Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Article En | MEDLINE | ID: mdl-38825526

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.


Disease Models, Animal , Gabapentin , Neuralgia , Animals , Gabapentin/pharmacology , Gabapentin/therapeutic use , Neuralgia/drug therapy , Rats , Male , Methylmercury Compounds , Analgesics/pharmacology , Analgesics/therapeutic use , Amines/pharmacology , Amines/therapeutic use , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , gamma-Aminobutyric Acid/pharmacology , Rats, Wistar
2.
Toxicology ; 505: 153833, 2024 Jun.
Article En | MEDLINE | ID: mdl-38759721

Electronic waste (e-waste) contains numerous metals and organic pollutants that have detrimental impacts on human health. We studied 199 e-waste recycling workers and 104 non-exposed workers; analyzed blood, urine, and hair samples to measure heavy metals, hormonal, liver, and renal function. We used quantile regression models to evaluate the impact of Pb, Cd, and Hg on hormonal, liver and renal function, and the role of DNA oxidative damage in mediating the relationship between exposures and outcomes. Exposed workers had higher blood lead (Pb) (median 11.89 vs 3.63 µg/dL), similar blood cadmium (Cd) (1.04 vs 0.99 µg/L) and lower total mercury (Hg) in hair (0.38 vs 0.57 ppm) than non-exposed group. Exposed workers also had elevated median concentrations of total triiodothyronine (TT3), aspartate aminotransferase (AST), alanine aminotransferase (ALT), urinary albumin, albumin creatinine ratio (ACR) and estimated glomerular filtration rate (eGFR) were significantly higher than non-exposed group (p≤0.05). Sex hormones including luteinizing hormone, follicle stimulating hormone, estrogen, progesterone and testosterone concentrations were not significantly different between exposed and non-exposed (all p≥0.05). The median concentration of ALT was 4.00 (95% CI: 0.23, 7.77), urinary albumin was 0.09 (95% CI: 0.06, 0.12) and ACR was 1.31 (95% CI: 0.57, 2.05) units higher in the exposed group compared to non-exposed group. Pb was associated with a 3.67 unit increase in the ALP (95% CI: 1.53, 5.80), 0.01 unit increase in urinary albumin (95% CI: 0.002, 0.01), and 0.07 unit increase in ACR (95% CI: 0.01, 0.13). However, no hormonal, renal, and hepatic parameters were associated with Cd or Hg. Oxidative DNA damage did not mediate exposure-outcome relationships (p≥0.05). Our data indicate e-waste exposure impairs liver and renal functions secondary to elevated Pb levels. Continuous monitoring, longitudinal studies to evaluate the dose-response relationship and effective control measure are required to protect workers from e-waste exposure.


Electronic Waste , Kidney , Occupational Exposure , Humans , Electronic Waste/adverse effects , Adult , Male , Occupational Exposure/adverse effects , Occupational Exposure/analysis , Female , Bangladesh/epidemiology , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Liver/drug effects , Liver/metabolism , Middle Aged , Metals, Heavy/urine , Metals, Heavy/blood , Metals, Heavy/toxicity , Recycling , Young Adult , Cadmium/blood , Cadmium/urine , Cadmium/toxicity , Hair/chemistry , Lead/blood , Lead/toxicity , Hormones/blood , DNA Damage
3.
Int J Hyg Environ Health ; 257: 114340, 2024 Apr.
Article En | MEDLINE | ID: mdl-38422602

BACKGROUND: Electronic waste (e-waste) recycling activities release toxic metals, which pose substantial hazard to the environment and human health. We evaluated metal concentrations in biological and environmental samples, and examined the associations between biological lead (Pb), cadmium (Cd), and mercury (Hg) with soil and dust metals, and other possible determinants, among populations exposed and non-exposed to e-waste in Bangladesh. METHODS: A total of 199 e-waste workers and 104 non-exposed individuals were recruited. We measured blood Pb (BPb) and Cd (BCd) concentrations and total Hg (THg) from hair samples. Data were collected on occupational, and behavioral factors. We fitted an elastic net regression (ENET) to model the relationship between a set of influencing factors and metals as outcome variables while controlling for potential covariates. RESULTS: The median concentrations of BPb (11.89 µg/dL) and BCd (1.04 µg/L) among exposed workers were higher than those of non-exposed workers (BPb: 3.63 µg/dL and BCd: 0.83 µg/L respectively). A 100 ppm increment in soil Pb level was associated with an increase in ln-Pb (transformed) in blood (ß = 0.002; 95% CI = 0.00, 0.02). Similarly, ln-BCd level increased (ß = 0.02; 95% CI = 0.001, 0.07) with every ppm increase in dust Cd level. The number of years worked in e-waste activities was associated with elevated ln-BPb (ß = 0.01; 95% CI = 0.01, 0.02) and ln-BCd levels (ß = 0.003; 95% CI = 0.00, 0.05). Smoking significantly contributed to elevated levels of ln-BCd (ß = 0.46; 95% CI = 0.43, 0.73). An increment of 100 kg of e-waste handling per week led to an increase in ln-BPb levels (ß = 0.002; 95% CI = 0.00, 0.01), while respondents knowledge about adverse impact on e-waste reduced the ln-BPb level (ß = -0.14; 95% CI = -0.31, -0.03). Fish consumption frequency had a positive association with THg in hair. CONCLUSIONS: Our data show the need for workplace controls to reduce exposure to Pb and Cd with a broader view of exposure source taken.


Electronic Waste , Mercury , Humans , Cadmium , Lead , Dust/analysis , Bangladesh , Recycling , Hair/chemistry
4.
Arch Toxicol ; 98(4): 1163-1175, 2024 Apr.
Article En | MEDLINE | ID: mdl-38367039

Methylmercury (MeHg) is a well-known environmental neurotoxicant that causes severe brain disorders such as Minamata disease. Although some patients with Minamata disease develop olfactory dysfunction, the underlying pathomechanism is largely unknown. We examined the effects of MeHg on the olfactory system using a model of MeHg poisoning in which mice were administered 30 ppm MeHg in drinking water for 8 weeks. Mice exposed to MeHg displayed significant mercury accumulation in the olfactory pathway, including the nasal mucosa, olfactory bulb, and olfactory cortex. The olfactory epithelium was partially atrophied, and olfactory sensory neurons were diminished. The olfactory bulb exhibited an increase in apoptotic cells, hypertrophic astrocytes, and amoeboid microglia, mainly in the granular cell layer. Neuronal cell death was observed in the olfactory cortex, particularly in the ventral tenia tecta. Neuronal cell death was also remarkable in higher-order areas such as the orbitofrontal cortex. Correlation analysis showed that neuronal loss in the olfactory cortex was strongly correlated with the plasma mercury concentration. Our results indicate that MeHg is an olfactory toxicant that damages the central regions involved in odor perception. The model described herein is useful for analyzing the mechanisms and treatments of olfactory dysfunction in MeHg-intoxicated patients.


Mercury Poisoning, Nervous System , Mercury , Methylmercury Compounds , Olfaction Disorders , Humans , Mice , Animals , Methylmercury Compounds/toxicity , Microglia/pathology , Olfaction Disorders/chemically induced , Olfaction Disorders/complications
5.
Environ Toxicol ; 39(5): 3149-3159, 2024 May.
Article En | MEDLINE | ID: mdl-38323385

Methylmercury (MeHg) causes selective neuronal damage to cerebrocortical neurons (CCNs) in the central nervous system, but not to hippocampal neurons (HiNs), which are highly vulnerable to neurodegenerative diseases. In our previous study using cultured rat neurons, we performed a comprehensive gene expression analysis and found that the brain-derived neurotrophic factor (BDNF), a neurotrophin (NT), was specifically expressed in HiNs. Therefore, to elucidate the causal factors of MeHg toxicity resistance in HiNs, we conducted a comparative study of the protein expression and function of several NTs, including BDNF, using CCNs showing vulnerability to MeHg toxicity and HiNs showing resistance. BDNF was specifically expressed in HiNs, whereas nerve growth factor was barely detectable in either neuron type. In addition, other NTs, NT3 and NT4/5, were expressed in small but nearly equal amounts in both neuron types. Furthermore, among the various pathways involved in MeHg neurotoxicity, the p44/42 MAPK pathway was specifically activated in HiNs, even without MeHg treatment. siRNAs were used to reduce NTs in both neuron types. Only a specific reduction in BDNF attenuated the resistance to MeHg toxicity and p44/42 MAPK activation in HiNs. In addition, the external addition of BDNF and NT4/5, which act on the same tyrosine receptor kinase (Trk), TrkB, suppressed MeHg neurotoxicity in both neuron types. These results suggest that BDNF, expressed specifically in HiNs, is involved in the resistance to MeHg neurotoxicity via TrkB. Additionally, the activation of the p44/42 MAPK pathway may contribute to the inhibitory effect of BDNF on MeHg neurotoxicity.


Methylmercury Compounds , Neurotoxicity Syndromes , Rats , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Methylmercury Compounds/toxicity , Neurons , Neurotoxicity Syndromes/metabolism , Hippocampus/metabolism
6.
J Toxicol Sci ; 49(2): 55-60, 2024.
Article En | MEDLINE | ID: mdl-38296529

Apoptosis is one of the hallmarks of MeHg-induced neuronal cell death; however, its molecular mechanism remains unclear. We previously reported that MeHg exposure induces neuron-specific ER stress in the mouse brain. Excessive ER stress contributes to apoptosis, and CHOP induction is considered to be one of the major mechanisms. CHOP is also increased by MeHg exposure in the mouse brain, suggesting that it correlates with increased apoptosis. In this study, to clarify whether CHOP mediates MeHg-induced apoptosis, we examined the effect of CHOP deletion on MeHg exposure in CHOP-knockout mice. Our data showed that CHOP deletion had no effect on MeHg exposure-induced weight loss or hindlimb impairment in mice, nor did it increase apoptosis or inhibit neuronal cell loss. Hence, CHOP plays little role in MeHg toxicity, and other apoptotic pathways coupled with ER stress may be involved in MeHg-induced cell death.


Brain , Methylmercury Compounds , Neurons , Animals , Mice , Apoptosis , Brain/pathology , Methylmercury Compounds/toxicity , Methylmercury Compounds/metabolism , Mice, Knockout , Neurons/pathology
7.
Ambio ; 52(5): 877-896, 2023 May.
Article En | MEDLINE | ID: mdl-36790578

Mercury (Hg) is a chemical of health concern worldwide that is now being acted upon through the Minamata Convention. Operationalizing the Convention and tracking its effectiveness requires empathy of the diversity and variation of mercury exposure and risk in populations worldwide. As part of the health plenary for the 15th International Conference on Mercury as a Global Pollutant (ICMGP), this review paper details how scientific understandings have evolved over time, from tragic poisoning events in the mid-twentieth century to important epidemiological studies in the late-twentieth century in the Seychelles and Faroe Islands, the Arctic and Amazon. Entering the twenty-first century, studies on diverse source-exposure scenarios (e.g., ASGM, amalgams, contaminated sites, cosmetics, electronic waste) from across global regions have expanded understandings and exemplified the need to consider socio-environmental variables and local contexts when conducting health studies. We conclude with perspectives on next steps for mercury health research in the post-Minamata Convention era.


Environmental Exposure , Environmental Pollutants , Mercury , Humans , Arctic Regions , Denmark , Environmental Pollutants/toxicity , Environmental Pollutants/analysis , Mercury/toxicity , Mercury/analysis
8.
Int J Mol Sci ; 23(23)2022 Dec 06.
Article En | MEDLINE | ID: mdl-36499738

Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures specific areas of the brain. MeHg is known to induce oxidative and endoplasmic reticulum (ER) stress. The unfolded protein response (UPR) pathway has a dual nature in that it regulates and protects cells from an overload of improperly folded proteins in the ER, whereas excessively stressed cells are eliminated by apoptosis. Oxidative stress/ER stress induced by methylmercury exposure may tilt the UPR toward apoptosis, but there is little in vivo evidence of a direct link to actual neuronal cell death. Here, by using the ER stress-activated indicator (ERAI) system, we investigated the time course signaling alterations of UPR in vivo in the most affected areas, the somatosensory cortex and striatum. In the ERAI-Venus transgenic mice exposed to MeHg (30 or 50 ppm in drinking water), the ERAI signal, which indicates the activation of the cytoprotective pathway of the UPR, was only transiently enhanced, whereas the apoptotic pathway of the UPR was persistently enhanced. Furthermore, detailed analysis following the time course showed that MeHg-induced apoptosis is strongly associated with alterations in UPR signaling. Our results suggest that UPR modulation could be a therapeutic target for treating neuropathy.


Methylmercury Compounds , Unfolded Protein Response , Mice , Animals , Endoplasmic Reticulum Stress , Cell Death , Signal Transduction , Apoptosis , Methylmercury Compounds/toxicity , Mice, Transgenic , Brain
9.
Toxicol Lett ; 371: 38-45, 2022 Dec 01.
Article En | MEDLINE | ID: mdl-36244566

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.

10.
JMIR Res Protoc ; 11(8): e38201, 2022 Aug 16.
Article En | MEDLINE | ID: mdl-35972788

BACKGROUND: e-Waste is a rapidly growing waste stream worldwide, and Bangladesh is a hub of e-waste handling. Informal e-waste recycling operations involve crude methods for dismantling, repairing, sorting, and recycling electronic goods with bare hands and without personal health protections. Direct inhalation or dermal exposure to toxicants during informal recycling is common. Evidence suggests that e-waste-derived toxicants pollute the terrestrial ecosystem and have been linked with adverse health effects. However, e-waste recycling-related occupational health hazards have not been adequately explored in the context of Bangladesh. OBJECTIVE: Our study aims to expand the current understanding of exposure to e-waste. This study will measure the metal concentrations in biological and environmental samples and evaluate the relationship between heavy metals and the biochemical systems of the e-waste workers. METHODS: The study uses a cross-sectional study design consisting of an exposed site and a nonexposed control site. The trained team collected information on individual exposures, detailed work and medical history, and biological samples (blood, urine, and hair) from each subject. This study will measure heavy metal levels (lead, cadmium, and mercury) and biochemical parameters (hematological, hormonal, renal, and others) from the biological samples with reported physical function as outcomes of interest. In addition, we also collected soil and dust samples from both exposed and nonexposed control sites to measure the health risk. All the environmental samples will be analyzed using inductively coupled plasma mass spectrometer to determine metal concentrations. We will also conduct a qualitative investigation for a deeper understanding of the e-waste management system in Bangladesh. RESULTS: The protocol has been approved by the Institutional Review Boards of the International Centre for Diarrheal Disease Research, Bangladesh, and The University of Queensland's Human Behavioral Ethics Committee. Informed written consent was obtained from all participants. We recruited 199 workers from the e-waste sites with at least 5 years of exposure and 104 control subjects with no industrial or e-waste exposure. Sample analysis is estimated to be completed in 2022. CONCLUSIONS: Although many studies have identified potential adverse health outcomes from exposure to e-waste, there is a lack of published epidemiological research in Bangladesh. Research in this field is particularly pressing in the context of the current e-waste trend and the need to deepen the understanding of exposures and outcomes. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/38201.

11.
Int J Mol Sci ; 23(13)2022 Jun 29.
Article En | MEDLINE | ID: mdl-35806222

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.


Mercury , Methylmercury Compounds , Neurotoxicity Syndromes , Adult , Child , Humans , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Oxidative Stress
12.
J Toxicol Sci ; 47(5): 211-219, 2022.
Article En | MEDLINE | ID: mdl-35527009

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.


Methylmercury Compounds , Animals , Antioxidants/pharmacology , Cells, Cultured , Gene Expression Profiling , Hippocampus , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Neurons , Rats
13.
J Toxicol Sci ; 47(1): 31-37, 2022.
Article En | MEDLINE | ID: mdl-34987139

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.


Methylmercury Compounds , Animals , Brain , Cerebellum , Methylmercury Compounds/toxicity , Rats , Sulfides , Sulfur
14.
Biol Pharm Bull ; 44(4): 522-527, 2021.
Article En | MEDLINE | ID: mdl-33790104

Methylmercury (MeHg) exposure during pregnancy is a concern because of its potential health risks to fetuses. Intestinal microbiota has important roles in the decomposition and fecal excretion of MeHg. We investigated the effect of nondigestible saccharides on the accumulation and excretion of Hg after MeHg exposure. Female BALB/cByJ mice were fed a basal diet or the same diet supplemented with 5% fructooligosaccharides (FOS) or 2.5% glucomannan. Six weeks after feeding, mice were administered MeHg chloride (4 mg Hg/kg, per os (p.o.)), and urine and feces were collected for 28 d. FOS-fed mice had lower total Hg levels in all tissues (including the brain) compared with that of controls. The glucomannan diet had no effect on tissue Hg levels. No differences in tissue concentrations of inorganic Hg among groups were found. Fecal Hg excretion was markedly higher in FOS-fed mice than that in controls, but urinary Hg excretion was similar. FOS-fed mice had a higher proportion of inorganic Hg in feces than that of controls, with a significant increase in fecal Hg excretion. Analysis of fecal bacterial population showed the relative abundance of Bacteroides in FOS-fed mice to be higher than that in controls. The results suggest that FOS enhanced fecal Hg excretion and decreased tissue Hg levels after MeHg administration, possibly by accelerating MeHg demethylation by intestinal bacteria (the candidate genus Bacteroides). This demethylation also reduces MeHg absorption in the large intestine. In conclusion, daily FOS intake may decrease tissue Hg levels in animals and humans exposed to MeHg.


Brain/metabolism , Dietary Supplements , Mercury/metabolism , Methylmercury Compounds/pharmacokinetics , Oligosaccharides/pharmacology , Animals , Bacteria/genetics , Bacteria/isolation & purification , Feces/chemistry , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Kidney/metabolism , Liver/metabolism , Mannans/pharmacology , Methylmercury Compounds/blood , Mice, Inbred BALB C , RNA, Ribosomal, 16S
15.
Arch Toxicol ; 95(6): 2151-2162, 2021 06.
Article En | MEDLINE | ID: mdl-33847776

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.


Hyperalgesia/chemically induced , Inflammation/chemically induced , Methylmercury Compounds/toxicity , Animals , Cytokines/metabolism , Hyperalgesia/physiopathology , Inflammation/pathology , Male , Microglia/drug effects , Microglia/pathology , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/pathology
16.
Arch Toxicol ; 95(4): 1227-1239, 2021 04.
Article En | MEDLINE | ID: mdl-33454822

Methylmercury (MeHg) is a chemical substance that causes adverse effects on fetal development. However, the molecular mechanisms by which environmental MeHg affects fetal development have not been clarified. Recently, it has been suggested that the toxic effects of chemicals on fetal development are related alterations in epigenetics, such as DNA methylation and histone modification. In order to analyze the epigenetic effects of low-level MeHg exposure on neuronal development, we evaluated neuronal development both in vivo and in vitro. Pregnant mice (C57BL/6J) were orally administrated 3 mg/kg of MeHg once daily from embryonic day 12-14. Fetuses were removed on embryonic day 19 and brain tissues were collected. LUHMES cells were treated with 1 nM of MeHg for 6 days and collected on the last day of treatment. In both in vivo and in vitro samples, MeHg significantly suppressed neurite outgrowth. Decreased acetylated histone H3 (AcH3) levels and increased histone deacetylase (HDAC) 3 and HDAC6 levels were observed in response to MeHg treatment in both in vivo and in vitro experiments. In addition, increased DNA methylation and DNA methyltransferase 1 (DNMT1) levels were observed in both in vivo and in vitro experiments. The inhibition of neurite outgrowth resulting from MeHg exposure was restored by co-treatment with DNMT inhibitor or HDAC inhibitors. Our results suggest that neurological effects such as reduced neurite outgrowth due to low-level MeHg exposure result from epigenetic changes, including a decrease in AcH3 via increased HDAC levels and an increase in DNA methylation via increased DNMT1 levels.


Epigenesis, Genetic/drug effects , Maternal Exposure/adverse effects , Methylmercury Compounds/toxicity , Neurogenesis/drug effects , Animals , Cell Line , DNA Methylation/drug effects , DNA Modification Methylases/metabolism , Female , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Methylmercury Compounds/administration & dosage , Mice , Mice, Inbred C57BL , Neuronal Outgrowth/drug effects , Pregnancy
17.
Arch Toxicol ; 95(4): 1241-1250, 2021 04.
Article En | MEDLINE | ID: mdl-33454823

Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures a specific area of the brain. MeHg-mediated neurotoxicity is believed to be caused by oxidative stress and endoplasmic reticulum (ER) stress but the mechanism by which those stresses lead to neuronal loss is unclear. Here, by utilizing the ER stress-activated indicator (ERAI) system, we investigated the signaling alterations in the unfolded protein response (UPR) prior to neuronal apoptosis in the mouse brain. In ERAI transgenic mice exposed to MeHg (25 mg/kg, S.C.), the ERAI signal, which indicates activation of the cytoprotective pathway of the UPR, was detected in the brain. Interestingly, detailed ex vivo analysis showed that the ERAI signal was localized predominantly in neurons. Time course analysis of MeHg exposure (30 ppm in drinking water) showed that whereas the ERAI signal was gradually attenuated at the late phase after increasing at the early phase, activation of the apoptotic pathway of the UPR was enhanced in proportion to the exposure time. These results suggest that MeHg induces not only ER stress but also neuronal cell death via a UPR shift. UPR modulation could be a therapeutic target for treating neuropathy caused by electrophiles similar to MeHg.


Brain/drug effects , Endoplasmic Reticulum Stress/drug effects , Methylmercury Compounds/toxicity , Unfolded Protein Response/drug effects , Animals , Apoptosis/drug effects , Brain/pathology , Cell Death/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Spatio-Temporal Analysis , Time Factors
18.
Antioxidants (Basel) ; 9(10)2020 Oct 16.
Article En | MEDLINE | ID: mdl-33081221

Methylmercury (MeHg) is a well-known neurotoxicant that causes severe intoxication in humans. In Japan, it is referred to as Minamata disease, which involves two characteristic clinical forms: fetal type and adult type depending on the exposed age. In addition to MeHg burden level, individual susceptibility to MeHg plays a role in the manifestation of MeHg toxicity. Research progress has pointed out the importance of oxidative stress in the pathogenesis of MeHg toxicity. MeHg has a high affinity for selenohydryl groups, sulfhydryl groups, and selenides. It has been clarified that such affinity characteristics cause the impairment of antioxidant enzymes and proteins, resulting in the disruption of antioxidant systems. Furthermore, MeHg-induced intracellular selenium deficiency due to the greater affinity of MeHg for selenohydryl groups and selenides leads to failure in the recoding of a UGA codon for selenocysteine and results in the degradation of antioxidant selenoenzyme mRNA by nonsense-mediated mRNA decay. The defect of antioxidant selenoenzyme replenishment exacerbates MeHg-mediated oxidative stress. On the other hand, it has also been revealed that MeHg can directly activate the antioxidant Keap1/Nrf2 signaling pathway. This review summarizes the incidence of MeHg-mediated oxidative stress from the viewpoint of the individual intracellular redox system interactions and the MeHg-mediated aforementioned intracellular events. In addition, the mechanisms of cellular stress pathways and neuronal cell death triggered by MeHg-mediated oxidative stress and direct interactions of MeHg with reactive residues of proteins are mentioned.

19.
Food Chem Toxicol ; 146: 111810, 2020 Dec.
Article En | MEDLINE | ID: mdl-33058990

The main target organ for MeHg is the nervous system, and its neurological dysfunction remains irreversible. Therefore, predictive biomarkers associated with individual susceptibility to MeHg and future clinical severity are needed to protect against the progression of MeHg toxicity. In this study, we demonstrated that plasma thiol antioxidant capacity (-SHp) is a useful predictive biomarker associated with future clinical severity using MeHg-intoxicated rats administered 1 mg/kg/day for 4 weeks. Blood samples were collected from the subclavian vein of each rat once a week to examine total blood mercury concentrations and the levels of plasma oxidative stress markers. Time course analyses of the correlation between these weekly blood examination values and hind limb crossing signs score after 4 weeks of MeHg exposure were performed, and plasma -SHp levels after 2 weeks of MeHg exposure showed strong correlations with future hind limb crossing sign scores. Neuropathological changes also developed in parallel with hind limb crossing sign scores. Quantitative analysis of vacuolar areas in the spinal cord showed a strong correlation with hind limb crossing sign scores. In conclusion, evaluation of plasma -SHp levels allowed us to detect individuals at risk for health damage and could protect the sensitive population against MeHg toxicity.


Antioxidants/metabolism , Central Nervous System/drug effects , Methylmercury Compounds/toxicity , Sulfhydryl Compounds/blood , Animals , Biomarkers/blood , Blood Proteins/metabolism , Central Nervous System/physiopathology , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
20.
Arch Toxicol ; 94(4): 1335-1347, 2020 04.
Article En | MEDLINE | ID: mdl-32140736

Methylmercury (MeHg) is a potent neurotoxic chemical, and gestational exposure to MeHg is known to cause developmental impairments in fetuses. Although it is well established that fetuses are extremely susceptible to MeHg toxicity, limited studies have investigated the effect of low-level MeHg exposure on mothers. In this study, we demonstrated that exposure of pregnant rats to low-level MeHg (1 ppm in drinking water) induced cerebellar synaptic and neuritic remodeling during the perinatal period between gestational day 20 and postnatal day (PND) 1. MeHg-induced neurodegeneration, for example, cerebellar granule cell death, was not detected and fetuses were delivered normally and exhibited normal development. The maternal cerebellar synaptic and neuritic changes were restored by PND 21. To elucidate the mechanisms underlying these perinatal changes in MeHg-exposed pregnant rats, we investigated proteins related to synapse formation and neurite outgrowth. We identified suppression of the tropomyosin receptor kinase (Trk) A pathway and reduced activity-regulated cytoskeleton-associated protein (Arc) expression in MeHg-exposed pregnant rats during the perinatal period, mirroring the decreased expression of synaptic and neuritic proteins. MeHg-exposed pregnant rats also exhibited increased perinatal plasma corticosterone levels and decreased estradiol levels compared to vehicle-exposed pregnant rats. Similar to the synaptic and neuritic changes, TrkA pathway activity, Arc expression, and plasma hormone levels were subsequently normalized. These results suggest that exposure of pregnant rats to low-level MeHg affected perinatal cerebellar synaptic and neuritic remodeling through modulation of the TrkA pathway and Arc expression which may be caused by MeHg-induced hormonal changes.


Chromosome Pairing/drug effects , Hazardous Substances/toxicity , Methylmercury Compounds/toxicity , Neuronal Plasticity/physiology , Animals , Brain , Cell Death , Cerebellum , Female , Humans , Male , Maternal Exposure , Motor Activity , Neurites , Neurons , Pregnancy , Rats
...