, such as adenosine triphosphate, glutathione, glutamate and ions, resulting in unbalanced cell homeostasis and impaired cell function. It is becoming increasingly clear that these channels can be activated in response to external stimuli and are involved in toxicity, yet their concrete mechanistic roles in the toxic effects induced by stress and various environmental changes remain poorly defined. This review provides an updated understanding of connexin hemichannels and pannexin channels in response to multiple extrinsic stressors and how these activated channels and their permeable messengers participate in toxicological pathways and processes, including inflammation, oxidative damage, intracellular calcium imbalance, bystander DNA damage and excitotoxicity.
Collapse
Affiliation(s)
- Di Yang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Mengyuan Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Sijia Yang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China.
| |
Collapse
|
7
|
Zuo K, Xu Q, Wang Y, Sui Y, Niu Y, Liu Z, Liu M, Liu X, Liu D, Sun W, Wang Z, Liu X, Liu J. L-Ascorbic Acid 2-Phosphate Attenuates Methylmercury-Induced Apoptosis by Inhibiting Reactive Oxygen Species Accumulation and DNA Damage in Human SH-SY5Y Cells. TOXICS 2023; 11:144. [PMID: 36851019 PMCID: PMC9967424 DOI: 10.3390/toxics11020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Methylmercury (MeHg) is a toxin that causes severe neuronal oxidative damage. As vitamin C is an antioxidant well-known to protect neurons from oxidative damage, our goal was to elucidate its protective mechanism against MeHg-induced oxidative stress in human neuroblastomas (SHSY5Y). We treated cells with MeHg, L-ascorbic acid 2-phosphate (AA2P), or both, and used MTT, flow cytometry, and Western blot analyses to assess cell damage. We found that MeHg significantly decreased the survival rate of SH-SY5Y cells in a time- and dose-dependent manner, increased apoptosis, downregulated PAR and PARP1 expression, and upregulated AIF, Cyto C, and cleaved Caspase-3 expression. A time course study showed that MeHg increased reactive oxygen species (ROS) accumulation; enhanced apoptosis; increased DNA damage; upregulated expression ofγH2A.X, KU70, 67 and 57 kDa AIF, CytoC, and cleaved Caspase-3; and downregulated expression of 116 kDa PARP1, PAR, BRAC1, and Rad51. Supplementation with AA2P significantly increased cell viability and decreased intrinsic ROS accumulation. It also reduced ROS accumulation in cells treated with MeHg and decreased MeHg-induced apoptosis. Furthermore, AA2P conversely regulated gene expression compared to MeHg. Collectively, we demonstrate that AA2P attenuates MeHg-induced apoptosis by alleviating ROS-mediated DNA damage and is a potential treatment for MeHg neurotoxicity.
Collapse
Affiliation(s)
- Kuiyang Zuo
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Qi Xu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Yujie Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Yutong Sui
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Ye Niu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Zinan Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Mingsheng Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Xinpeng Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Dan Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Wei Sun
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Ziyu Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Xiaomei Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| |
Collapse
|
8
|
Almikhlafi MA, Karami MM, Jana A, Alqurashi TM, Majrashi M, Alghamdi BS, Ashraf GM. Mitochondrial Medicine: A Promising Therapeutic Option Against Various Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:1165-1183. [PMID: 36043795 PMCID: PMC10286591 DOI: 10.2174/1570159x20666220830112408] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/05/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022] Open
Abstract
Abnormal mitochondrial morphology and metabolic dysfunction have been observed in many neurodegenerative disorders (NDDs). Mitochondrial dysfunction can be caused by aberrant mitochondrial DNA, mutant nuclear proteins that interact with mitochondria directly or indirectly, or for unknown reasons. Since mitochondria play a significant role in neurodegeneration, mitochondriatargeted therapies represent a prosperous direction for the development of novel drug compounds that can be used to treat NDDs. This review gives a brief description of how mitochondrial abnormalities lead to various NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. We further explore the promising therapeutic effectiveness of mitochondria- directed antioxidants, MitoQ, MitoVitE, MitoPBN, and dimebon. We have also discussed the possibility of mitochondrial gene therapy as a therapeutic option for these NDDs.
Collapse
Affiliation(s)
- Mohannad A. Almikhlafi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Mohammed M. Karami
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Thamer M. Alqurashi
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Majrashi
- Department of Pharmacology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| |
Collapse
|
9
|
Zhang Y, Chen S, Fan F, Xu N, Meng XL, Zhang Y, Lin JM. Neurotoxicity mechanism of aconitine in HT22 cells studied by microfluidic chip-mass spectrometry. J Pharm Anal 2023; 13:88-98. [PMID: 36820076 PMCID: PMC9937797 DOI: 10.1016/j.jpha.2022.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Aconitine, a common and main toxic component of Aconitum, is toxic to the central nervous system. However, the mechanism of aconitine neurotoxicity is not yet clear. In this work, we had the hypothesis that excitatory amino acids can trigger excitotoxicity as a pointcut to explore the mechanism of neurotoxicity induced by aconitine. HT22 cells were simulated by aconitine and the changes of target cell metabolites were real-time online investigated based on a microfluidic chip-mass spectrometry system. Meanwhile, to confirm the metabolic mechanism of aconitine toxicity on HT22 cells, the levels of lactate dehydrogenase, intracellular Ca2+, reactive oxygen species, glutathione and superoxide dismutase, and ratio of Bax/Bcl-2 protein were detected by molecular biotechnology. Integration of the detected results revealed that neurotoxicity induced by aconitine was associated with the process of excitotoxicity caused by glutamic acid and aspartic acid, which was followed by the accumulation of lactic acid and reduction of glucose. The surge of extracellular glutamic acid could further lead to a series of cascade reactions including intracellular Ca2+ overload and oxidative stress, and eventually result in cell apoptosis. In general, we illustrated a new mechanism of aconitine neurotoxicity and presented a novel analysis strategy that real-time online monitoring of cell metabolites can provide a new approach to mechanism analysis.
Collapse
Affiliation(s)
- Yingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China,Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shiyu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China,Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Fangfang Fan
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ning Xu
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xian-Li Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China,Corresponding author.
| | - Jin-Ming Lin
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China,Corresponding author.
| |
Collapse
|
10
|
Augustyniak J, Lipka G, Kozlowska H, Caloni F, Buzanska L. Oxygen as an important factor modulating in vitro MeHgCl toxicity associated with mitochondrial genes in hiPSCs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113737. [PMID: 35696963 DOI: 10.1016/j.ecoenv.2022.113737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Mitochondria are energy factories of cells and important targets for methylmercury chloride (MgHgCl). Methylmercury (MeHg) is a well-known environmental toxicant that bioaccumulates in fish and shellfish. It readily crosses the placental barrier, making it a threat to correct fetal development. Despite being comprehensively investigated for years, this compound has not been assessed for its in vitro mitochondrial toxicity under different oxygen conditions. In this study, human induced pluripotent stem cells (hiPSCs) were used to evaluate the dependence of the expression of genes associated with pluripotency and mitochondria on atmospheric (21% O2) and low (5% O2) oxygen concentrations upon MeHgCl treatment. We showed that the toxicity of MeHgCl was strongly related to an increased mtDNA copy number and downregulation of the expression of an mtDNA replication and damage repair-associated gene POLG1 (Mitochondrial Polymerase Gamma Catalytic Subunit) in both tested oxygen conditions. In addition, the viability and mitochondrial membrane potential of hiPSCs were significantly lowered by MeHgCl regardless of the oxygen concentration. However, reactive oxygen species accumulation significantly increased only under atmospheric oxygen conditions; what was associated with increased expression of TFAM (Transcription Factor A, Mitochondrial) and NRF1 (Nuclear Respiratory Factor 1) and downregulation of PARK2 (Parkin RBR E3 Ubiquitin Protein Ligase). Taken together, our results demonstrated that MeHgCl could induce in vitro toxicity in hiPSCs through altering mitochondria-associated genes in an oxygen level-dependent manner. Thus, our work suggests that oxygen should be considered a factor was modulating the in vitro toxicity of environmental pollutants. Typical atmospheric conditions of in vitro culture significantly lower the predictive value of studies of such toxicity.
Collapse
Affiliation(s)
- J Augustyniak
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - G Lipka
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - H Kozlowska
- Laboratory of Advanced Microscopy Technique, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - F Caloni
- Department of Environmental Science and Policy (ESP), Università degli Studi di Milano, Milan, Italy
| | - L Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
11
|
Pan J, Wei Y, Ni L, Li X, Deng Y, Xu B, Yang T, Sun J, Liu W. Unbalanced ER-mitochondrial calcium homeostasis promotes mitochondrial dysfunction and associated apoptotic pathways activation in methylmercury exposed rat cortical neurons. J Biochem Mol Toxicol 2022; 36:e23136. [PMID: 35678294 DOI: 10.1002/jbt.23136] [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: 11/16/2021] [Revised: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 11/06/2022]
Abstract
Methylmercury (MeHg) is a cumulative environmental pollutant that can easily cross the blood-brain barrier and cause damage to the brain, mainly targeting the central nervous system. The purpose of this study is to investigate the role of calcium ion (Ca2+ ) homeostasis between the endoplasmic reticulum (ER) and mitochondria in MeHg-induced neurotoxicity. Rat primary cortical neurons exposed to MeHg (0.25-1 μm) underwent dose-dependent cell damage, accompanied by increased Ca2+ release from the ER and elevated levels of free Ca2+ in cytoplasm and mitochondria. MeHg also increased the protein and messenger RNA expressions of the inositol 1,4,5-triphosphate receptor, ryanodine receptor 2, and mitochondrial calcium uniporter. Ca2+ channel inhibitors 2-aminoethyl diphenylborinate and procaine reduced the release of Ca2+ from ER, while RR and 4,4'-diisothiocyanatostilbene-2,2'-disulfonate inhibited Ca2+ uptake from mitochondria. In addition, pretreatment with Ca2+ chelator BAPTA-AM effectively restored mitochondrial membrane potential levels, inhibited over opening of mitochondrial permeability transition pore, and maintained mitochondrial function stability. Meanwhile, the expression of mitochondrial apoptosis-related proteins recovered to some extent, along with the reduction of the early apoptosis ratio. These results suggest that Ca2+ homeostasis plays an essential role in mitochondrial damage and apoptosis induced by MeHg, which may be one of the important mechanisms of MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Jingyi Sun
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
12
|
Wildner G, Loreto JS, de Almeida P, Claro MT, Ferreira SA, Barbosa NV. Short exposure to ethyl and methylmercury prompts similar toxic responses in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 2022; 252:109216. [PMID: 34710619 DOI: 10.1016/j.cbpc.2021.109216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 12/30/2022]
Abstract
Methylmercury (MeHg) and ethylmercury (EtHg) are important mercury organic forms in terms of human poisoning. Since the comparative effects of compounds are mainly in vitro, this study was designed to investigate the toxicities induced by MeHg and EtHg in an in vivo study using adult Drosophila melanogaster (D. melanogaster). Firstly, we performed a survival curve, where the flies were fed on a medium containing MeHg and EtHg at concentrations ranging from 2.5 to 200 μM, until the end of their lifespan. After that, the concentrations 25 and 200 μM of MeHg and EtHg were chosen to be tested in a short exposure for 5 days. The analysis of survival by Kaplan-Meier plot revealed that all concentrations of MeHg and EtHg reduced significantly the lifespan of the flies. Short exposure to both concentrations of MeHg and EtHg impaired the ability of flies in the climbing assay and induced lipid peroxidation. Only the flies exposed to the highest concentration had viability loss, thiol depletion, and increased reactive species (RS) and Hg levels in the whole body. Our findings indicate that MeHg and EtHg exhibit similar toxic effects in vivo, and that oxidative stress is a phenomenon behind the toxicity of both mercurials. The data obtained also reinforce the use of D. melanogaster as a useful organism for basic toxicological research.
Collapse
Affiliation(s)
- Guilherme Wildner
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900 Santa Maria, RS, Brazil
| | - Julia Sepel Loreto
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900 Santa Maria, RS, Brazil
| | - Pamela de Almeida
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900 Santa Maria, RS, Brazil
| | - Mariana Torri Claro
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900 Santa Maria, RS, Brazil
| | - Sabrina Antunes Ferreira
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900 Santa Maria, RS, Brazil
| | - Nilda Vargas Barbosa
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900 Santa Maria, RS, Brazil.
| |
Collapse
|
13
|
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
|
14
|
The Role of Human LRRK2 in Acute Methylmercury Toxicity in Caenorhabditis elegans. Neurochem Res 2021; 46:2991-3002. [PMID: 34272628 DOI: 10.1007/s11064-021-03394-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
Methylmercury (MeHg) exposure and its harmful effects on the developing brain continue to be a global environmental health concern. Decline in mitochondrial function is central to the toxic effects of MeHg and pathogenesis of mitochondria-related diseases including Parkinson's disease (PD). LRRK2 (Leucine-rich repeat kinase 2) mutation is one of the most common genetic risk factors for PD. In this study, we utilize an acute toxicity model of MeHg exposure in the model organism Caenorhabditis elegans (C. elegans) to compare lifespan, developmental progression, mitochondrial membrane potential and reactive oxygen species (ROS) between the wild-type N2 strain, wild-type LRRK2 transgenic strain (WLZ1), and mutant LRRK2(G2019S) transgenic strain (WLZ3). Additionally, the expression levels of skn-1 and gst-4 were investigated. Our results show that acute MeHg exposure (5 and 10 µM) caused a significant developmental delay in the N2 and WLZ3 worms. Notably, the worms expressing wild-type LRRK2 were resistant to 5 µM MeHg- induced developmental retardation. ROS levels in response to MeHg exposure were increased in the N2 worms, but not in the WLZ1 or WLZ3 worms. The mitochondrial membrane potential was decreased in the N2 worms but increased in the WLZ1 and WLZ3 worms following MeHg exposure. Furthermore, MeHg exposure increased the expression of skn-1 in N2, but not in WLZ1 worms. Although skn-1 expression was increased in the WLZ3 worms following MeHg exposure, gst-4 expression was not induced. Both skn-1 and gst-4 had higher basal expression levels in LRRK2s transgenic than wild-type N2 worms. Knocking down of skn-1 with feeding RNAi had a significant developmental effect in WLZ1 worms; however, the effect was not found in WLZ3 worms. These results suggest that mitochondrial dysfunction and a defect in the SKN-1 signaling in the LRRK2 G2019S worms contribute to the severe developmental delay, establishing a modulatory role of LRRK2 mutation in MeHg-induced acute toxicity.
Collapse
|
15
|
Hernández-Fernández J, Pinzón-Velasco A, López EA, Rodríguez-Becerra P, Mariño-Ramírez L. Transcriptional Analyses of Acute Exposure to Methylmercury on Erythrocytes of Loggerhead Sea Turtle. TOXICS 2021; 9:70. [PMID: 33805397 PMCID: PMC8066450 DOI: 10.3390/toxics9040070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 01/09/2023]
Abstract
To understand changes in enzyme activity and gene expression as biomarkers of exposure to methylmercury, we exposed loggerhead turtle erythrocytes (RBCs) to concentrations of 0, 1, and 5 mg L-1 of MeHg and de novo transcriptome were assembled using RNA-seq. The analysis of differentially expressed genes (DEGs) indicated that 79 unique genes were dysregulated (39 upregulated and 44 downregulated genes). The results showed that MeHg altered gene expression patterns as a response to the cellular stress produced, reflected in cell cycle regulation, lysosomal activity, autophagy, calcium regulation, mitochondrial regulation, apoptosis, and regulation of transcription and translation. The analysis of DEGs showed a low response of the antioxidant machinery to MeHg, evidenced by the fact that genes of early response to oxidative stress were not dysregulated. The RBCs maintained a constitutive expression of proteins that represented a good part of the defense against reactive oxygen species (ROS) induced by MeHg.
Collapse
Affiliation(s)
- Javier Hernández-Fernández
- Department of Natural and Environmental Science, Marine Biology Program, Faculty of Science and Engineering, Genetics, Molecular Biology and Bioinformatic Research Group–GENBIMOL, Jorge Tadeo Lozano University, Cra. 4 No 22-61, Bogotá 110311, Colombia;
- Faculty of Sciences, Department of Biology, Pontificia Universidad Javeriana, Calle 45, Cra. 7, Bogotá 110231, Colombia
| | - Andrés Pinzón-Velasco
- Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Calle 45, Cra. 30, Bogotá 111321, Colombia;
| | - Ellie Anne López
- IDEASA Research Group-Environment and Sustainability, Institute of Environmental Studies and Services, Sergio Arboleda University, Bogotá 111711, Colombia;
| | - Pilar Rodríguez-Becerra
- Department of Natural and Environmental Science, Marine Biology Program, Faculty of Science and Engineering, Genetics, Molecular Biology and Bioinformatic Research Group–GENBIMOL, Jorge Tadeo Lozano University, Cra. 4 No 22-61, Bogotá 110311, Colombia;
| | - Leonardo Mariño-Ramírez
- NCBI, NLM, NIH Computational Biology Branch, Building 38A, Room 6S614M 8600 Rockville Pike, MSC 6075, Bethesda, MD 20894-6075, USA;
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Chen N, Tang X, Ye Z, Wang S, Xiao X. Methylmercury disrupts autophagic flux by inhibiting autophagosome-lysosome fusion in mouse germ cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 198:110667. [PMID: 32339925 DOI: 10.1016/j.ecoenv.2020.110667] [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: 10/11/2019] [Revised: 04/03/2020] [Accepted: 04/19/2020] [Indexed: 06/11/2023]
Abstract
Methylmercury (MeHg) is an extremely toxic environmental pollutant that can cause serious male reproductive developmental dysplasia in humans and animals. However, the molecular mechanisms underlying MeHg-induced male reproductive injury are not fully clear. The purpose of this study was to explore whether mitophagy and lysosome dysfunction contribute to MeHg-induced apoptosis of germ cell and to determine the potential mechanism. First, we confirmed the exposure of GC2-spd cells to mercury. In GC2-spd cells (a mouse spermatocyte cell line), we found that MeHg treatment led to an obvious increase of cell apoptosis accompanied by a marked rise of LC3-II expression and an elevated number of autophagosomes. These results were associated with the induction of oxidative stress and mitophagy. Interestingly, we found that MeHg did not promote but prevented autophagosome-lysosome fusion by impairing the lysosome function. Furthermore, as a lysosome inhibitor, chloroquine pre-treatment obviously enhanced LC3-II expression and mitophagy formation in MeHg-treated cells. This further proved that the induction of mitophagy and the injury of the lysosome played an important role in the GC2-spd cell apoptosis induced by MeHg. Our findings indicate that MeHg caused apoptosis in the GC2-spd cells, which were dependent on oxidative stress-mediated mitophagy and the lysosome damaging-mediated inhibition of autophagic flux induced by MeHg.
Collapse
Affiliation(s)
- Na Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiaofeng Tang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Zhaoyang Ye
- Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Shanshan Wang
- Key Laboratory of Agro-product Safety and Quality, Institute of Quality Standards & Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, 100081, PR China.
| | - Xianjin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
19
|
Pérez CA, Shah EG, Butler IJ. Mercury-induced autoimmunity: Report of two adolescent siblings with Morvan syndrome "plus" and review of the literature. J Neuroimmunol 2020; 342:577197. [PMID: 32126315 DOI: 10.1016/j.jneuroim.2020.577197] [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: 12/21/2019] [Revised: 02/08/2020] [Accepted: 02/21/2020] [Indexed: 02/06/2023]
Abstract
Heavy metal toxicity is a global health concern. Mercury intoxication has been implicated in the etiology and pathogenesis of autoimmune disease, including Morvan syndrome. We describe two siblings with overlapping features of distinct autoimmune syndromes following accidental exposure to elemental mercury. Morvan syndrome was the predominant clinical phenotype. In addition to the characteristic anti-leucine-rich glioma-inactivated protein 1 (LGI1) and anti-contactin-associated protein-like 2 (Caspr2) autoantibodies, glutamic acid decarboxylase 65-kilodalton isoform (GAD65), and N-type and P/Q-type voltage-gated calcium channel (VGCC) antibodies were detected. Treatment with chelation therapy, glucocorticoids, and intravenous immunoglobulin was unsuccessful, but complete resolution of symptoms was achieved following treatment with rituximab. Herein, we perform an extensive review of the literature with a focus on the emerging concepts of mercury-induced autoimmunity and the role of mercury in the etiopathogenesis of autoimmune diseases of the nervous system.
Collapse
Affiliation(s)
- Carlos A Pérez
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Ekta G Shah
- Division of Child and Adolescent Neurology, Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ian J Butler
- Division of Child and Adolescent Neurology, Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
20
|
Rzajew J, Radzik T, Rebas E. Calcium-Involved Action of Phytochemicals: Carotenoids and Monoterpenes in the Brain. Int J Mol Sci 2020; 21:ijms21041428. [PMID: 32093213 PMCID: PMC7073062 DOI: 10.3390/ijms21041428] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Neurodegenerative and mood disorders represent growing medical and social problems, many of which are provoked by oxidative stress, disruption in the metabolism of various neurotransmitters, and disturbances in calcium homeostasis. Biologically active plant compounds have been shown to exert a positive impact on the function of calcium in the central nervous system. Methods: The present paper reviews studies of naturally occurring terpenes and derivatives and the calcium-based aspects of their mechanisms of action, as these are known to act upon a number of targets linked to neurological prophylaxis and therapy. Results: Most of the studied phytochemicals possess anticancer, antioxidative, anti-inflammatory, and neuroprotective properties, and these have been used to reduce the risk of or treat neurological diseases. Conclusion: The neuroprotective actions of some phytochemicals may employ mechanisms based on regulation of calcium homeostasis and should be considered as therapeutic agents.
Collapse
|
21
|
Ke T, Tsatsakis A, Santamaría A, Antunes Soare FA, Tinkov AA, Docea AO, Skalny A, Bowman AB, Aschner M. Chronic exposure to methylmercury induces puncta formation in cephalic dopaminergic neurons in Caenorhabditis elegans. Neurotoxicology 2020; 77:105-113. [PMID: 31935438 DOI: 10.1016/j.neuro.2020.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 01/09/2023]
Abstract
The neurotransmitter dopamine is a neuromodulator in the positive and negative regulation of brain circuits. Dopamine insufficiency or overload has been implicated in aberrant activities of neural circuits that play key roles in the pathogenesis of neurological and psychiatric diseases. Dopaminergic neurons are vulnerable to environmental insults. The neurotoxin methylmercury (MeHg) produces dopaminergic neuron damage in rodent as well as in Caenorhabditis elegans (C. elegans) models. Previous studies have demonstrated the utility of C. elegans as an alternative and complementary experimental model in dissecting out mechanism of MeHg-induced dopaminergic neurodegeneration. However, a sensitive pathological change that marks early events in neurodegeneration induced by environmental level of MeHg, is still lacking. By establishing a chronic exposure C. elegans model, for the first time, we have shown the propensity of MeHg (5 μM, 10 days) to induce bright puncta of dat-1::mCherry aggreagtes in the dendrites of cephalic (2 CEPs) dopaminergic neurons in a dose- and time-dependent manner, while these changes were not found in other dopaminergic neurons: anterior deirids (2 ADEs) and posterior deirids (2 PDEs), cholinergic neurons (2 AIYs) or glutamatergic neurons (2 PVDs). The bright puncta appear as an aggregation of mCherry proteins accumulating in dendrites. Further staining shows that the puncta were not inclusions in lysosome, or amyloid protein aggregates. In addition, features of the puncta including enlarged sphere shape (0.5-2 μm diameters), bright and accompanying with the shrinkage of the dendrite suggest that the puncta are likely composed of homologous mCherry molecules packaged at the dendritic site for exportation. Moreover, in the glutathione S-transferase 4 (gst-4) transcriptional reporter strain and RT-PCR assay, the expression levels of gst-4 and tubulins (tba-1 and tba-2) genes were not significantly modified under this chronic exposure paradigm, but gst-4 did show significant changes in an one day exposure paradigm. Collectively, these results suggest that CEP dopaminergic neurons are a sensitive target of MeHg, and the current exposure paradigm could be used as a model to investigate mechanism of dopaminergic neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Aristidis Tsatsakis
- Laboratory of Toxicology and Forensic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico
| | - Félix Alexandre Antunes Soare
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Alexey A Tinkov
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy, Faculty of Pharmacy, Craiova, 200349, Romania
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia.
| |
Collapse
|
22
|
Lin T, Ruan S, Huang D, Meng X, Li W, Wang B, Zou F. MeHg-induced autophagy via JNK/Vps34 complex pathway promotes autophagosome accumulation and neuronal cell death. Cell Death Dis 2019; 10:399. [PMID: 31113939 PMCID: PMC6529499 DOI: 10.1038/s41419-019-1632-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/21/2019] [Accepted: 05/06/2019] [Indexed: 12/20/2022]
Abstract
Methylmercury (MeHg), an environmental toxin, may specifically cause neurological disorders. Recent studies have reported that autophagy can be induced by metals and be involved in metal cytotoxicity. However, the role of autophagy in MeHg-induced neurotoxicity remains unknown. Here, we demonstrate that MeHg induces mTOR-independent autophagy through JNK/Vps34 complex pathway, which further promotes autophagosome accumulation and neuronal cell death. In addition to cell death, MeHg increased LC3-II expression in a concentration- and time-dependent manner in neuronal cells; furthermore, western blot analysis of LC3-II expression under baf A1-treated condition indicates that MeHg activates autophagy induction. However, we found lysosomal degradative function was impaired by MeHg. Under this condition, MeHg-activated autophagy induction would elicit autophagosome accumulation and cell death. Consistent with this inference, the autophagy inhibitor decreased the MeHg-induced autophagosome accumulation and neuronal cells death, whereas the autophagy inducers further augmented MeHg cytotoxicity. Then, the mechanism of autophagy induction is investigated. We show that MeHg-induced autophagy is mTOR-independent. Vacuolar protein sorting 34 (Vps34) complex is critical for mTOR-independent autophagy. MeHg induced the interaction between Beclin1 and Vps34 to form Vps34 complex. Importantly, knockdown of Vps34 inhibited autophagy induction by MeHg. Furthermore, we found that JNK, but not p38 or ERK, promoted the formation of Vps34 complex and autophagy induction. Finally, inhibition of JNK or downregulation of Vps34 decreased autophagosome accumulation and alleviated MeHg-induced neuronal cell death. The present study implies that inhibiting JNK/Vps34 complex autophagy induction pathway may be a novel therapeutic approach for the treatment of MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tianji Lin
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Shijuan Ruan
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Dingbang Huang
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Wenjun Li
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Bin Wang
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China.
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, 510515, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Zhang W, Zhu B, Ren J, Lu F, Qi Y, Weng W, Gao R. Two methods for modeling of sick sinus syndrome in rats: Ischemia reperfusion and sodium hydroxide induced injury. Biomed Pharmacother 2019; 111:778-784. [PMID: 30612002 DOI: 10.1016/j.biopha.2018.11.091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/13/2018] [Accepted: 11/25/2018] [Indexed: 01/23/2023] Open
Abstract
The Sick Sinus Syndrome (SSS) is a serious life-threatening heart disease. It is important to establish a credible and stable sinus node damage model. In this study, we use two methods to construct an SSS damage model in rats. One is to inject sodium hydroxide to the SSS area through internal jugular vein. Another is to cause ischemia-reperfusion injury on the SSS area. 43 healthy SD rats were randomly divided into 4 groups, namely ischemia-reperfusion injury group (IRIG), inject sodium hydroxide group (ISHG), and propranolol group (PG) and the control group (CG). The achievement ratio of modeling was 67% in the IRIG and 83% in the ISHG. The HR significantly decreased after operation in the IRIG and ISHG compared with pre-operation (P<0.01). The HR was reduced by above 30% in these 2 groups after modeling, while the reduction was better maintained in IRIG. Additionally, the sinoatrial node recovery time (SNRT) and sinoatrial conduction time (SACT) were significantly prolonged compared with pre-modeling in 2 groups (P < 0.01). Morphology results showed blurry in structure and boundaries with pale cytoplasm. It is speculated that IRIG and ISHG modeling might influence the calcium concentration and damage the sinus node function by decrease the expression of HCN4 and SCN5A, which impaired the driving ability of sinus node and leading to apoptosis. Ischemia reperfusion injury and sodium hydroxide injury could construct stable SSS models which could represent clinic pathological damage. Thus, both methods could be used for further studies of the SSS mechanisms and drugs.
Collapse
Affiliation(s)
- Wantong Zhang
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Baochen Zhu
- Beijing University of Chinese Medicine, 100029, China
| | - Jianxun Ren
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Fang Lu
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Yi Qi
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Weiliang Weng
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Rui Gao
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China.
| |
Collapse
|
24
|
Pereira LC, de Paula ES, Pazin M, Carneiro MFH, Grotto D, Barbosa F, Dorta DJ. Niacin prevents mitochondrial oxidative stress caused by sub-chronic exposure to methylmercury. Drug Chem Toxicol 2018; 43:64-70. [PMID: 30192646 DOI: 10.1080/01480545.2018.1497045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Humans and animals can be exposed to different chemical forms of mercury (Hg) in the environment. For example, methylmercury (MeHg)-contaminated fish is part of the basic diet of the riparian population in the Brazilian Amazon Basin, which leads to high total blood and plasma Hg levels in people living therein. Hg induces toxic effects mainly through oxidative stress. Different compounds have been used to prevent the damage caused by MeHg-induced reactive oxygen species (ROS). This study aims to investigate the in vivo effects of sub-chronic exposure to low MeHg levels on the mitochondrial oxidative status and to evaluate the niacin protective effect against MeHg-induced oxidative stress. For this purpose, Male Wistar rats were divided into four groups: control group, treated with drinking water on a daily basis; group exposed to MeHg at a dose of 100 µg/kg/day; group that received niacin at a dose of 50 mg/kg/day in drinking water, with drinking water being administered by gavage; group that received niacin at a dose of 50 mg/kg/day in drinking water as well as MeHg at a dose of 100 µg/kg/day. After 12 weeks, the rats, which weighed 500-550 g, were sacrificed, and their liver mitochondria were isolated by standard differential centrifugation. Sub-chronic exposure to MeHg (100 µg/kg/day for 12 weeks) led to mitochondrial swelling (p < 0.05) and induced ROS overproduction as determined by increased DFCH oxidation (p < 0.05), increased gluthatione oxidation (p < 0.05), and reduced protein thiol content (p < 0.05). In contrast, niacin supplementation inhibited oxidative stress, which counteracted and minimized the toxic MeHg effects on mitochondria.
Collapse
Affiliation(s)
- Lílian Cristina Pereira
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil.,Faculdade de Ciências Agronômicas, Departamento de Bioprocessos e Biotecnologia, Universidade Estadual Paulista, Botucatu, São Paulo, Brasil.,Departamento de Patologia, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, TOXICAM - Núcleo de Avaliação do Impacto Ambiental sobre a Saúde Humana, Botucatu, São Paulo, Brazil
| | - Eloisa Silva de Paula
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Murilo Pazin
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Maria Fernanda Hornos Carneiro
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Denise Grotto
- Laboratório de Pesquisa em Toxicologia, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade de Sorocaba, Sorocaba, São Paulo, Brasil
| | - Fernando Barbosa
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Daniel Junqueira Dorta
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Departamento de Química, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil.,Instituto Nacional de Tecnologias Alternativas de Detecção, Avaliação Toxicológica e Remoção de Micropututantes e Radioativos (INCT-DATREM), Unesp, Instituto de Química, Araraquara, São Paulo, Brasil
| |
Collapse
|
25
|
Sampaio GSA, Oliveira KRHM, Kauffmann N, do Nascimento JLM, Souza GS, Gomes BD, de Lima SMA, Silveira LCL, Rocha FAF, Herculano AM. Methylmercury alters the number and topography of NO-synthase positive neurons in embryonic retina: Protective effect of alpha-tocopherol. Toxicol In Vitro 2018; 53:89-98. [PMID: 30075186 DOI: 10.1016/j.tiv.2018.07.018] [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: 04/13/2018] [Revised: 06/25/2018] [Accepted: 07/30/2018] [Indexed: 10/28/2022]
Abstract
Vertebrate retina has been shown to be an important target for mercury toxicity and very studies have shown the effect of mercury on the retinal ontogenesis. The nitrergic system plays an important role in the retinal development. The current work studied the effects of methylmercury (MeHg) exposure on the NO-synthase positive neurons (NADPH-diaphorase neurons or NADPH-d+) of the chick retinal ganglion cell layer at embryonic E15 and postnatal P1 days. Retinal flat mounts were stained for NADPH-diaphorase histochemistry and mosaic properties of NADPH-d + were studied by plotting isodensity maps and employing density recovery profile technique. It was also evaluated the protective effect of alpha-tocopherol treatment on retinal tissues exposed to MeHg. MeHg exposure decreased the density of NADPH-d + neurons and altered cell mosaic properties at E15 but had very little or no effect at P1 retinas. Alpha-tocopherol has a protective effect against MeHg exposure at E15. MeHg alterations and alpha-tocopherol protective effect in embryonic retinas were demonstrated to be at work in experimental conditions. MeHg effect in the early phases of visual system development in natural conditions might use the nitrergic pathway and supplementary diet could have a protective effect. At later stages, this mechanism seems to be naturally protected.
Collapse
Affiliation(s)
- Gabriela S A Sampaio
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | | | - Nayara Kauffmann
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - José Luiz M do Nascimento
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil; Universidade Ceuma, São Luís, Maranhão, Brazil
| | - Givago S Souza
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil; Núcleo de Medicina Tropical, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Bruno D Gomes
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | | | - Luiz Carlos L Silveira
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil; Núcleo de Medicina Tropical, Universidade Federal do Pará, Belém, Pará, Brazil; Universidade Ceuma, São Luís, Maranhão, Brazil
| | | | - Anderson M Herculano
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil.
| |
Collapse
|
26
|
Tucker EK, Nowak RA. Methylmercury alters proliferation, migration, and antioxidant capacity in human HTR8/SV-neo trophoblast cells. Reprod Toxicol 2018; 78:60-68. [PMID: 29581082 DOI: 10.1016/j.reprotox.2018.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/01/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Abstract
Methylmercury, a potent neurotoxin, is able to pass through the placenta, but its effects on the placenta itself have not been elucidated. Using an immortalized human trophoblast cell line, HTR8/SV-neo, we assessed the in vitro toxicity of methylmercury. We found that 1 μg/mL methylmercury decreased viability, proliferation, and migration; and it had effects on antioxidant genes similar to those seen in neural cells. However, methylmercury led to decreased expression of superoxide dismutase 1 and increased expression of surfactant protein D. HTR cells treated 0.01 or 0.1 μg/mL methylmercury had increased migration rates along with decreased expression of an adhesion gene, cadherin 3, suggesting that low doses of methylmercury promote migration in HTR cells. Our results indicate that trophoblast cells react differently to methylmercury relative to neural cell lines, and thus investigation of methylmercury toxicity in placental cells is needed to understand the effects of this heavy metal on the placenta.
Collapse
Affiliation(s)
- Emily K Tucker
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207, W. Gregory Dr., Urbana, Illinois, USA.
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207, W. Gregory Dr., Urbana, Illinois, USA
| |
Collapse
|
27
|
Prince LM, Rand MD. Methylmercury exposure causes a persistent inhibition of myogenin expression and C2C12 myoblast differentiation. Toxicology 2018; 393:113-122. [PMID: 29104120 PMCID: PMC5757876 DOI: 10.1016/j.tox.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxicant, best known for its selective targeting of the developing nervous system. MeHg exposure has been shown to cause motor deficits such as impaired gait and coordination, muscle weakness, and muscle atrophy, which have been associated with disruption of motor neurons. However, recent studies have suggested that muscle may also be a target of MeHg toxicity, both in the context of developmental myogenic events and of low-level chronic exposures affecting muscle wasting in aging. We therefore investigated the effects of MeHg on myotube formation, using the C2C12 mouse myoblast model. We found that MeHg inhibits both differentiation and fusion, in a concentration-dependent manner. Furthermore, MeHg specifically and persistently inhibits myogenin (MyoG), a transcription factor involved in myocyte differentiation, within the first six hours of exposure. MeHg-induced reduction in MyoG expression is contemporaneous with a reduction of a number of factors involved in mitochondrial biogenesis and mtDNA transcription and translation, which may implicate a role for mitochondria in mediating MeHg-induced change in the differentiation program. Unexpectedly, inhibition of myoblast differentiation with MeHg parallels inhibition of Notch receptor signaling. Our research establishes muscle cell differentiation as a target for MeHg toxicity, which may contribute to the underlying etiology of motor deficits with MeHg toxicity.
Collapse
Affiliation(s)
- Lisa M Prince
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| |
Collapse
|
28
|
Barbosa NV, Nogueira CW, Nogara PA, de Bem AF, Aschner M, Rocha JBT. Organoselenium compounds as mimics of selenoproteins and thiol modifier agents. Metallomics 2017; 9:1703-1734. [PMID: 29168872 DOI: 10.1039/c7mt00083a] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Selenium is an essential trace element for animals and its role in the chemistry of life relies on a unique functional group: the selenol (-SeH) group. The selenol group participates in critical redox reactions. The antioxidant enzymes glutathione peroxidase (GPx) and thioredoxin reductase (TrxR) exemplify important selenoproteins. The selenol group shares several chemical properties with the thiol group (-SH), but it is much more reactive than the sulfur analogue. The substitution of S by Se has been exploited in organic synthesis for a long time, but in the last 4 decades the re-discovery of ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one) and the demonstration that it has antioxidant and therapeutic properties has renovated interest in the field. The ability of ebselen to mimic the reaction catalyzed by GPx has been viewed as the most important molecular mechanism of action of this class of compound. The term GPx-like or thiol peroxidase-like reaction was previously coined in the field and it is now accepted as the most important chemical attribute of organoselenium compounds. Here, we will critically review the literature on the capacity of organoselenium compounds to mimic selenoproteins (particularly GPx) and discuss some of the bottlenecks in the field. Although the GPx-like activity of organoselenium compounds contributes to their pharmacological effects, the superestimation of the GPx-like activity has to be questioned. The ability of these compounds to oxidize the thiol groups of proteins (the thiol modifier effects of organoselenium compounds) and to spare selenoproteins from inactivation by soft-electrophiles (MeHg+, Hg2+, Cd2+, etc.) might be more relevant for the explanation of their pharmacological effects than their GPx-like activity. In our view, the exploitation of the thiol modifier properties of organoselenium compounds can be harnessed more rationally than the use of low mass molecular structures to mimic the activity of high mass macromolecules that have been shaped by millions to billions of years of evolution.
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, RS, Brazil.
| | - Cristina W Nogueira
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| | - Pablo A Nogara
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| | - Andreza F de Bem
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
29
|
Farina M, Aschner M, da Rocha JBT. The catecholaminergic neurotransmitter system in methylmercury-induced neurotoxicity. ADVANCES IN NEUROTOXICOLOGY 2017; 1:47-81. [PMID: 32346666 DOI: 10.1016/bs.ant.2017.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology , Albert Einstein College of Medicine , Bronx , NY , United States
| | - João Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
30
|
Developmental neurotoxicity of the hippocampus following in utero exposure to methylmercury: impairment in cell signaling. Arch Toxicol 2017; 92:513-527. [PMID: 28821999 DOI: 10.1007/s00204-017-2042-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/10/2017] [Indexed: 01/01/2023]
Abstract
In this study, we assessed some hippocampal signaling cascades and behavioral impairments in 30-day-old rat pups prenatally exposed to methylmercury (MeHg). Pregnant rats were exposed to 1.0 or 2.0 mg/kg MeHg by gavage in alternated days from gestational day 5 until parturition. We found increased anxiety-like and decreased exploration behavior evaluated by open field test and deficit of both short- and long-term memories by novel object recognition task, respectively, in MeHg-treated pups. Downregulated PI3K/Akt/mTOR pathway and activated/hypophosphorylated (Ser9) GSK3β in MeHg-treated pups could be upstream of hyperphosphorylated Tau (Ser396) destabilizing microtubules and contributing to neural dysfunction in the hippocampus of these rats. Hyperphosphorylated/activated p38MAPK and downregulated phosphoErk1/2 support a role for mitogen-activated protein kinase (MAPK) cascade on MeHg neurotoxicity. Decreased receptor of advanced glycation end products (RAGE) immunocontent supports the assumption that downregulated RAGE/Erk1/2 pathway could be involved in hypophosphorylated lysine/serine/proline (KSP) repeats on neurofilament subunits and disturbed axonal transport. Downregulated myelin basic protein (MBP), the major myelin protein, is compatible with dysmyelination and neurofilament hypophosphorylation. Increased glial fibrillary acidic protein (GFAP) levels suggest reactive astrocytes, and active apoptotic pathways BAD/BCL-2, BAX/BCL-XL, and caspase 3 suggest cell death. Taken together, our findings get light on important signaling mechanisms that could underlie the behavioral deficits in 30-day-old pups prenatally exposed to MeHg.
Collapse
|
31
|
The Putative Role of Environmental Mercury in the Pathogenesis and Pathophysiology of Autism Spectrum Disorders and Subtypes. Mol Neurobiol 2017; 55:4834-4856. [DOI: 10.1007/s12035-017-0692-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023]
|
32
|
Ghizoni H, de Souza V, Straliotto MR, de Bem AF, Farina M, Hort MA. Superoxide anion generation and oxidative stress in methylmercury-induced endothelial toxicity in vitro. Toxicol In Vitro 2017; 38:19-26. [DOI: 10.1016/j.tiv.2016.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/12/2016] [Accepted: 10/26/2016] [Indexed: 12/23/2022]
|
33
|
Das S, Paul A, Mumbrekar KD, Rao SBS. Harmonization of Mangiferin on methylmercury engendered mitochondrial dysfunction. ENVIRONMENTAL TOXICOLOGY 2017; 32:630-644. [PMID: 28071871 DOI: 10.1002/tox.22265] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 06/06/2023]
Abstract
Mangiferin (MGN), a C-glucosylxanthone abundantly found in mango plants, was studied for its potential to ameliorate methylmercury (MeHg) induced mitochondrial damage in HepG2 (human hepatocarcinoma) cell line. Cell viability experiments performed using 3-[4,5-dimethylthiazol-2-yl]-2,5- diphenyltetrazolium bromide (MTT) showed protective property of MGN in annulling MeHg-induced cytotoxicity. Conditioning the cells with optimal dose of MGN (50 µM) lowered MeHg-induced oxidative stress, calcium influx/efflux, depletion of mitochondrial trans-membrane potential and prevented mitochondrial fission as observed by decrease in Mitotracker red fluorescence, expression of pDRP1 (serine 616), and DRP1 levels. MGN pre-treated cells demonstrated elevation in the activities of glutathione (GSH), Glutathione-S-transferase (GST), Glutathione peroxidase (GPx), Glutathione reductase (GR), reduced levels of Aspartate aminotransferase (AST) and Alanine aminotransferase (ALT) and mitochondrial electron transport chain (ETC) enzyme complexes. In addition, the anti-apoptotic effect of MGN was clearly indicated by the reduction in MeHg-induced apoptotic cells analyzed by flowcytometric analysis after Annexin V-FITC/propidium iodide staining. In conclusion, the present work demonstrates the ability of a dietary polyphenol, MGN to ameliorate MeHg-mediated mitochondrial dysfunction in human hepatic cells in vitro. This hepatoprotective potential may be attributed predominantly to the free radical scavenging/antioxidant property of MGN, by facilitating the balancing of cellular Ca2+ ions, maintenance of redox homeostasis and intracellular antioxidant activities, ultimately preserving the mitochondrial function and cell viability after MeHg intoxication. As MeHg intoxication occurs over a period of time, continuous consumption of such dietary compounds may prove to be very useful in promoting human health. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 630-644, 2017.
Collapse
Affiliation(s)
- Shubhankar Das
- Department of Radiation Biology and Toxicology, School of Life Sciences, Manipal University, Manipal, Karnataka, India, 576104
| | - Ajanta Paul
- Department of Biotechnology, School of Life Sciences, Manipal University, Manipal, Karnataka, India, 576104
| | - Kamalesh D Mumbrekar
- Department of Radiation Biology and Toxicology, School of Life Sciences, Manipal University, Manipal, Karnataka, India, 576104
| | - Satish B S Rao
- Department of Radiation Biology and Toxicology, School of Life Sciences, Manipal University, Manipal, Karnataka, India, 576104
| |
Collapse
|
34
|
Rudgalvyte M, Peltonen J, Lakso M, Wong G. Chronic MeHg exposure modifies the histone H3K4me3 epigenetic landscape in Caenorhabditis elegans. Comp Biochem Physiol C Toxicol Pharmacol 2017; 191:109-116. [PMID: 27717699 DOI: 10.1016/j.cbpc.2016.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 09/29/2016] [Accepted: 10/02/2016] [Indexed: 01/05/2023]
Abstract
Methylmercury (MeHg) is a persistent environmental pollutant that occurs in the food chain, at occupational sites, and via medical procedures. Exposure in humans and animal models results in renal, neuro, and reproductive toxicities. In this study, we demonstrate that chronic exposure to MeHg (10μM) causes epigenetic landscape modifications of histone H3K4 trimethylation (H3K4me3) marks in Caenorhabditis elegans using chromatin immuno-precipitation sequencing (ChIP-seq). The modifications correspond to the locations of 1467 genes with enhanced and 508 genes with reduced signals. Among enhanced genes are those encoding glutathione-S-transferases, lipocalin-related protein and a cuticular collagen. ChIP-seq enhancement of these genes was confirmed with increased mRNA expression levels revealed by qRT-PCR. Furthermore, we observed enhancement of H3K4me3 marks in these genes in animals exposed to MeHg in utero and assayed at L4 stage. In utero exposure enhanced marks without alterations in mRNA expression except for the lpr-5 gene. Finally, knockdown of lipocalin-related protein gene lpr-5, which is involved in intercellular signaling, and cuticular collagen gene dpy-7, structural component of the cuticle, by RNA interference (RNAi) resulted in increased lethality of animals after MeHg exposure. Our results provide new data on the epigenetic landscape changes elicited by MeHg exposure, as well as describe a unique model for studying in utero effects of heavy metals. Together, these findings may help to understand the toxicological effects of MeHg at the molecular level.
Collapse
Affiliation(s)
- Martina Rudgalvyte
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Kuopio, Finland; Faculty of Health Sciences, University of Macau, Macau, S.A.R., China
| | - Juhani Peltonen
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Kuopio, Finland
| | - Merja Lakso
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Kuopio, Finland
| | - Garry Wong
- Faculty of Health Sciences, University of Macau, Macau, S.A.R., China.
| |
Collapse
|
35
|
Memantine, a Low-Affinity NMDA Receptor Antagonist, Protects against Methylmercury-Induced Cytotoxicity of Rat Primary Cultured Cortical Neurons, Involvement of Ca2+ Dyshomeostasis Antagonism, and Indirect Antioxidation Effects. Mol Neurobiol 2016; 54:5034-5050. [DOI: 10.1007/s12035-016-0020-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/01/2016] [Indexed: 01/20/2023]
|
36
|
Low-Dose Methylmercury-Induced Apoptosis and Mitochondrial DNA Mutation in Human Embryonic Neural Progenitor Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5137042. [PMID: 27525052 PMCID: PMC4972916 DOI: 10.1155/2016/5137042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/06/2016] [Accepted: 06/22/2016] [Indexed: 01/12/2023]
Abstract
Methylmercury (MeHg) is a long-lasting organic pollutant primarily found in the aquatic environment. The developing brain is particularly sensitive to MeHg due to reduced proliferation of neural stem cell. Although several mechanisms of MeHg-induced apoptosis have been defined in culture models, it remains unclear whether mitochondrial DNA (mtDNA) mutation is involved in the toxic effect of MeHg, especially in the neural progenitor cells. In the present study, the ReNcell CX cell, a human neural progenitor cells (hNPCs) line, was exposed to nanomolar concentrations of MeHg (≤50 nM). We found that MeHg altered mitochondrial metabolic function and induced apoptosis. In addition, we observed that MeHg induced ROS production in a dose-dependent manner in hNPCs cells, which was associated with significantly increased expressions of ND1, Cytb, and ATP6. To elucidate the mechanism underlying MeHg toxicity on mitochondrial function, we examined the ATP content and mitochondrial membrane potential in MeHg-treated hNPCs. Our study showed that MeHg exposure led to decreased ATP content and reduced mitochondrial membrane potential, which failed to match the expansion in mtDNA copy number, suggesting impaired mtDNA. Collectively, these results demonstrated that MeHg induced toxicity in hNPCs through altering mitochondrial function and inducing oxidative damage to mtDNA.
Collapse
|
37
|
Yuan Y, Atchison WD. Multiple Sources of Ca2+ Contribute to Methylmercury-Induced Increased Frequency of Spontaneous Inhibitory Synaptic Responses in Cerebellar Slices of Rat. Toxicol Sci 2016; 150:117-30. [PMID: 26732885 DOI: 10.1093/toxsci/kfv314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We previously showed that elevated intracellular Ca(2+) ([Ca(2+)]i) in the molecular layer and granule cells in cerebellar slices is responsible for the initial increases in frequency of spontaneous or miniature inhibitory postsynaptic currents (sIPSCs or mIPSCs) of Purkinje cells following methylmercury (MeHg) treatment. To identify the contribution of different Ca(2+) sources to MeHg-induced stimulation of spontaneous GABA release, we examined sIPSC or mIPSC frequency of Purkinje cells in acutely prepared cerebellar slices using whole-cell patch-clamp recording techniques under conditions of lowered [Ca(2+)]o, pretreatment with caffeine, cyclopiazonic acid (CPA), thapsigargin or ruthenium red (RR) to deplete ryanodine-sensitive and insensitive intracellular Ca(2+) stores or mitochondria, or a combination of lowering [Ca(2+)]o and increased BAPTA buffering. Lowering [Ca(2+)]o significantly reduced sIPSC or mIPSC frequency and amplitudes, but failed to completely prevent MeHg-induced increase in these events frequency. Caffeine, CPA, or thapisgargin also minimized MeHg-induced increase in sIPSC frequency, yet none of them completely blocked MeHg-induced increase in sIPSC frequency. Similarly, the mitochondrial Ca(2+) transport inhibitor RR, or a combination of lowering [Ca(2+)]o and BAPTA buffering reduced but did not prevent MeHg-induced changes in mIPSC frequency. Consistently, confocal Ca(2+) imaging under low [Ca(2+)]o conditions or in the presence of caffeine or CPA exhibited a marked reduction of MeHg-induced increases in [Ca(2+)]i in both molecular and granule layers. Thus, these results verify that a combination of extracellular Ca(2+) influx and Ca(2+) release from different intracellular Ca(2+) pools all contribute to MeHg-induced increase in [Ca(2+)]i and spontaneous GABA release, although extracellular Ca(2+) appears to be the primary contributor.
Collapse
Affiliation(s)
- Yukun Yuan
- Department of Pharmacology/Toxicology, Michigan State University, B-331 Life Sciences Building, 1355 Bogue Street, East Lansing, Michigan 48824-1317
| | - William D Atchison
- Department of Pharmacology/Toxicology, Michigan State University, B-331 Life Sciences Building, 1355 Bogue Street, East Lansing, Michigan 48824-1317
| |
Collapse
|
38
|
Abstract
Mitochondrial dynamics, fission and fusion, were first identified in yeast with investigation in heart cells beginning only in the last 5 to 7 years. In the ensuing time, it has become evident that these processes are not only required for healthy mitochondria, but also, that derangement of these processes contributes to disease. The fission and fusion proteins have a number of functions beyond the mitochondrial dynamics. Many of these functions are related to their membrane activities, such as apoptosis. However, other functions involve other areas of the mitochondria, such as OPA1's role in maintaining cristae structure and preventing cytochrome c leak, and its essential (at least a 10 kDa fragment of OPA1) role in mtDNA replication. In heart disease, changes in expression of these important proteins can have detrimental effects on mitochondrial and cellular function.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| | - T T Liu
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| |
Collapse
|
39
|
Shettigar NB, Das S, Rao NB, Rao SBS. Thymol, a monoterpene phenolic derivative of cymene, abrogates mercury-induced oxidative stress resultant cytotoxicity and genotoxicity in hepatocarcinoma cells. ENVIRONMENTAL TOXICOLOGY 2015; 30:968-980. [PMID: 24574037 DOI: 10.1002/tox.21971] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/03/2014] [Accepted: 02/09/2014] [Indexed: 06/03/2023]
Abstract
Thymol (TOH) was investigated for its ability to protect against mercuric chloride (HgCl2 )-induced cytotoxicity and genotoxicity using human hepatocarcinoma (HepG2) cell line. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay confirmed the efficacy of TOH pretreatment in attenuating HgCl2 -induced cytotoxicity. Pretreatment with TOH inhibited HgCl2 -induced genotoxicity, depolarization of mitochondrial membrane, oxidative stress, and mitochondrial superoxide levels. Interestingly, TOH (100 µM) alone elevated the intracellular basal glutathione S-transferase (GST) levels and TOH pretreatment abrogated the decrease in glutathione, GST, superoxide dismutase, and catalase levels even after HgCl2 intoxication. Furthermore, TOH was also capable of inhibiting HgCl2 -induced apoptotic as well as necrotic cell death analyzed by flowcytometric analysis of cells dual stained with Annexin-FITC/propidium iodide. The present findings clearly indicate the cytoprotective potential of TOH against HgCl2 -induced toxicity, which may be attributed to its free radical scavenging ability which facilitated in reducing oxidative stress and mitochondrial damage thereby inhibiting cell death.
Collapse
Affiliation(s)
- Nishan B Shettigar
- Department of Biotechnology, School of Life Sciences, Manipal University, Manipal, Karnataka, 576 104, India
| | - Shubhankar Das
- Division of Radiobiology and Toxicology, School of Life Sciences, Manipal University, Manipal, Karnataka, 576 104, India
| | - Nageshwar B Rao
- Division of Radiobiology and Toxicology, School of Life Sciences, Manipal University, Manipal, Karnataka, 576 104, India
| | - Satish B S Rao
- Division of Radiobiology and Toxicology, School of Life Sciences, Manipal University, Manipal, Karnataka, 576 104, India
| |
Collapse
|
40
|
Shiraishi M, Hangai M, Yamamoto M, Sasaki M, Tanabe A, Sasaki Y, Miyamoto A. Alteration in MARCKS phosphorylation and expression by methylmercury in SH-SY5Y cells and rat brain. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:1256-1263. [PMID: 24835554 DOI: 10.1016/j.etap.2014.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 03/27/2014] [Accepted: 04/21/2014] [Indexed: 06/03/2023]
Abstract
The molecular mechanisms mediating methylmercury (MeHg)-induced neurotoxicity are not completely understood. Because myristoylated alanine-rich C kinase substrate (MARCKS) plays an essential role in the differentiation and development of neuronal cells, we studied the alteration of MARCKS expression and phosphorylation in MeHg-induced neurotoxicity of neuroblastoma SH-SY5Y cells and in the rat brain. Exposure to MeHg induced a decrease in cell viability of SH-SY5Y cells, which was accompanied by a significant increase in phosphorylation and a reduction in MARCKS expression. Pretreatment of cells with a protein kinase C inhibitor or an extracellular Ca(2+) chelator suppressed MeHg-induced MARCKS phosphorylation. In MARCKS knock-down cells, MeHg-induced cell death was significantly augmented in comparison to control siRNA. In brain tissue from MeHg-treated rats, MARCKS phosphorylation was enhanced in the olfactory bulb in comparison to control rats. The present study may indicate that alteration in MARCKS expression or phosphorylation has consequences for MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Mitsuya Shiraishi
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.
| | - Makoto Hangai
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Megumi Yamamoto
- Department of Basic Medical Sciences, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan
| | - Masanori Sasaki
- Department of Basic Medical Sciences, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan
| | - Atsuhiro Tanabe
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, 307 Fukasaku, Minuma-ku, Saitama 337-8570, Japan
| | - Yasuharu Sasaki
- Laboratory of Pharmacology, School of Pharmaceutical Science, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Atsushi Miyamoto
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
41
|
Exploring cross-talk between oxidative damage and excitotoxicity and the effects of riluzole in the rat cortex after exposure to methylmercury. Neurotox Res 2014; 26:40-51. [PMID: 24519665 DOI: 10.1007/s12640-013-9448-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 11/07/2013] [Accepted: 12/05/2013] [Indexed: 12/22/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxin that causes neurologic and developmental diseases. Oxidative damage and excitotoxicity are putative mechanisms, which underlie MeHg-induced neurotoxicity. In this study, the cross-talk between the oxidative damage and excitotoxicity pathways and the protective effects of riluzole in the rat cortex were explored. Rats were injected with MeHg and/or riluzole, and cold vapor atomic fluorescence spectrometry, hematoxylin and eosin staining, flow cytometry, fluorescence assays, spectrophotometry, real-time PCR, and Western blotting were used to evaluate neurotoxicity. The present study showed that (1) MeHg accumulated in the cerebral cortex and caused pathology. (2) MeHg caused oxidative damage by inducing glutathione (GSH) depletion, reactive oxygen species (ROS) production, inhibition of antioxidant enzyme activity, and alteration of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. (3) MeHg disrupted the glutamate transporters (GluTs), glutamate-glutamine cycle, and N-methyl-D-aspartate receptor expression and induced excitotoxicity. (4) Excitotoxicity resulted in disruption of GSH synthesis, calcium overloading, oxidative damage, and excessive ROS production. (5) Pretreatment with riluzole antagonized MeHg neurotoxicity by down regulating cross-talk between the oxidative damage and excitotoxicity pathways. In conclusion, the cross-talk between the oxidative damage and excitotoxicity pathways caused by MeHg exposure was linked by GluTs and calcium and inhibited by riluzole treatment.
Collapse
|
42
|
Rudgalvyte M, VanDuyn N, Aarnio V, Heikkinen L, Peltonen J, Lakso M, Nass R, Wong G. Methylmercury exposure increases lipocalin related (lpr) and decreases activated in blocked unfolded protein response (abu) genes and specific miRNAs in Caenorhabditis elegans. Toxicol Lett 2013; 222:189-96. [PMID: 23872261 DOI: 10.1016/j.toxlet.2013.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/10/2013] [Accepted: 07/10/2013] [Indexed: 01/15/2023]
Abstract
Methylmercury (MeHg) is a persistent environmental and dietary contaminant that causes serious adverse developmental and physiologic effects at multiple cellular levels. In order to understand more fully the consequences of MeHg exposure at the molecular level, we profiled gene and miRNA transcripts from the model organism Caenorhabditis elegans. Animals were exposed to MeHg (10 μM) from embryo to larval 4 (L4) stage and RNAs were isolated. RNA-seq analysis on the Illumina platform revealed 541 genes up- and 261 genes down-regulated at a cutoff of 2-fold change and false discovery rate-corrected significance q < 0.05. Among the up-regulated genes were those previously shown to increase under oxidative stress conditions including hsp-16.11 (2.5-fold), gst-35 (10.1-fold), and fmo-2 (58.5-fold). In addition, we observed up-regulation of 6 out of 7 lipocalin related (lpr) family genes and down regulation of 7 out of 15 activated in blocked unfolded protein response (abu) genes. Gene Ontology enrichment analysis highlighted the effect of genes related to development and organism growth. miRNA-seq analysis revealed 6-8 fold down regulation of mir-37-3p, mir-41-5p, mir-70-3p, and mir-75-3p. Our results demonstrate the effects of MeHg on specific transcripts encoding proteins in oxidative stress responses and in ER stress pathways. Pending confirmation of these transcript changes at protein levels, their association and dissociation characteristics with interaction partners, and integration of these signals, these findings indicate broad and dynamic mechanisms by which MeHg exerts its harmful effects.
Collapse
Affiliation(s)
- Martina Rudgalvyte
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Kuopio, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Dórea JG, Farina M, Rocha JBT. Toxicity of ethylmercury (and Thimerosal): a comparison with methylmercury. J Appl Toxicol 2013; 33:700-11. [PMID: 23401210 DOI: 10.1002/jat.2855] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/10/2012] [Accepted: 12/11/2012] [Indexed: 12/18/2022]
Abstract
Ethylmercury (etHg) is derived from the metabolism of thimerosal (o-carboxyphenyl-thio-ethyl-sodium salt), which is the most widely used form of organic mercury. Because of its application as a vaccine preservative, almost every human and animal (domestic and farmed) that has been immunized with thimerosal-containing vaccines has been exposed to etHg. Although methylmercury (meHg) is considered a hazardous substance that is to be avoided even at small levels when consumed in foods such as seafood and rice (in Asia), the World Health Organization considers small doses of thimerosal safe regardless of multiple/repetitive exposures to vaccines that are predominantly taken during pregnancy or infancy. We have reviewed in vitro and in vivo studies that compare the toxicological parameters among etHg and other forms of mercury (predominantly meHg) to assess their relative toxicities and potential to cause cumulative insults. In vitro studies comparing etHg with meHg demonstrate equivalent measured outcomes for cardiovascular, neural and immune cells. However, under in vivo conditions, evidence indicates a distinct toxicokinetic profile between meHg and etHg, favoring a shorter blood half-life, attendant compartment distribution and the elimination of etHg compared with meHg. EtHg's toxicity profile is different from that of meHg, leading to different exposure and toxicity risks. Therefore, in real-life scenarios, a simultaneous exposure to both etHg and meHg might result in enhanced neurotoxic effects in developing mammals. However, our knowledge on this subject is still incomplete, and studies are required to address the predictability of the additive or synergic toxicological effects of etHg and meHg (or other neurotoxicants).
Collapse
Affiliation(s)
- José G Dórea
- Department of Nutrition, Faculty of Health Sciences, Universidade de Brasilia, 70919-970, Brasilia, DF, Brazil.
| | | | | |
Collapse
|
44
|
Diphenyl diselenide prevents methylmercury-induced mitochondrial dysfunction in rat liver slices. Tetrahedron 2012. [DOI: 10.1016/j.tet.2012.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|