1
|
Panzenhagen AC, Petry FDS, Alves-Teixeira A, Santos L, Carazza-Kessler FG, Gelain DP, Moreira JCF. Biomarkers of methylmercury neurotoxicity and neurodevelopmental features: A systematic review. Food Chem Toxicol 2024; 191:114851. [PMID: 38986832 DOI: 10.1016/j.fct.2024.114851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
The issue of MeHg contamination is a significant concern due to its detrimental impact on the environment. This study aimed to thoroughly investigate the effects of MeHg on neurodevelopmental biomarkers, as there is a lack of systematic reviews in this area. We conducted a comprehensive search of three databases (PubMed, Scopus, and Web of Science) and found 522 records, which were then meticulously reviewed by two independent reviewers. A total of 66 studies were included, with biomarkers related to oxidative stress, neurotransmission, inflammation, epigenetics, and apoptosis being the most prominent. The results of both in vitro and in vivo models indicate that antioxidant enzymes and other oxidative stress-related markers are indeed, altered following MeHg exposure. Moreover, MeHg exposure causes significant disruptions to neurotransmitter levels, activities of neurotransmitter synthesis enzymes, receptor densities, and proteins involved in synaptic function. Proinflammatory biomarkers are consistently overexpressed in both MeHg-treated cells and the brains of exposed rats. Furthermore, studies on DNA methylation and biomarker activity suggest that MeHg exposure may lead to neurotoxicity and neurodevelopmental issues via perturbations to epigenetic markers and the apoptosis pathway.
Collapse
Affiliation(s)
- Alana Castro Panzenhagen
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| | - Fernanda Dos Santos Petry
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Alexsander Alves-Teixeira
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Lucas Santos
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Flávio Gabriel Carazza-Kessler
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Centro de Estudos Em Estresse Oxidativo, Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| |
Collapse
|
2
|
Nakamura R, Iwai T, Takanezawa Y, Shirahata T, Konishi N, Ohshiro Y, Uraguchi S, Tanabe M, Kobayashi Y, Sakamoto K, Nakahara T, Yamamoto M, Kiyono M. Oleanolic acid-3-glucoside, a synthetic oleanane-type saponin, ameliorates methylmercury-induced dysfunction of synaptic transmission in mice. Toxicology 2024; 506:153867. [PMID: 38906242 DOI: 10.1016/j.tox.2024.153867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Methylmercury (MeHg) is widely distributed in nature and is known to cause neurotoxic effects. This study aimed to examine the anti-MeHg activity of oleanolic acid-3-glucoside (OA3Glu), a synthetic oleanane-type saponin derivative, by evaluating its effects on motor function, pathology, and electrophysiological properties in a mouse model of MeHg poisoning. Mice were orally administered 2 or 4 mg·kg-1·d-1 MeHg with or without 100 µg·kg-1·d-1 OA3Glu 5x/week for four weeks. Motor function was evaluated using beam-walking and dynamic weight-bearing (DWB) tests. High-dose MeHg exposure significantly increased the frequency of stepping off the hind leg while crossing the beam in the beam-walking test, and increased weight on forelegs when moving freely in the DWB test. OA3Glu treatment alleviated motor abnormality caused by high-dose MeHg exposure in both motor function tests. Additionally, OA3Glu treatment reduced the number of contracted Purkinje cells frequently observed in the cerebellum of MeHg-treated groups, although cerebrum histology was similar in all experimental groups. The synaptic potential amplitude in the cerebellum decreased as MeHg exposure increased, which was restored by OA3Glu treatment. Even in the cerebrum, where the effects of MeHg were not observed, the amplitude of the field potential was suppressed with increasing MeHg exposure but was restored with OA3Glu treatment. Taken together, the study findings suggest that OA3Glu improves neurotransmission and movement disorders associated with MeHg exposure via protection of Purkinje cells in the cerebellum while ameliorating pre/post-synaptic deficits in the cerebral cortex in which no changes were observed at the tissue level, potentially providing a treatment to mitigate MeHg toxicity.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Takashi Iwai
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yasukazu Takanezawa
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tatsuya Shirahata
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Naruki Konishi
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yuka Ohshiro
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shimpei Uraguchi
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Mitsuo Tanabe
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshinori Kobayashi
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kenji Sakamoto
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan; Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Tsutomu Nakahara
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Megumi Yamamoto
- Department of Environment and Public Health, National Institute for Minamata Disease, 4058-18, Hama, Minamata, Kumamoto 867-0008, Japan
| | - Masako Kiyono
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
3
|
Pall ML. Central Causation of Autism/ASDs via Excessive [Ca 2+]i Impacting Six Mechanisms Controlling Synaptogenesis during the Perinatal Period: The Role of Electromagnetic Fields and Chemicals and the NO/ONOO(-) Cycle, as Well as Specific Mutations. Brain Sci 2024; 14:454. [PMID: 38790433 PMCID: PMC11119459 DOI: 10.3390/brainsci14050454] [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: 03/08/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The roles of perinatal development, intracellular calcium [Ca2+]i, and synaptogenesis disruption are not novel in the autism/ASD literature. The focus on six mechanisms controlling synaptogenesis, each regulated by [Ca2+]i, and each aberrant in ASDs is novel. The model presented here predicts that autism epidemic causation involves central roles of both electromagnetic fields (EMFs) and chemicals. EMFs act via voltage-gated calcium channel (VGCC) activation and [Ca2+]i elevation. A total of 15 autism-implicated chemical classes each act to produce [Ca2+]i elevation, 12 acting via NMDA receptor activation, and three acting via other mechanisms. The chronic nature of ASDs is explained via NO/ONOO(-) vicious cycle elevation and MeCP2 epigenetic dysfunction. Genetic causation often also involves [Ca2+]i elevation or other impacts on synaptogenesis. The literature examining each of these steps is systematically examined and found to be consistent with predictions. Approaches that may be sed for ASD prevention or treatment are discussed in connection with this special issue: The current situation and prospects for children with ASDs. Such approaches include EMF, chemical avoidance, and using nutrients and other agents to raise the levels of Nrf2. An enriched environment, vitamin D, magnesium, and omega-3s in fish oil may also be helpful.
Collapse
Affiliation(s)
- Martin L Pall
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
4
|
Fujimura M. Gabapentin improves neuropathic pain in Minamata disease model rats. Environ Health Prev Med 2024; 29:31. [PMID: 38825526 PMCID: PMC11157338 DOI: 10.1265/ehpm.24-00035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Methylmercury (MeHg), the causative agent of Minamata disease, damages the cranial nervous system and causes specific sensory disturbances, especially hypoesthesia, in the extremities. However, recent reports demonstrate that patients with chronic Minamata disease conversely develop neuropathic pain in the lower extremities. Studies on our established Minamata disease model rats showed that MeHg-mediated neurodegeneration might induce neuropathic pain by over time through inducing rewiring with neuronal activation in the somatosensory cortex via microglial activation in the spinal dorsal horn. METHODS In this study, the effects of gabapentin, a potentially effective treatment for neuropathic pain, was evaluated using this Minamata disease model rats. To further elucidate the mechanism of its medicinal effects, histochemical and biochemical analyses of the nervous system of Minamata disease model rats were conducted. RESULTS Gabapentin treatment restored the reduction in the pain threshold caused by MeHg exposure in rats. Histochemical and biochemical analyses revealed that gabapentin showed no effect on MeHg-induced neurodegeneration in entire nervous system and microglial activation in the spinal dorsal horn. However, it was shown that gabapentin may reduce excessive synaptogenesis through its antagonist action on the alpha2-delta-1 subunit of calcium channels in the somatosensory cortex. CONCLUSIONS These results indicate that gabapentin may alleviated neuropathic pain in MeHg poisoning, as typified by Minamata disease, by reversibly modulation synaptic rewiring in the somatosensory cortex.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Minamata, Japan
| |
Collapse
|
5
|
Camacho JA, Welch B, Sprando RL, Hunt PR. Reproductive-Toxicity-Related Endpoints in C. elegans Are Consistent with Reduced Concern for Dimethylarsinic Acid Exposure Relative to Inorganic Arsenic. J Dev Biol 2023; 11:18. [PMID: 37218812 PMCID: PMC10204422 DOI: 10.3390/jdb11020018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Exposures to arsenic and mercury are known to pose significant threats to human health; however, the effects specific to organic vs. inorganic forms are not fully understood. Caenorhabditis elegans' (C. elegans) transparent cuticle, along with the conservation of key genetic pathways regulating developmental and reproductive toxicology (DART)-related processes such as germ stem cell renewal and differentiation, meiosis, and embryonic tissue differentiation and growth, support this model's potential to address the need for quicker and more dependable testing methods for DART hazard identification. Organic and inorganic forms of mercury and arsenic had different effects on reproductive-related endpoints in C. elegans, with methylmercury (meHgCl) having effects at lower concentrations than mercury chloride (HgCl2), and sodium arsenite (NaAsO2) having effects at lower concentrations than dimethylarsinic acid (DMA). Progeny to adult ratio changes and germline apoptosis were seen at concentrations that also affected gravid adult gross morphology. For both forms of arsenic tested, germline histone regulation was altered at concentrations below those that affected progeny/adult ratios, while concentrations for these two endpoints were similar for the mercury compounds. These C. elegans findings are consistent with corresponding mammalian data, where available, suggesting that small animal model test systems may help to fill critical data gaps by contributing to weight of evidence assessments.
Collapse
Affiliation(s)
- Jessica A. Camacho
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA
| | | | | | | |
Collapse
|
6
|
Fujimura M. Fasudil, a ROCK inhibitor, prevents neuropathic pain in Minamata disease model rats. Toxicol Lett 2022; 371:38-45. [PMID: 36244566 DOI: 10.1016/j.toxlet.2022.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/24/2022] [Accepted: 10/11/2022] [Indexed: 02/13/2023]
Abstract
Methylmercury (MeHg), an environmental toxicant, is known to cause sensory impairment by inducing neurodegeneration of sensory nervous systems. However, in recent years, it has been revealed that neuropathic pain occurs in the chronic phase of MeHg poisoning, that is, in current Minamata disease patients. Our recent study using Minamata disease model rats demonstrated that MeHg-mediated neurodegeneration in the sensory nervous system may induce inflammatory microglia production in the dorsal horn of the spinal cord and subsequent somatosensory cortical rewiring, leading to neuropathic pain. We hypothesized that inhibition of the Rho-associated coiled coil-forming protein kinase (ROCK) pathway could prevent MeHg-induced neuropathic pain because the ROCK pathway is known to be involved in inducing the production of inflammatory microglia. Here, we showed for the first time that Fasudil, a ROCK inhibitor, can prevent neuropathic pain in Minamata disease model rats. In this model, Fasudil significantly suppressed nerve injury-induced inflammatory microglia production in the dorsal horn of the spinal cord and prevented subsequent somatosensory cortical rewiring. These results suggest that the ROCK pathway is involved in the onset and development of neuropathic pain in the chronic phase of Minamata disease, and that its inhibition is effective in pain prevention.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan.
| |
Collapse
|
7
|
Fujimura M, Usuki F. Cellular Conditions Responsible for Methylmercury-Mediated Neurotoxicity. Int J Mol Sci 2022; 23:ijms23137218. [PMID: 35806222 PMCID: PMC9266708 DOI: 10.3390/ijms23137218] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Methylmercury (MeHg) is a widely known environmental pollutant that causes severe neurotoxicity. MeHg-induced neurotoxicity depends on various cellular conditions, including differences in the characteristics of tissues and cells, exposure age (fetal, childhood, or adulthood), and exposure levels. Research has highlighted the importance of oxidative stress in the pathogenesis of MeHg-induced toxicity and the site- and cell-specific nature of MeHg-induced neurotoxicity. The cerebellar granule cells and deeper layer cerebrocortical neurons are vulnerable to MeHg. In contrast, the hippocampal neurons are resistant to MeHg, even at high mercury accumulation levels. This review summarizes the mechanisms underlying MeHg-mediated intracellular events that lead to site-specific neurotoxicity. Specifically, we discuss the mechanisms associated with the redox ability, neural outgrowth and synapse formation, cellular signaling pathways, epigenetics, and the inflammatory conditions of microglia.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto 867-0008, Japan
- Correspondence: ; Tel.: +81-966-63-3111; Fax: +81-966-61-1145
| | - Fusako Usuki
- Division of Neuroimmunology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan;
| |
Collapse
|
8
|
Fujimura M, Unoki T. Preliminary evaluation of the mechanism underlying vulnerability/resistance to methylmercury toxicity by comparative gene expression profiling of rat primary cultured cerebrocortical and hippocampal neurons. J Toxicol Sci 2022; 47:211-219. [PMID: 35527009 DOI: 10.2131/jts.47.211] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Methylmercury (MeHg), an environmentally toxic substance, causes site-specific neuronal cell death; while MeHg exposure causes death in cerebrocortical neurons, interestingly, it does not in hippocampal neurons, which are generally considered to be vulnerable to toxic substances. This phenomenon of site-specific neuronal cell death can be reproduced in animal experiments; however, the mechanism underlying the resistance of hippocampal neurons to MeHg toxicity has not been clarified. In this study, we comparatively analyzed the response to MeHg exposure in terms of viability and the expression characteristics of primary cultured cerebrocortical neurons and hippocampal neurons derived from fetal rat brain. Neuronal differentiated hippocampal neurons were more resistant to MeHg toxicity than cerebrocortical neurons, as indicated by a 2‒3 fold higher half-maximal inhibitory concentration (IC50; 3.3 μM vs. 1.2 μM), despite similar intracellular mercury concentrations in both neuronal cell types. Comprehensive RNA sequencing-based gene expression analysis of non-MeHg-exposed cells revealed that 80 out of 15,208 genes showed at least 10-fold higher expression in hippocampal neurons than in cerebrocortical neurons, whereas six genes showed at least 10-fold higher expression in cerebrocortical neurons than in hippocampal neurons. In particular, genes related to neuronal function, including those encoding transthyretin and brain-derived neurotrophic factor, showed approximately 50-fold higher expression in hippocampal neurons than in cerebrocortical neurons. In conclusion, the resistance of hippocampal neurons to MeHg toxicity may be related to the high expression of neuronal function-related proteins.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease
| | - Takamitsu Unoki
- Department of Basic Medical Sciences, National Institute for Minamata Disease
| |
Collapse
|
9
|
Unoki T, Akiyama M, Shinkai Y, Kumagai Y, Fujimura M. Spatio-temporal distribution of reactive sulfur species during methylmercury exposure in the rat brain. J Toxicol Sci 2022; 47:31-37. [PMID: 34987139 DOI: 10.2131/jts.47.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Brain susceptibility to methylmercury (MeHg) is developmentally and regionally specific in both humans and rodents, but the mechanism is not well clarified. Reactive sulfur species (RSS) with high nucleophilicity can react with MeHg, leading to the formation of a less toxic metabolite bismethylmercury sulfide, thus exerting cytoprotection. In this study, we assessed the variation of RSS content in the rat brain and evaluated its relevance in sensitivity to MeHg. Analyses of fetal/juvenile rat brains showed low RSS levels in early developmental stages. Site-specific analysis of adult rat brains revealed that cerebellar RSS levels were lower than those of the hippocampus. Microscopically, RSS levels of the granular cell layer were lower than those of the molecular layer in the cerebellum. Thus, low RSS levels corresponded with age and site of the brain that is vulnerable to MeHg. Taken together with the finding that brain RSS were consumed during MeHg exposure, these results indicate that RSS is a factor that defines the specificity of MeHg vulnerability in the brain.
Collapse
Affiliation(s)
- Takamitsu Unoki
- Department of Basic Medical Sciences, National Institute for Minamata Disease
| | - Masahiro Akiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University
| | - Yasuhiro Shinkai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba
| | - Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease
| |
Collapse
|
10
|
Gunderson JT, Peppriell AE, Krout IN, Vorojeikina D, Rand MD. Neuroligin-1 Is a Mediator of Methylmercury Neuromuscular Toxicity. Toxicol Sci 2021; 184:236-251. [PMID: 34546366 DOI: 10.1093/toxsci/kfab114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmercury (MeHg) is a developmental toxicant capable of eliciting neurocognitive and neuromuscular deficits in children with in utero exposure. Previous research in Drosophila melanogaster uncovered that developmental MeHg exposure simultaneously targets the developing musculature and innervating motor neuron in the embryo, along with identifying Drosophila neuroligin 1 (nlg1) as a gene associated with developmental MeHg sensitivity. Nlg1 and its transsynaptic partner neurexin 1 (Nrx1) are critical for axonal arborization and NMJ maturation. We investigated the effects of MeHg exposure on indirect flight muscle (IFM) morphogenesis, innervation, and function via flight assays and monitored the expression of NMJ-associated genes to characterize the role of Nlg1 mediating the neuromuscular toxicity of MeHg. Developmental MeHg exposure reduced the innervation of the IFMs, which corresponded with reduced flight ability. In addition, nlg1 expression was selectively reduced during early metamorphosis, whereas a subsequent increase was observed in other NMJ-associated genes, including nrx1, in late metamorphosis. Developmental MeHg exposure also resulted in persistent reduced expression of most nlg and nrx genes during the first 11 days of adulthood. Transgenic modulation of nlg1 and nrx1 revealed that developing muscle is particularly sensitive to nlg1 levels, especially during the 20-36-h window of metamorphosis with reduced nlg1 expression resulting in adult flight deficits. Muscle-specific overexpression of nlg1 partially rescued MeHg-induced deficits in eclosion and flight. We identified Nlg1 as a muscle-specific, NMJ structural component that can mediate MeHg neuromuscular toxicity resulting from early life exposure.
Collapse
Affiliation(s)
- Jakob T Gunderson
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Ashley E Peppriell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Ian N Krout
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| |
Collapse
|
11
|
Fujimura M, Usuki F, Nakamura A. Methylmercury induces hyperalgesia/allodynia through spinal cord dorsal horn neuronal activation and subsequent somatosensory cortical circuit formation in rats. Arch Toxicol 2021; 95:2151-2162. [PMID: 33847776 DOI: 10.1007/s00204-021-03047-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/01/2021] [Indexed: 12/30/2022]
Abstract
Methylmercury (MeHg) is known to cause serious neurological deficits in humans. In this study, we investigated the occurrence of MeHg-mediated neuropathic pain and identified the underlying pathophysiological mechanism in a rat model of MeHg exposure. Rats were exposed to MeHg (20 ppm in drinking water) for 3 weeks. Neurological damage was observed in the primary afferent neuronal system, including the dorsal root nerve and the dorsal column of the spinal cord. The MeHg-exposed rats showed hyperalgesia/allodynia, compared to controls, as evidenced by a significant decrease in the threshold of mechanical pain evaluated using an algometer with calibrated forceps. Immunohistochemistry revealed the accumulation of activated microglia in the dorsal root nerve, dorsal column, and dorsal horn of the spinal cord. Western blot analyses of the dorsal part of the spinal cord demonstrated an increase in inflammotoxic and inflammatory cytokines and a neuronal activation related protein, phospho-CRE bunding protein (CREB). The results suggest that dorsal horn neuronal activation was mediated by inflammatory factors excreted by accumulated microglia. Furthermore, analyses of the cerebral cortex demonstrated increased expression of phospho-CREB and thrombospondin-1, which is known to be an important factor for excitatory synapse formation, specifically in the somatosensory cortical area. In addition, the expression of pre- and post-synaptic markers was increased in this cortex area. These results suggested that the new cortical circuit was wired specifically in the somatosensory cortex. In conclusion, MeHg-mediated dorsal horn neuronal activation with inflammatory microglia might induce somatosensory cortical rewiring, leading to hyperalgesia/allodynia.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto, Japan.
| | - Fusako Usuki
- Division of Neuroimmunology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Atsushi Nakamura
- Department of Clinical Medicine, National Institute for Minamata Disease, Kumamoto, Japan
| |
Collapse
|
12
|
Rand MD, Conrad K, Marvin E, Harvey K, Henderson D, Tawil R, Sobolewski M, Cory-Slechta DA. Developmental exposure to methylmercury and resultant muscle mercury accumulation and adult motor deficits in mice. Neurotoxicology 2020; 81:1-10. [PMID: 32735808 PMCID: PMC7708410 DOI: 10.1016/j.neuro.2020.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 11/20/2022]
Abstract
Developmental methylmercury (MeHg) exposure can have lasting consequences on neural development and motor function across the lifespan. Recent evidence for MeHg targeting of myogenic pathways has drawn attention to the possibility that developing skeletal muscle plays a role in the motor deficits stemming from early life MeHg exposure. In this study we examined a potential role for muscle in influencing MeHg developmental toxicity in offspring of female mice exposed to MeHg via drinking water. Dams had access to 0, 0.5 or 5.0 ppm MeHg chloride in drinking water from two weeks prior to mating through weaning. Blood, brain and muscle tissue was harvested from dams at weaning and pups at postnatal days (PND) 6, 21 and 60 for analysis of total Hg. Muscle tissue sections were examined with histological stains. Behavioral testing of offspring was conducted at PND 60 and included locomotor activity, inverted screen, grip strength and rotarod tests to assess motor function. Total Hg (tHg) levels in dam muscles at weaning were 1.7-3-fold higher than Hg levels in blood or brain. In PND6 male and female pups, muscle and brain tHg levels were 2 to 4-fold higher than blood tHg. Brain tHg levels decreased more rapidly than muscle tHg levels between PND 6 and 21. Premised on modeling of growth dilution, brain tissue demonstrated an elimination of tHg while muscle tissue exhibited a net uptake of tHg between PND 6 and 21. Despite overall elevated Hg levels in developing muscle, no gross morphological or cytological phenotypes were observed in muscle at PND 60. At the higher MeHg dose, grip strength was reduced in both females and males at PND 60, whereas only male specific deficits were observed in locomotor activity and inverted screen tests with marginally significant deficits on rotarod. These findings highlight a potential role for developing skeletal muscle in mediating the neuromuscular insult of early life MeHg exposure.
Collapse
Affiliation(s)
| | | | - Elena Marvin
- Department of Environmental Medicine, United States
| | | | - Don Henderson
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Rabi Tawil
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | | | | |
Collapse
|
13
|
Gunderson JT, Peppriell AE, Vorojeikina D, Rand MD. Tissue-specific Nrf2 signaling protects against methylmercury toxicity in Drosophila neuromuscular development. Arch Toxicol 2020; 94:4007-4022. [PMID: 32816092 DOI: 10.1007/s00204-020-02879-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Methylmercury (MeHg) can elicit cognitive and motor deficits due to its developmental neuro- and myotoxic properties. While previous work has demonstrated that Nrf2 antioxidant signaling protects from MeHg toxicity, in vivo tissue-specific studies are lacking. In Drosophila, MeHg exposure shows greatest developmental toxicity in the pupal stage resulting in failed eclosion (emergence of adults) and an accompanying 'myosphere' phenotype in indirect flight muscles (IFMs). To delineate tissue-specific contributions to MeHg-induced motor deficits, we investigated the potential of Nrf2 signaling in either muscles or neurons to moderate MeHg toxicity. Larva were exposed to various concentrations of MeHg (0-20 µM in food) in combination with genetic modulation of the Nrf2 homolog cap-n-collar C (CncC), or its negative regulator Keap1. Eclosion behavior was evaluated in parallel with the morphology of two muscle groups, the thoracic IFMs and the abdominal dorsal internal oblique muscles (DIOMs). CncC signaling activity was reported with an antioxidant response element construct (ARE-GFP). We observed that DIOMs are distinguished by elevated endogenous ARE-GFP expression, which is only transiently seen in the IFMs. Dose-dependent MeHg reductions in eclosion behavior parallel formation of myospheres in the DIOMs and IFMs, while also increasing ARE-GFP expression in the DIOMs. Modulating CncC signaling via muscle-specific Keap1 knockdown and upregulation gives a rescue and exacerbation, respectively, of MeHg effects on eclosion and myospheres. Interestingly, muscle-specific CncC upregulation and knockdown both induce lethality. In contrast, neuron-specific upregulation of CncC, as well as Keap1 knockdown, rescued MeHg effects on eclosion and myospheres. Our findings indicate that enhanced CncC signaling localized to either muscles or neurons is sufficient to rescue muscle development and neuromuscular function from a MeHg insult. Additionally, there may be distinct roles for CncC signaling in myo-morphogenesis.
Collapse
Affiliation(s)
- Jakob T Gunderson
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ashley E Peppriell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
14
|
Fu X, Yang X, Du X, Cui Q. Deciphering the possible role of H2O2 in methylmercury-induced neurotoxicity in Xenopus laevis. Mol Cell Toxicol 2020. [DOI: 10.1007/s13273-020-00082-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Abstract
Backgrounds
Methylmercury (MeHg) is regarded as a developmental neurotoxicant but the detailed mechanism remains not completely clear.
Methods
The Xenopus laevis embryos were exposed to methylmercury chloride and the expression of neurodevelopment and oxidative stress genes was detected by qRT-PCR or Western blotting. PC12 cells were exposed to various levels of H2O2, and then cell cycle, neurite length, neurodevelopment-related genes, protein expression of apoptosis and autophagy were detected.
Results
The genes of neurodevelopment and oxidative stress were disrupted by methylmercury chloride and H2O2 were increased interestingly in X. laevis embryos. Then, PC12 cells were exposed to H2O2 and the results showed the cell cycle, neurite length, and neurodevelopment-related genes, the proteins apoptosis and autophagy were changed.
Conclusion
These results supported the idea that neurodevelopment-related gene expression was regulated by oxidative stress and that apoptosis and autophagy pathways were activated by H2O2 and involved in methylmercury neurotoxicity.
Collapse
|
15
|
Rosa-Silva HTD, Panzenhagen AC, Schmidtt V, Alves Teixeira A, Espitia-Pérez P, de Oliveira Franco Á, Mingori M, Torres-Ávila JF, Schnorr CE, Hermann PRS, Moraes DP, Almeida RF, Moreira JCF. Hepatic and neurobiological effects of foetal and breastfeeding and adulthood exposure to methylmercury in Wistar rats. CHEMOSPHERE 2020; 244:125400. [PMID: 31809933 DOI: 10.1016/j.chemosphere.2019.125400] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/20/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
Methylmercury (MeHg) is an organic bioaccumulated mercury derivative that strongly affects the environment and represents a public health problem primarily to riparian communities in South America. Our objective was to investigate the hepatic and neurological effects of MeHg exposure during the phases foetal and breast-feeding and adult in Wistar rats. Wistar rats (n = 10) were divided into 3 groups. Control group received mineral oil; The simple exposure (SE) group was exposed only in adulthood (0.5 mg/kg/day); and double exposure (DE) was pre-exposed to MeHg 0.5 mg/kg/day during pregnancy and breastfeeding (±40 days) and re-exposed to MeHg for 45 days from day 100. After, we evaluated possible abnormalities. Behavioral and biochemical parameters in liver and occipital cortex (CO), markers of liver injury, redox and AKT/GSK3β/mTOR signaling pathway. Our results showed that both groups treated with MeHg presented significant alterations, such as decreased locomotion and exploration and impaired visuospatial perception. The rats exposed to MeHg showed severe liver damage and increased hepatic glycogen concentration. The MeHg groups showed significant impairment in redox balance and oxidative damage to liver macromolecules and CO. MeHg upregulated the AKT/GSK3β/mTOR pathway and the phosphorylated form of the Tau protein. In addition, we found a reduction in NeuN and GFAP immunocontent. These results represent the first approach to the hepatotoxic and neural effects of foetal and adult MeHg exposure.
Collapse
Affiliation(s)
- Helen Tais da Rosa-Silva
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Alana Castro Panzenhagen
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victória Schmidtt
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Alexsander Alves Teixeira
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Pedro Espitia-Pérez
- Facultad de Ciencias de la Salud, Laboratorio de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Córdoba, Colombia
| | - Álvaro de Oliveira Franco
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Moara Mingori
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - José F Torres-Ávila
- Unit for Development and Innovation in Genetics and Molecular Biology, Universidad Simón Bolívar, Barranquilla, Atlántico, Colombia
| | - Carlos Eduardo Schnorr
- Departamento de Civil y Ambiental, Programa de Ingeniería Ambiental, Universidad de la Costa, Barranquilla, Atlántico, Colombia
| | - Paolla Rissi Silva Hermann
- Instituto de Química, Sala 103, Campus do Vale, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diogo Pompéu Moraes
- Instituto de Química, Sala 103, Campus do Vale, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Roberto Farina Almeida
- Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
16
|
Yang L, Zhang Y, Wang F, Luo Z, Guo S, Strähle U. Toxicity of mercury: Molecular evidence. CHEMOSPHERE 2020; 245:125586. [PMID: 31881386 DOI: 10.1016/j.chemosphere.2019.125586] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/28/2019] [Accepted: 12/08/2019] [Indexed: 05/25/2023]
Abstract
Minamata disease in Japan and the large-scale poisoning by methylmercury (MeHg) in Iraq caused wide public concerns about the risk emanating from mercury for human health. Nowadays, it is widely known that all forms of mercury induce toxic effects in mammals, and increasing evidence supports the concern that environmentally relevant levels of MeHg could impact normal biological functions in wildlife. The information of mechanism involved in mercurial toxicity is growing but knowledge gaps still exist between the adverse effects and mechanisms of action, especially at the molecular level. A body of data obtained from experimental studies on mechanisms of mercurial toxicity in vivo and in vitro points to that disruption of the antioxidant system may play an important role in the mercurial toxic effects. Moreover, the accumulating evidence indicates that signaling transduction, protein or/and enzyme activity, and gene regulation are involving in mediating toxic and adaptive response to mercury exposure. We conducted here a comprehensive review of mercurial toxic effects on wildlife and human, in particular synthesized key findings of molecular pathways involved in mercurial toxicity from the cells to human. We discuss the molecular evidence related mercurial toxicity to the adverse effects, with particular emphasis on the gene regulation. The further studies relying on Omic analysis connected to adverse effects and modes of action of mercury will aid in the evaluation and validation of causative relationship between health outcomes and gene expression.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| | - Yuanyuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Zidie Luo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Uwe Strähle
- Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
17
|
Fujimura M, Usuki F, Nakamura A. Fasudil, a Rho-Associated Coiled Coil-Forming Protein Kinase Inhibitor, Recovers Methylmercury-Induced Axonal Degeneration by Changing Microglial Phenotype in Rats. Toxicol Sci 2020; 168:126-136. [PMID: 30462329 DOI: 10.1093/toxsci/kfy281] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmercury (MeHg) is an environmental neurotoxicant that induces neuropathological changes. In this study, we established chronic MeHg-intoxicated rats. These rats survived, and sustained MeHg-induced axonal degeneration, including the dorsal root nerve and the dorsal column of the spinal cord; these changes persisted 12 weeks after MeHg withdrawal. We demonstrated for the first time the restorative effect of Fasudil, a specific inhibitor of Rho-associated coiled coil-forming protein kinase, on axonal degeneration and corresponding neural dysfunction in the established chronic MeHg-intoxicated rats. To investigate the mechanism of this restorative effect, we focused on the expression of Rho protein families. This was supported by our previous study, which demonstrated that cotreatment with Fasudil prevented axonal degeneration by mitigating neurite extension/retraction incoordination caused by MeHg-induced suppression of Rac1 in vitro and in subacute MeHg-intoxicated rats. However, the mechanism of the restorative effect of Fasudil on axonal degeneration in chronic MeHg-intoxicated rats differed from MeHg-mediated neuritic extension/retraction incoordination. We found that the restorative effect of Fasudil was caused by the Fasudil-induced change of microglial phenotype, from proinflammatory to anti-inflammatory; moreover, Fasudil suppressed Rho-associated coiled coil-forming protein kinase activity. Treatment with Fasudil decreased the expression of proinflammatory factors, including tumor necrosis factor-α, inducible nitric oxide synthase, interleukin-1β, and interleukin-6; furthermore, it inactivated the nuclear factor kappa-light-chain-enhancer of activated B cells pathway. Additionally, Fasudil treatment was associated with increased levels of anti-inflammatory factors arginase-1 and interleukin-10. These results suggest that Rho-associated coiled coil-forming protein kinase inhibition may recover MeHg-mediated axonal degeneration and neural dysfunction in chronic MeHg intoxication.
Collapse
Affiliation(s)
| | - Fusako Usuki
- Department of Clinical Medicine, National Institute for Minamata Disease, Kumamoto 867-0008, Japan
| | - Atsushi Nakamura
- Department of Clinical Medicine, National Institute for Minamata Disease, Kumamoto 867-0008, Japan
| |
Collapse
|
18
|
Pregnant rats exposed to low-level methylmercury exhibit cerebellar synaptic and neuritic remodeling during the perinatal period. Arch Toxicol 2020; 94:1335-1347. [DOI: 10.1007/s00204-020-02696-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/02/2020] [Indexed: 12/14/2022]
|
19
|
Rodrigues KE, de Oliveira FR, Barbosa BRC, Paraense RSO, Bannwart CM, Pinheiro BG, Botelho ADS, Muto NA, do Amarante CB, Hamoy M, Macchi BDM, Maia CDSF, do Prado AF, do Nascimento JLM. Aqueous Coriandrum sativum L. extract promotes neuroprotection against motor changes and oxidative damage in rat progeny after maternal exposure to methylmercury. Food Chem Toxicol 2019; 133:110755. [PMID: 31408720 DOI: 10.1016/j.fct.2019.110755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/03/2019] [Accepted: 08/08/2019] [Indexed: 01/18/2023]
Abstract
This study aimed to investigate the effects of Coriandrum sativum aqueous extract (CSAE) on the rat progeny of mothers exposed to methylmercury (MeHg). The presence of bioactive compounds and CSAE's antioxidant capacity been evaluated, and the offspring were assessed for their total mercury levels, motor behavioral parameters and oxidative stress in the cerebellum. The analysis of the bioactive compounds revealed significant amounts of polyphenols, flavonoids, and anthocyanins, as well as a variety of minerals. A DPPH test showed the CSAE had important antioxidant activity. The MeHg + CSAE group performed significantly better spontaneous locomotor activity, palmar grip strength, balance, and motor coordination in behavioral tests compared the MeHg group, as well as in the parameters of oxidative stress, with similar results to those of the control group. The MeHg + CSAE group also had significantly reduced mercury levels in comparison to the MeHg group. Based on the behavioral tests, which detected large locomotor, balance, and coordination improvements, as well as a reduction in oxidative stress, we conclude that CSAE had positive functional results in the offspring of rats exposed to MeHg.
Collapse
Affiliation(s)
- Keuri Eleutério Rodrigues
- Neuroscience and Cellular Biology Post Graduation Program, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil; Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Fábio Rodrigues de Oliveira
- Neuroscience and Cellular Biology Post Graduation Program, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil; Bromatology and Quality Control Laboratory, Health and Biological Sciences Department, Federal University of Amapa (UNIFAP), Macapa, Ap, Brazil
| | - Benilson Ramos Cassunde Barbosa
- Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Ricardo S Oliveira Paraense
- Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Cahy Manoel Bannwart
- Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Bruno Gonçalves Pinheiro
- Behavioral and Inflammatory Pharmacology Laboratory, Health Sciences Institute, Pharmacy College, Federal University of Para, Belem, PA, Brazil
| | | | - Nilton Akio Muto
- Amazonian Bioactive Compounds Valorization Center, Federal University of Para, Belem, PA, Brazil
| | | | - Moises Hamoy
- Natural Products' Toxicology and Pharmacology Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Barbarella de Matos Macchi
- Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - Cristiane do Socorro Ferraz Maia
- Behavioral and Inflammatory Pharmacology Laboratory, Health Sciences Institute, Pharmacy College, Federal University of Para, Belem, PA, Brazil
| | - Alejandro Ferraz do Prado
- Structural Biology Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil
| | - José Luiz Martins do Nascimento
- Neuroscience and Cellular Biology Post Graduation Program, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil; Molecular and Cellular Neurochemistry Laboratory, Biological Sciences Institute, Federal University of Para, Belem, PA, Brazil; National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, RJ, Brazil; Pharmaceutical Sciences Post Graduation Program, Health and Biological Sciences Department, Federal University of Amapa (UNIFAP), Macapa, Ap, Brazil.
| |
Collapse
|
20
|
Reardon AJF, Karathra J, Ribbenstedt A, Benskin JP, MacDonald AM, Kinniburgh DW, Hamilton TJ, Fouad K, Martin JW. Neurodevelopmental and Metabolomic Responses from Prenatal Coexposure to Perfluorooctanesulfonate (PFOS) and Methylmercury (MeHg) in Sprague-Dawley Rats. Chem Res Toxicol 2019; 32:1656-1669. [PMID: 31340646 DOI: 10.1021/acs.chemrestox.9b00192] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Methylmercury (MeHg) and perfluorooctanesulfonate (PFOS) are major contaminants of human blood that are both common in dietary fish, thereby raising questions about their combined impact on human development. Here, pregnant Sprague-Dawley rats ingested a daily dose, from gestational day 1 through to weaning, of either 1 mg/kg bw PFOS (PFOS-only), 1 mg/kg MeHg (MeHg-only), a mixture of 0.1 mg/kg PFOS and 1 mg/kg MeHg (Low-Mix), or of 1 mg/kg of PFOS and 1 mg/kg MeHg (High-Mix). Newborns were monitored for physical milestones and reflexive developmental responses, and in juveniles the spontaneous activity, anxiety, memory, and cognition were assessed. Targeted metabolomics of 199 analytes was applied to sectioned brain regions of juvenile offspring. Newborns in the High-Mix group had decreased weight gain as well as delayed reflexes and innate behavioral responses compared to controls and individual chemical groups indicating a toxicological interaction on early development. In juveniles, cumulative mixture effects increased in a dose-dependent manner in tests of anxiety-like behavior. However, other developmental test results suggested antagonism, as PFOS-only and MeHg-only juveniles had increased hyperactivity and thigmotaxic behavior, respectively, but fewer effects in Low-Mix and High-Mix groups. Consistent with these behavioral observations, a pattern of antagonism was also observed in neurochemicals measured in rat cortex, as PFOS-only and MeHg-only juveniles had altered concentrations of metabolites (e.g., lipids, amino acids, and biogenic amines), while no changes were evident in the combined exposures. The cortical metabolites altered in PFOS-only and MeHg-only exposed groups are involved in inhibitory and excitatory neurotransmission. These proof-of-principle findings at relatively high doses indicate the potential for toxicological interaction between PFOS and MeHg, with developmental-stage specific effects. Future mixture studies at lower doses are warranted, and prospective human birth cohorts should consider possible confounding effects from PFOS and mercury exposure on neurodevelopment.
Collapse
Affiliation(s)
- Anthony J F Reardon
- Department of Laboratory Medicine and Pathology , University of Alberta , Edmonton , Alberta T6G 2G3 , Canada
| | - Jacqueline Karathra
- Department of Laboratory Medicine and Pathology , University of Alberta , Edmonton , Alberta T6G 2G3 , Canada
| | - Anton Ribbenstedt
- Department of Environmental Science and Analytical Chemistry (ACES) , Stockholm University , Stockholm SE-11418 , Sweden
| | - Jonathan P Benskin
- Department of Environmental Science and Analytical Chemistry (ACES) , Stockholm University , Stockholm SE-11418 , Sweden
| | - Amy M MacDonald
- Alberta Centre for Toxicology , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
| | - David W Kinniburgh
- Alberta Centre for Toxicology , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
| | - Trevor J Hamilton
- Department of Psychology , MacEwan University , Edmonton , Alberta T5J 4S2 , Canada
| | - Karim Fouad
- Department of Physical Therapy , University of Alberta , Edmonton , Alberta T6G 2G4 , Canada
| | - Jonathan W Martin
- Department of Laboratory Medicine and Pathology , University of Alberta , Edmonton , Alberta T6G 2G3 , Canada.,Department of Environmental Science and Analytical Chemistry (ACES) , Stockholm University , Stockholm SE-11418 , Sweden
| |
Collapse
|
21
|
Kaushal P, Kumar P, Dhar P. Ameliorative role of antioxidant supplementation on sodium-arsenite induced adverse effects on the developing rat cerebellum. J Ayurveda Integr Med 2019; 11:455-463. [PMID: 30635247 PMCID: PMC7772504 DOI: 10.1016/j.jaim.2018.02.138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/01/2018] [Accepted: 02/08/2018] [Indexed: 11/26/2022] Open
Abstract
Background Arsenic is an environmental contaminant of global concern. Consumption of ground water contaminated with inorganic arsenic (iAs) continues to be the major source of its exposure. The developing nervous system is especially vulnerable to environmental insults due to its higher rate of oxygen consumption and provision of weaker antioxidant (AOX) machinery. Objective Since oxidative stress has been reported as one of the major factors underlying iAs induced toxicity, the aim of the present study is to study the effect of two AOXs i.e., Alpha Lipoic Acid (ALA) and Curcumin (Cur) in developing cerebellum of rats exposed to arsenic during postnatal period. Materials and Methods The study was carried out on mother reared neonatal rat pups grouped as normal (Ia) and sham (vehicle) controls (Ib,c,d), while the experimental groups IIa/ IIb received sodium arsenite (NaAsO2) [(1.5/2.5 mg/kg body weight (bw)] alone or along with ALA (70 mg/kg bw)- IIIa/ IIIb or along with Cur (150 mg/kg bw)- IVa/ IVb. Behavioural, biochemical and immunohistochemical procedures were carried out to understand the underlying mechanisms. Results The observations indicated deficits in locomotor function, accumulation of iAs, increased levels of oxidative stress markers along with downregulation of the expression of proteins closely associated with synaptic functioning (Synaptophysin and Postsynaptic density protein95) in the cerebellum of iAs treated animals. Substantial recovery in all these parameters was observed in AOX co-treated groups. Conclusion Our results support the potential of ALA and Cur in amelioration of iAs induced developmental neurotoxicity. ALA and Cur can be proposed as dietary adjuvants amongst populations inhabiting areas with high iAs contamination as a safe and cost effective antidotes.
Collapse
Affiliation(s)
- Parul Kaushal
- Department of Anatomy, All Institute of Medical Sciences, New Delhi 110029, India
| | - Pavan Kumar
- Department of Anatomy, All Institute of Medical Sciences, New Delhi 110029, India
| | - Pushpa Dhar
- Department of Anatomy, All Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
22
|
Neurobehavioral and oxidative stress alterations following methylmercury and retinyl palmitate co-administration in pregnant and lactating rats and their offspring. Neurotoxicology 2018; 69:164-180. [DOI: 10.1016/j.neuro.2018.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/26/2022]
|
23
|
Ghizoni H, Ventura M, Colle D, Gonçalves CL, de Souza V, Hartwig JM, Santos DB, Naime AA, Cristina de Oliveira Souza V, Lopes MW, Barbosa F, Brocardo PS, Farina M. Effects of perinatal exposure to n-3 polyunsaturated fatty acids and methylmercury on cerebellar and behavioral parameters in mice. Food Chem Toxicol 2018; 120:603-615. [DOI: 10.1016/j.fct.2018.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
|
24
|
Delayed neurochemical effects of prenatal exposure to MeHg in the cerebellum of developing rats. Toxicol Lett 2017; 284:161-169. [PMID: 29258870 DOI: 10.1016/j.toxlet.2017.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/03/2017] [Accepted: 12/09/2017] [Indexed: 01/09/2023]
Abstract
Human fetuses and neonates are particularly vulnerable to methylmercury (MeHg)-induced brain damage and are sensitive even to low exposure levels. Previous work of our group evidence that prenatal exposure to MeHg causes cognitive and behavioral alterations and disrupt hippocampus signaling. The current study aimed to investigate the effect of gestational exposure of rats to MeHg at low doses (1 or 2 mg/kg) on parameters of redox imbalance and key signaling pathways in the cerebellum of their offspring. Pregnant females received MeHg (treated group) or 0.9% saline water (control group) by gavage in alternated days from gestational day 5 (GD5) until parturition and analyzes were proceed in the cerebellum of 30-day-old pups. We found increased lipid peroxidation and protein carbonylation levels as well as decreased SH content in pups prenatally exposed to 2 mg/kg MeHg. In addition, misregulated SOD/catalase activities supported imbalanced redox equilibrium. We found decreased GSK3β(Ser9) phosphorylation, suggesting activation of this enzyme and dephosphorylation/inhibition of ERK1/2 and JNK pathways. Increased PKAα catalytic subunit could be upstream of hyperphosphorylated c-Raf(Ser259) and downregulated MAPK pathway. In addition, we found raised levels of the Ca2+-dependent protein phosphatase 2 B (PP2B). We also found preserved immunohistochemical staining for both glial fibrillary acidic protein (GFAP) and NeuN in MeHg-exposed pups. Western blot analysis showed unaltered levels of BAX/BCL-XL, BAD/BCL-2 and active caspase 3. Together, these findings support absence of reactive astrocytes, neuronal damage and apoptotic cell death in the cerebellum of MeHg treated pups. The present study provides evidence that prenatal exposure to MeHg leads to later redox imbalance and disrupted signaling mechanisms in the cerebellum of 30-day-old pups potentially predisposing them to long-lasting neurological impairments in CNS.
Collapse
|
25
|
Fujimura M, Usuki F. In situ different antioxidative systems contribute to the site-specific methylmercury neurotoxicity in mice. Toxicology 2017; 392:55-63. [PMID: 29030019 DOI: 10.1016/j.tox.2017.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/28/2017] [Accepted: 10/06/2017] [Indexed: 01/17/2023]
Abstract
Methylmercury (MeHg), an environmental toxicant, induces site-specific neurotoxicity in adult human and animal models. In this study, we demonstrated that MeHg-induced neuropathological changes of the brain in mice were remarkable in the cerebrocortical neurons of deeper layers (dl-CCNs), but not in the CCNs of shallow layers (sl-CCNs) and the hippocampal neurons of cornu ammonis 1 (CA1-HNs). Total mercury concentration was not corresponded to the pathological changes. Here, we investigated the cause of such site-specific MeHg neurotoxicity with a focus on in situ antioxidative systems due to its critical role in MeHg intoxication. We performed in situ analyses of antioxidative enzymes expression using RT-qPCR analyses from laser microdissected sl-CCNs, dl-CCNs, and CA1-HNs samples, and immunohistochemistry. The results of antioxidative enzymes expression analyses demonstrated the lowest basal expression levels of mRNA and proteins, especially manganese superoxide dismutase (Mn-SOD) and glutathione peroxidase 1 (GPx1) in dl-CCNs. In addition, the Mn-SOD expression showed a lowest response to MeHg in dl-CCNs. We also performed enzymatic activity analyses for antioxidative enzymes using separated cerebral cortex and hippocampus. The results of enzymatic activity analyses indicate that the expression levels of antioxidative enzymes reflect their enzymatic activities. Immunostaining of thymidine glycerol, a sensitive oxidative stress marker, showed selectively increased expression in dl-CCNs after the exposure to MeHg but not in sl-CCNs and CA1-HNs, suggesting the occurrence of MeHg-induced oxidative stress in dl-CCNs. The differences in MeHg-induced occurrence of oxidative stress and pathological changes in sl-CCNs, dl-CCNs, and CA1-HNs corresponded to the basal level of Mn-SOD and GPx1 expression and the different protective response of Mn-SOD expression to MeHg. These findings suggest that the in situ different antioxidative systems play a role in the site-specific neurotoxicity of MeHg.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto, Japan.
| | - Fusako Usuki
- Department of Clinical Medicine, National Institute for Minamata Disease, Kumamoto, Japan
| |
Collapse
|
26
|
Site-specific neural hyperactivity via the activation of MAPK and PKA/CREB pathways triggers neuronal degeneration in methylmercury-intoxicated mice. Toxicol Lett 2017; 271:66-73. [DOI: 10.1016/j.toxlet.2017.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/17/2017] [Accepted: 03/02/2017] [Indexed: 11/20/2022]
|
27
|
Sahin D, Erdolu CO, Karadenizli S, Kara A, Bayrak G, Beyaz S, Demir B, Ates N. Effects of gestational and lactational exposure to low dose mercury chloride (HgCl2) on behaviour, learning and hearing thresholds in WAG/Rij rats. EXCLI JOURNAL 2016; 15:391-402. [PMID: 27540351 PMCID: PMC4983802 DOI: 10.17179/excli2016-315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/05/2016] [Indexed: 01/18/2023]
Abstract
We investigated the effects of inorganic mercury exposure during gestational/lactational periods on the behaviour, learning and hearing functions in a total of 32, 5-week-old and 5-month-old WAG/Rij rats (equally divided into 4 groups as 5-week and 5-month control mercury exposure groups). We evaluated the rats in terms of locomotor activity (LA), the Morris-water-maze (MWM) test and the passive avoidance (PA) test to quantify learning and memory performance; we used distortion product otoacoustic emission (DPOAE) tests to evaluate hearing ability. There were no significant differences between the 5-week-old rat groups in LA, and we detected a significant difference (p < 0.05) in the HgCl2-treated group in PA, MWM and DPOAE tests compared with the control group. The HgCl2-treated 5-week-old group exhibited worse emotional memory performance in PA, worse spatial learning and memory performances in MWM. There were no significant differences between the groups of 5-month-old rats in LA, MWM or PA. However, the DPOAE tests worsened in the mid- and high-frequency hearing thresholds. The HgCl2-treated 5-month-old group exhibited the most hearing loss of all groups. Our results convey that mercury exposure in young rats may worsen learning and memory performances as well as hearing at high-frequency levels. While there was no statistically significant difference in the behavior and learning tests in adult rats, the DPOAE test produced poorer results. Early detection of effects of mercury exposure provides medicals team with an opportunity to determinate treatment regimens and mitigate ototoxicity. DPOAE test can be used in clinical and experimental research investigating heavy metal ototoxicity.
Collapse
Affiliation(s)
- Deniz Sahin
- Kocaeli University / Medical Faculty, Physiology, Kocaeli, Turkey
| | | | | | - Ahmet Kara
- Sakarya University Training and Research Hospital, Otorhinolaryngology Department, Sakarya,Turkey
| | - Gunce Bayrak
- Kocaeli University / Medical Faculty, Kocaeli, Turkey
| | - Sumeyye Beyaz
- Kocaeli University / Medical Faculty, Kocaeli, Turkey
| | - Buse Demir
- Kocaeli University / Medical Faculty, Kocaeli, Turkey
| | - Nurbay Ates
- Kocaeli University / Medical Faculty, Physiology, Kocaeli, Turkey
| |
Collapse
|
28
|
Prenatal low-dose methylmercury exposure impairs neurite outgrowth and synaptic protein expression and suppresses TrkA pathway activity and eEF1A1 expression in the rat cerebellum. Toxicol Appl Pharmacol 2016; 298:1-8. [DOI: 10.1016/j.taap.2016.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 11/19/2022]
|
29
|
Umemori J, Winkel F, Castrén E, Karpova NN. Distinct effects of perinatal exposure to fluoxetine or methylmercury on parvalbumin and perineuronal nets, the markers of critical periods in brain development. Int J Dev Neurosci 2015; 44:55-64. [PMID: 25997908 DOI: 10.1016/j.ijdevneu.2015.05.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 12/14/2022] Open
Abstract
The in utero exposure to common chemical stressors, environmental pollutant methylmercury and antidepressant fluoxetine, results in behavioral impairments persistent into adulthood. Modulation of critical periods in brain development may alter proper network formation and lastingly impair brain function. To investigate whether early-life stressors can modulate critical periods, we analyzed the development of parvalbumin (PV) and perineuronal nets (PNNs) in the dentate gyrus and CA1 area of the hippocampus and the basolateral amygdala in mice perinatally exposed to either fluoxetine or methylmercury. The number of PV and PNN neurons, and PV intensity, were analyzed by fluorescent immunohistochemistry at the postnatal ages P17 (ongoing critical period) and P24 (closing critical period). The exposure to fluoxetine did not affect the number of PV cells and PV intensity but decreased PNN formation around the cells at P17 and P24 in all tissues. In contrast, perinatal methylmercury inhibited the development of PV interneurons and PV expression at P17 only, but at P24 these parameters were restored. Methylmercury strongly increased PNN formation from P17 to P24 in the amygdala only. We suggest that perinatal fluoxetine and methylmercury might delay the closure and the onset, respectively, of the critical periods in the amygdala and hippocampus.
Collapse
Affiliation(s)
- Juzoh Umemori
- Neuroscience Center, University of Helsinki, 00790 Helsinki, Finland
| | - Frederike Winkel
- Neuroscience Center, University of Helsinki, 00790 Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center, University of Helsinki, 00790 Helsinki, Finland
| | - Nina N Karpova
- Neuroscience Center, University of Helsinki, 00790 Helsinki, Finland.
| |
Collapse
|
30
|
Shao Y, Yamamoto M, Figeys D, Ning Z, Chan HM. Proteomic Analysis of Cerebellum in Common Marmoset Exposed to Methylmercury. Toxicol Sci 2015; 146:43-51. [DOI: 10.1093/toxsci/kfv069] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
31
|
Zhao W, Cheng J, Gu J, Liu Y, Fujimura M, Wang W. Assessment of neurotoxic effects and brain region distribution in rat offspring prenatally co-exposed to low doses of BDE-99 and methylmercury. CHEMOSPHERE 2014; 112:170-6. [PMID: 25048903 DOI: 10.1016/j.chemosphere.2014.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 05/17/2023]
Abstract
Exposure to polybrominated diphenyl ether (PDBE) and methylmercury (MeHg) can occur simultaneously as both contaminants are found in the same food sources, especially fish, seafood, marine mammals and milk. The aim of this study was to assess the effects of exposure to low levels of MeHg (2.0 μg mL(-1) in drinking water) and BDE-99 (0.2 mg kg(-1) d(-1)) from gestational day 6 to postnatal day (PND) 21, alone and in combination, on neurobehavioral development and redox responses in offspring. The present study demonstrated an interaction due to co-exposure with low doses of MeHg and BDE-99 enhanced developmental neurotoxic effects. These effects were manifested as the delayed appearance of negative geotaxis reflexes, impaired motor coordination, and induction of oxidative stress in the cerebellum. In particular, the cerebellum may be a sensitive target for combined MeHg and BDE-99 toxicity. The neurotoxicity of low dose MeHg was exacerbated by the presence of low dose of BDE-99. It is concluded that prenatal co-exposure to MeHg and BDE-99 causes oxidative stress in the cerebellum of offspring by altering the activity of different antioxidant enzymes and producing free radicals. Hg retention was not affected by co-exposure to BDE-99. However, MeHg co-exposure seemed to increase BDE-99 concentrations in selected brain regions in pups compared to pups exposed to BDE-99 only. These results showed that the adverse effects following prenatal co-exposure to MeHg and BDE-99 were associated with tissue concentrations very close to the current human body burden of this persistent bioaccumulative compound.
Collapse
Affiliation(s)
- Wenchang Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; School of Environmental and Safety Engineering, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Jinping Cheng
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jinmin Gu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuanyuan Liu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Masatake Fujimura
- Department of Basic Medical Science, National Institute for Minamata Disease, Minamata, Kumamoto 867-0008, Japan
| | - Wenhua Wang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
32
|
Bisen-Hersh EB, Farina M, Barbosa F, Rocha JBT, Aschner M. Behavioral effects of developmental methylmercury drinking water exposure in rodents. J Trace Elem Med Biol 2014; 28:117-124. [PMID: 24210169 PMCID: PMC3979511 DOI: 10.1016/j.jtemb.2013.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/19/2013] [Accepted: 09/19/2013] [Indexed: 10/26/2022]
Abstract
Early methylmercury (MeHg) exposure can have long-lasting consequences likely arising from impaired developmental processes, the outcome of which has been exposed in several longitudinal studies of affected populations. Given the large number of newborns at an increased risk of learning disabilities associated with in utero MeHg exposure, it is important to study neurobehavioral alterations using ecologically valid and physiologically relevant models. This review highlights the benefits of using the MeHg drinking water exposure paradigm and outlines behavioral outcomes arising from this procedure in rodents. Combination treatments that exacerbate or ameliorate MeHg-induced effects, and possible molecular mechanisms underlying behavioral impairment are also discussed.
Collapse
Affiliation(s)
- Emily B Bisen-Hersh
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Fernando Barbosa
- Department of Clinical, Toxicological and Bromatological Analyses, Faculty of Pharmaceutical Sciences of Ribeirão Preto, São Paulo, Brazil
| | - Joao B T Rocha
- Departamento de Química, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Michael Aschner
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, The Kennedy Center for Research on Human Development, and The Center for Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
33
|
Zimmermann LT, dos Santos DB, Colle D, dos Santos AA, Hort MA, Garcia SC, Bressan LP, Bohrer D, Farina M. Methionine stimulates motor impairment and cerebellar mercury deposition in methylmercury-exposed mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2014; 77:46-56. [PMID: 24555646 DOI: 10.1080/15287394.2014.865582] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Methylmercury (MeHg) is a highly toxic environmental contaminant that produces neurological and developmental impairments in animals and humans. Although its neurotoxic properties have been widely reported, the molecular mechanisms by which MeHg enters the cells and exerts toxicity are not yet completely understood. Taking into account that MeHg is found mostly bound to sulfhydryl-containing molecules such as cysteine in the environment and based on the fact that the MeHg-cysteine complex (MeHg-S-Cys) can be transported via the L-type neutral amino acid carrier transport (LAT) system, the potential beneficial effects of L-methionine (L-Met, a well known LAT substrate) against MeHg (administrated as MeHg-S-Cys)-induced neurotoxicity in mice were investigated. Mice were exposed to MeHg (daily subcutaneous injections of MeHg-S-Cys, 10 mg Hg/kg) and/or L-Met (daily intraperitoneal injections, 250 mg/kg) for 10 consecutive days. After treatments, the measured hallmarks of toxicity were mostly based on behavioral parameters related to motor performance, as well as biochemical parameters related to the cerebellar antioxidant glutathione (GSH) system. MeHg significantly decreased motor activity (open-field test) and impaired motor performance (rota-rod task) compared with controls, as well as producing disturbances in the cerebellar antioxidant GSH system. Interestingly, L-Met administration did not protect against MeHg-induced behavioral and cerebellar changes, but rather increased motor impairments in animals exposed to MeHg. In agreement with this observation, cerebellar levels of mercury (Hg) were higher in animals exposed to MeHg plus L-Met compared to those only exposed to MeHg. However, this event was not observed in kidney and liver. These results are the first to demonstrate that L-Met enhances cerebellar deposition of Hg in mice exposed to MeHg and that this higher deposition may be responsible for the greater motor impairment observed in mice simultaneously exposed to MeHg and L-Met.
Collapse
Affiliation(s)
- Luciana T Zimmermann
- a Departamento de Bioquímica, Centro de Ciências Biológicas , Universidade Federal de Santa Catarina , Florianópolis , Santa Catarina , Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Patel E, Reynolds M. Methylmercury impairs motor function in early development and induces oxidative stress in cerebellar granule cells. Toxicol Lett 2013; 222:265-72. [DOI: 10.1016/j.toxlet.2013.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/31/2013] [Accepted: 08/03/2013] [Indexed: 01/20/2023]
|
35
|
Mello-Carpes PB, Barros W, Borges S, Alves N, Rizzetti D, Peçanha FM, Vassallo DV, Wiggers GA, Izquierdo I. Chronic exposure to low mercury chloride concentration induces object recognition and aversive memories deficits in rats. Int J Dev Neurosci 2013; 31:468-72. [PMID: 23770019 DOI: 10.1016/j.ijdevneu.2013.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 10/26/2022] Open
Abstract
This work examines the effects of chronic exposure to low inorganic mercury (mercury chloride, HgCl(2)) concentration on the recognition and aversive memories. Forty male Wistar rats were divided into 4 groups treated during 30 or 60 days with saline (control) or HgCl(2) doses. After treated the animals were tested considering object recognition and inhibitory avoidance behavioral memory paradigms. Elevated plus maze, open field and tail flick tests were used to assess anxiety, locomotor and exploratory activity and pain thresholds. Only exposure for 60 days to HgCl(2) induced in memory deficits quantified in the object recognition task. In the inhibitory avoidance all the animals exposed to mercury (for 30 or 60 days) presented worst performance than control animals. Our results suggest that chronic exposure to low mercury chloride concentrations impairs memory formation.
Collapse
|
36
|
Cheng J, Fujimura M, Zhao W, Wang W. Neurobehavioral effects, c-Fos/Jun expression and tissue distribution in rat offspring prenatally co-exposed to MeHg and PFOA: PFOA impairs Hg retention. CHEMOSPHERE 2013; 91:758-764. [PMID: 23490179 DOI: 10.1016/j.chemosphere.2013.02.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 02/02/2013] [Accepted: 02/09/2013] [Indexed: 06/01/2023]
Abstract
Exposure to methylmercury (MeHg) and perfluorooctanoic acid (PFOA) can occur simultaneously as both contaminants are found in the same food sources, especially fish, seafood, marine mammals and milk. The aim of this study was to assess the effects of exposure to MeHg (10 μg mL(-1) in drinking water) and PFOA (10 μg mL(-1) in drinking water) from gestational day 1 to postnatal day (PND) 21, alone and in combination, on neurobehavioral development and the expression of c-Fos/Jun in different brain regions in the offspring. Our findings showed that exposure to MeHg alone, and exposure to MeHg combined with PFOA significantly induced cliff avoidance reflexes and negative geotaxis reflexes. And these effects appeared to be greater following exposure to MeHg alone. MeHg and/or PFOA exposure did not significantly impair motor coordination functions, or cause significant changes in c-Fos expression in the hippocampus and cerebellum, and spatial learning tests were similar to those in the controls, thus it was impossible to determine whether combined exposure to MeHg and PFOA had any additional effects on both hippocampus and cerebellum regions. However, a significant increase in the frequency of line crossing was observed in rats treated with MeHg or PFOA alone, and there were no significant differences between the MeHg+PFOA-treated group and the controls, suggesting that PFOA was antagonistic to MeHg toxicity in the locomotor activity test. Co-exposure to MeHg and PFOA decreased all tissue Hg concentrations in pups compared to the group exposed to MeHg only, suggesting that PFOA impaired Hg retention in different tissues.
Collapse
Affiliation(s)
- Jinping Cheng
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | | | | | | |
Collapse
|