Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(13)2022 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-35806222

RESUMO

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.


Assuntos
Mercúrio , Compostos de Metilmercúrio , Síndromes Neurotóxicas , Adulto , Criança , Humanos , Compostos de Metilmercúrio/metabolismo , Compostos de Metilmercúrio/toxicidade , Neurônios/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Estresse Oxidativo
2.
Arch Toxicol ; 95(6): 2151-2162, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33847776

RESUMO

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.


Assuntos
Hiperalgesia/induzido quimicamente , Inflamação/induzido quimicamente , Compostos de Metilmercúrio/toxicidade , Animais , Citocinas/metabolismo , Hiperalgesia/fisiopatologia , Inflamação/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Corno Dorsal da Medula Espinal/efeitos dos fármacos , Corno Dorsal da Medula Espinal/patologia
3.
Arch Toxicol ; 94(4): 1335-1347, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32140736

RESUMO

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.


Assuntos
Pareamento Cromossômico/efeitos dos fármacos , Substâncias Perigosas/toxicidade , Compostos de Metilmercúrio/toxicidade , Plasticidade Neuronal/fisiologia , Animais , Encéfalo , Morte Celular , Cerebelo , Feminino , Humanos , Masculino , Exposição Materna , Atividade Motora , Neuritos , Neurônios , Gravidez , Ratos
4.
Toxicol Appl Pharmacol ; 298: 1-8, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26965727

RESUMO

Methylmercury (MeHg) is a highly neurotoxic environmental chemical that can cause developmental impairments. Human fetuses and neonates are particularly susceptible to MeHg toxicity; however, the mechanisms governing its effects in the developing brain are unclear. In the present study, we investigated the effects of prenatal and lactational MeHg exposure on the developing cerebellum in rats. We demonstrated that exposure to 5ppm MeHg decreased postnatal expression of pre- and postsynaptic proteins, suggesting an impairment in synaptic development. MeHg exposure also reduced neurite outgrowth, as shown by a decrease in the expression of the neurite marker neurofilament H. These changes were not observed in rats exposed to 1ppm MeHg. In order to define the underlying mechanism, we investigated the effects of MeHg exposure on the tropomyosin receptor kinase (Trk) A pathway, which plays important roles in neuronal differentiation and synapse formation. We demonstrated suppression of the TrkA pathway on gestation day 20 in rats exposed to 5ppm MeHg. In addition, down-regulation of eukaryotic elongation factor 1A1 (eEF1A1) was observed on postnatal day 1. eEF1A1 knockdown in differentiating PC12 cells impaired neurite outgrowth and synaptic protein expression, similar to the results of MeHg exposure in the cerebellum. These results suggest that suppression of the TrkA pathway and subsequent decreases in eEF1A1 expression induced by prenatal exposure to MeHg may lead to reduced neurite outgrowth and synaptic protein expression in the developing cerebellum.


Assuntos
Cerebelo/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Compostos de Metilmercúrio/toxicidade , Neuritos/efeitos dos fármacos , Fator 1 de Elongação de Peptídeos/genética , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Receptor trkA/antagonistas & inibidores , Sinapses/efeitos dos fármacos , Animais , Western Blotting , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Cerebelo/patologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Feminino , Masculino , Neuritos/patologia , Células PC12 , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Wistar , Sinapses/genética
5.
Arch Toxicol ; 90(4): 917-26, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25975991

RESUMO

Manifestation of methylmercury (MeHg) toxicity depends on individual susceptibility to MeHg, as well as MeHg burden level. Therefore, biomarkers that reflect the protective capacity against MeHg are needed. The critical role of oxidative stress in the pathogenesis of MeHg cytotoxicity has been demonstrated. Because MeHg has high affinity for selenohydryl groups, sulfhydryl groups, and selenides, and causes posttranscriptional defects in selenoenzymes, proteins with selenohydryl and sulfhydryl groups should play a critical role in mediating MeHg-induced oxidative stress. Here, plasma oxidative stress markers and selenoproteins were investigated in MeHg-intoxicated rats showing neuropathological changes after 4 weeks of MeHg exposure. The thiol antioxidant barrier (-SHp) level significantly decreased 2 weeks after MeHg exposure, which is an early stage at which no systemic oxidative stress, histopathological changes, or clinical signs were detected. Diacron reactive oxidant metabolite (d-ROM) levels significantly increased 3 weeks after MeHg exposure, indicating the occurrence of systemic oxidative stress. Rats treated with lead acetate or cadmium chloride showed no changes in levels of -SHp and d-ROM. Selenoprotein P1 abundance significantly decreased in MeHg-treated rats, whereas it significantly increased in rats treated with Pb or Cd. Plasma selenium-dependent glutathione peroxidase (GPx3) activity also significantly decreased after MeHg exposure, whereas plasma non-selenoenzyme glutathione reductase activity significantly increased in MeHg-treated rats. The results suggest that decreased capacity of -SHp and selenoproteins (GPx3 and selenoprotein P) can be useful biomarkers of ongoing MeHg cytotoxicity and the individual protective capacity against the MeHg body burden.


Assuntos
Biomarcadores/sangue , Selenoproteínas/sangue , Animais , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Cádmio/toxicidade , Glutationa Peroxidase/metabolismo , Chumbo/toxicidade , Masculino , Compostos de Metilmercúrio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(37): 15037-42, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23983263

RESUMO

Nonsense-mediated mRNA decay (NMD) is an mRNA surveillance mechanism that eliminates aberrant mRNAs containing premature termination codons (PTCs). NMD inhibits the production of aberrant proteins that still retain, at least in part, wild-type function as well as dominant-negative peptides. Therefore, the selective inhibition of NMD has the potential to ameliorate NMD-exacerbated mutant phenotypes. However, we do not have sufficient knowledge of how to effectively suppress NMD with minimum cytotoxic effects. In this study, we aimed to identify NMD-related factors that can be targeted to efficiently inhibit NMD without causing significant cytotoxicity to restore the levels of truncated but partially functional proteins. We evaluated the knockdown of 15 NMD components in Ullrich congenital muscular dystrophy fibroblasts, which have a homozygous frameshift mutation causing a PTC in the collagen type VI α 2 gene. Of the 15 NMD factors tested, knockdown of SMG-8 produced the best effect for restoring defective mRNA and protein levels without affecting cell growth, cell-cycle progression, or endoplasmic reticulum stress. The efficacy of SMG-8 knockdown to improve the mutant phenotype was confirmed using another cell line, from a cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy patient who carries a PTC-containing mutation in HtrA serine peptidase 1. Our results suggest that SMG-8 is an appropriate target for inhibiting NMD to improve NMD-exacerbated mutant phenotypes. NMD inhibition by knockdown of SMG-8 may also be useful to induce synergy in combining the use of read-through drugs for patients with nonsense mutation-associated diseases.


Assuntos
Degradação do RNAm Mediada por Códon sem Sentido , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Quinases/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Códon sem Sentido , Colágeno Tipo VI/química , Colágeno Tipo VI/genética , DNA Complementar/genética , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Dados de Sequência Molecular , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutação , Proteínas Nucleares/genética , Fenótipo , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/genética , Proteínas Quinases/química , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Subunidades Proteicas , RNA Interferente Pequeno/genética , Esclerose/genética , Esclerose/metabolismo , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
7.
Toxicol Appl Pharmacol ; 288(1): 19-25, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26184774

RESUMO

Methylmercury (MeHg) is an environmental neurotoxicant. The developing nervous system is susceptible to low concentrations of MeHg; however, the effect of MeHg on neural progenitor cell (NPC) proliferation, a key stage of neurogenesis during development, remains to be clarified. In this study, we investigated the effect of low concentrations of MeHg on NPCs by using a primary culture system developed using the embryonic rat cerebral cortex. NPC proliferation was suppressed 48h after exposure to 10nM MeHg, but cell death was not observed. Western blot analyses for cyclins A, B, D1, and E demonstrated that MeHg down-regulated cyclin E, a promoter of the G1/S cell cycle transition. Cyclin E has been shown to be degraded following the phosphorylation by glycogen synthase kinase 3ß (GSK-3ß). The time course study showed that GSK-3ß was up-regulated 3h after exposure to 10nM MeHg, and cyclin E degradation 48h after MeHg exposure. We further demonstrated that GSK-3ß inhibitors, lithium and SB-415286, suppressed MeHg-induced inhibition of NPC proliferation by preventing cyclin E degradation. These results suggest that the inhibition of NPC proliferation induced by low concentration of MeHg was associated with up-regulation of GSK-3ß at the early stage and subsequent degeneration of cyclin E.


Assuntos
Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Ciclina E/metabolismo , Poluentes Ambientais/toxicidade , Quinase 3 da Glicogênio Sintase/metabolismo , Compostos de Metilmercúrio/toxicidade , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteólise , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima
8.
Toxicol Appl Pharmacol ; 282(3): 259-66, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25545984

RESUMO

Methylmercury (MeHg) is an environmental toxin which induces cell death specific for the nervous systems. Here we show that MeHg causes neuronal cell death through the suppression of the tropomyosin receptor kinase A (TrkA) pathway, and that compounds activating the TrkA pathway prevent MeHg-induced nerve damage in vitro and in vivo. We first investigated the mechanism of MeHg-induced neurotoxicity in differentiating neurons using PC12 cells. Exposure to 100nM MeHg for 1day induced apoptosis in differentiating PC12 cells. Further, MeHg-induced apoptosis was preceded by inhibition of neurite extension, as determined by ELISA analyses of the neurite-specific protein neurofilament triplet H protein (NF-H). To determine the mechanism of MeHg-induced apoptosis, we evaluated the effects of MeHg on the TrkA pathway, which is known to regulate neuronal differentiation and viability. Western blot analysis demonstrated that, like the TrkA phosphorylation inhibitor K252a, MeHg inhibited phosphorylation of TrkA and its downstream effectors. Furthermore, GM1 ganglioside and its analog MCC-257, which enhance TrkA phosphorylation, overcame the effect of MeHg in neurons, supporting the involvement of the TrkA pathway in MeHg-induced nerve damage. Finally, we demonstrated that MCC-257 rescued the clinical sign and pathological changes in MeHg-exposed rats. These findings indicate that MeHg-induced apoptosis in neuron is triggered by inhibition of the TrkA pathway, and that GM1 ganglioside and MCC-257 effectively prevent MeHg-induced nerve damage.


Assuntos
Poluentes Ambientais/toxicidade , Compostos de Metilmercúrio/toxicidade , Neurônios/efeitos dos fármacos , Receptor trkA/antagonistas & inibidores , Animais , Carbazóis/farmacologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Gangliosídeo G(M1)/análogos & derivados , Gangliosídeo G(M1)/farmacologia , Alcaloides Indólicos/farmacologia , Masculino , Neurônios/metabolismo , Células PC12 , Ratos , Ratos Wistar , Receptor trkA/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Sci Rep ; 14(1): 21832, 2024 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-39294331

RESUMO

Methylmercury (MeHg) is a well-known neurotoxicant that induces various cellular functions depending on cellular- and developmental-specific vulnerabilities. MeHg has a high affinity for selenol and thiol groups, thus impairing the antioxidant system. Such affinity characteristics of MeHg led us to develop sensor vectors to assess MeHg toxicity. In this study, MeHg-mediated defects in selenocysteine (Sec) incorporation were demonstrated using thioredoxin reductase 1 cDNA fused with the hemagglutinin tag sequence at the C-terminus. Taking advantage of such MeHg-mediated defects in Sec incorporation, a cDNA encoding luciferase with a Sec substituted for cysteine-491 was constructed. This construct showed MeHg-induced decreases in signaling in a dose-dependent manner. To directly detect truncated luciferase under MeHg exposure, we further constructed a new sensor vector fused with a target for proteasomal degradation. However, this construct was inadequate because of the low rate of Sec insertion, even in the absence of MeHg. Finally, a Krab transcriptional suppressor fused with Sec was constructed and assessed to demonstrate MeHg-dependent increases in signal intensity. We confirmed that the vector responded specifically and in a dose-dependent manner to MeHg in cultured cerebellar granule cells. This vector is expected to allow monitoring of MeHg-specific toxicity via spatial and temporal imaging.


Assuntos
Compostos de Metilmercúrio , Compostos de Metilmercúrio/toxicidade , Animais , Humanos , Camundongos , Técnicas Biossensoriais/métodos , Luciferases/metabolismo , Luciferases/genética , Cerebelo/metabolismo , Cerebelo/efeitos dos fármacos
10.
J Biol Chem ; 286(8): 6641-9, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21106535

RESUMO

Methylmercury (MeHg) toxicity is a continuous environmental problem to human health. The critical role of oxidative stress in the pathogenesis of MeHg cytotoxicity has been clarified, but the molecular mechanisms underlying MeHg-mediated oxidative stress remain to be elucidated. Here we demonstrate a post-transcriptional effect of MeHg on antioxidant selenoenzymes by using a MeHg-susceptible cell line. MeHg-induced selenium deficiency leads to failure of the recoding of a UGA codon for selenocysteine and results in degradation of the major antioxidant selenoenzyme glutathione peroxidase 1 (GPx1) mRNA by nonsense-mediated mRNA decay (NMD), a cellular mechanism that detects the premature termination codon (PTC) located 5'-upstream of the last exon-exon junction and degrades PTC-containing mRNAs. In contrast, thioredoxin reductase 1 (TrxR1), another antioxidant selenoenzyme of the thioredoxin system, was likely skipped by NMD because of a UGA codon in the last exon. However, TrxR1 activity was decreased despite mRNA up-regulation, which was probably due to the synthesis of aberrant TrxR1 protein without selenocysteine. Changes in selenoenzyme GPx1 and TrxR1 mRNAs were observed earlier than was the incidence of oxidative stress and up-regulation of other antioxidant enzyme mRNAs. Results indicated that the MeHg-induced relative selenium-deficient condition affects the major antioxidant selenoenzymes GPx1 and TrxR1 through a post-transcriptional effect, resulting in the disturbance of cellular redox systems and the incidence of oxidative stress. Treatment with ebselen, a seleno-organic compound, effectively suppressed oxidative stress and protected cells against MeHg-induced relative selenium deficiency and cytotoxicity.


Assuntos
Glutationa Peroxidase/biossíntese , Compostos de Metilmercúrio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Selênio/metabolismo , Selenoproteínas/biossíntese , Tiorredoxina Redutase 1/biossíntese , Animais , Antioxidantes/farmacologia , Azóis/farmacologia , Linhagem Celular , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Isoindóis , Compostos Organosselênicos/farmacologia , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/biossíntese , Ratos , Glutationa Peroxidase GPX1
11.
Toxicol Appl Pharmacol ; 250(1): 1-9, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20869980

RESUMO

Methylmercury (MeHg) is an environmental neurotoxicant which induces neuropathological changes in both the central nervous and peripheral sensory nervous systems. Our recent study demonstrated that down-regulation of Ras-related C3 botulinum toxin substrate 1 (Rac1), which is known to promote neuritic extension, preceded MeHg-induced damage in cultured cortical neurons, suggesting that MeHg-mediated axonal degeneration is due to the disturbance of neuritic extension. Therefore we hypothesized that MeHg-induced axonal degeneration might be caused by neuritic extension/retraction incoordination. This idea brought our attention to the Ras homolog gene (Rho)/Rho-associated coiled coil-forming protein kinase (ROCK) pathway because it has been known to be associated with the development of axon and apoptotic neuronal cell death. Here we show that inhibition of the Rho/ROCK pathway prevents MeHg-intoxication both in vitro and in vivo. A Rho inhibitor, C3 toxin, and 2 ROCK inhibitors, Fasudil and Y-27632, significantly protected against MeHg-induced axonal degeneration and apoptotic neuronal cell death in cultured cortical neuronal cells exposed to 100 nM MeHg for 3 days. Furthermore, Fasudil partially prevented the loss of large pale neurons in dorsal root ganglia, axonal degeneration in dorsal spinal root nerves, and vacuolar degeneration in the dorsal columns of the spinal cord in MeHg-intoxicated model rats (20 ppm MeHg in drinking water for 28 days). Hind limb crossing sign, a characteristic MeHg-intoxicated sign, was significantly suppressed in this model. The results suggest that inhibition of the Rho/ROCK pathway rescues MeHg-mediated neuritic extension/retraction incoordination and is effective for the prevention of MeHg-induced axonal degeneration and apoptotic neuronal cell death.


Assuntos
Compostos de Metilmercúrio/toxicidade , Degeneração Neural/prevenção & controle , Neurônios/efeitos dos fármacos , Quinases Associadas a rho/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Toxinas Botulínicas/farmacologia , Regulação para Baixo , Masculino , Degeneração Neural/induzido quimicamente , Neurônios/patologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Antioxidants (Basel) ; 9(10)2020 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-33081221

RESUMO

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.

13.
Food Chem Toxicol ; 146: 111810, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33058990

RESUMO

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.


Assuntos
Antioxidantes/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Compostos de Metilmercúrio/toxicidade , Compostos de Sulfidrila/sangue , Animais , Biomarcadores/sangue , Proteínas Sanguíneas/metabolismo , Sistema Nervoso Central/fisiopatologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
14.
Neurotoxicology ; 30(1): 16-22, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19000711

RESUMO

Methylmercury (MeHg) has been recognized as a neurotoxicant targeted on the central nervous system including cerebellum and cerebral cortex. Some molecular targets of MeHg have been identified using cerebellar neuronal cells, but little is known in the cerebrocortical neuronal cells. In this study, the molecular mechanism underlying MeHg-induced cell death in cerebrocortical neurons was investigated using a primary culture of embryonic rat cortical neuronal cells. The cultured cells exhibited apoptosis 3 days after exposure to 100 nM MeHg, suggesting the involvement of caspase-dependent apoptotic pathways. We demonstrated for the first time that neuritic degeneration precedes MeHg-induced apoptotic death in neurons exposed to 100 nM MeHg. Immunocytochemical and ELISA analyses for neurite-specific proteins namely, tau and MAP2, showed that injury to tau-positive axons was first induced followed by damage to the dendrites and cellular bodies. To further investigate the factors responsible for neuronal death, we investigated the expression levels of Rho-family proteins (Rac1, Cdc42, and RhoA), which regulate neuritic functions and apoptosis in neurons. Western blot analysis demonstrated that MeHg downregulated the expression levels of Rac1 and Cdc42 but did not affect RhoA. The exposure concentration and time course studies confirmed that Rac1 is targeted during an early stage of MeHg-induced cytotoxicity. The results indicate that neuritic degeneration, in particular axonal degeneration triggered by the downregulation of Rac1 expression, contributes to MeHg-induced apoptotic cell death in cultured cerebrocortical neurons.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Compostos de Metilmercúrio/toxicidade , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Apoptose , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Dendritos/química , Dendritos/efeitos dos fármacos , Regulação para Baixo , Proteínas Associadas aos Microtúbulos/análise , Neuritos/química , Neurônios/química , Ratos , Ratos Sprague-Dawley , Proteína cdc42 de Ligação ao GTP/análise , Proteína rhoA de Ligação ao GTP/análise , Proteínas tau/análise
15.
Toxicol Sci ; 168(1): 126-136, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30462329

RESUMO

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-1ß, 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.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/metabolismo , Masculino , Compostos de Metilmercúrio/efeitos adversos , Compostos de Metilmercúrio/toxicidade , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Raízes Nervosas Espinhais/efeitos dos fármacos , Raízes Nervosas Espinhais/patologia , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia , Proteínas rac1 de Ligação ao GTP/metabolismo
16.
Sci Rep ; 9(1): 1279, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718659

RESUMO

Nonsense-mediated mRNA decay (NMD) is a cellular mechanism that eliminates mRNAs that harbor premature translation termination codons (PTCs). Here, we investigated the effects of environmental stresses (oxidative stress and endoplasmic reticulum (ER) stress) on NMD activity. Methylmercury (MeHg) was used to cause oxidative stress and thapsigargin to stress the ER. NMD suppression, evidenced by upregulation of NMD-sensitive mRNAs and a decrease in UPF1 phosphorylation, was observed in MeHg-treated myogenic cells, cerebral cortical neuronal cells, and astroglial cells. Mild ER stress amplified NMD suppression caused by MeHg. To elucidate the cause of stress-induced NMD suppression, the role of the phospho-eIF2α/ATF4 pathway was investigated. Knockdown and non-phosphorylatable eIF2α-transfection studies demonstrated the critical role of phospho-eIF2α-mediated repression of translation in mild ER stress-induced NMD suppression. However, NMD suppression was also observed in phospho-eIF2α-deficient cells under mild ER stress. Mechanistic target of rapamycin suppression-induced inhibition of cap-dependent translation, and downregulation of the NMD components UPF1, SMG7, and eIF4A3, were probably involved in stress-induced NMD suppression. Our results indicate that stress-induced NMD suppression has the potential to affect the condition of cells and phenotypes of PTC-related diseases under environmental stresses by stabilizing NMD-targeted gene expression.


Assuntos
Estresse do Retículo Endoplasmático , Degradação do RNAm Mediada por Códon sem Sentido , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 4A em Eucariotos/genética , Fator de Iniciação 4A em Eucariotos/metabolismo , Humanos , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Mensageiro/genética , Transativadores/genética , Transativadores/metabolismo
17.
Front Physiol ; 10: 759, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281262

RESUMO

Muscle atrophy can be caused by unloading stress such as microgravity environments or cast immobilization. Therapies for such disuse muscle atrophy and their underlying mechanisms are incompletely understood. Here, we investigated the therapeutic effects of local vibration stimulation on immobilization-induced skeletal muscle atrophy. A rat model was made by placing the left hindlimb in a cast for 1 week, leading to oxidative myofiber atrophy without myopathic changes in soleus skeletal muscle. Vibration stimulus (90 Hz, 15 min) to the plantar fascia of the atrophic hindlimb was performed once a day using a hand-held vibration massager after removal of a cast at the end of the immobilization period. After 2 weeks, rats were dissected, and quantitative analysis of the cross-sectional areas of soleus myofibers was performed. The results revealed that vibration induced significant recovery from disuse muscle atrophy, compared with untreated immobilized samples. Furthermore, vibration treatment suppressed the fiber transition from slow to fast fiber types compared with vibration-untreated immobilized samples. Western blotting analyses of mechanical stress-induced factors revealed that the expression of mechano-growth factor (MGF), systemic insulin-like growth factor I, and the mechanotransduction protein, Yes-associated protein 1 (YAP1), was decreased in untreated immobilized soleus muscle, whereas vibration stimulation restored their expression. No change in the level of phosphorylation of YAP1Ser127 was observed, leading to no change in p-YAP1/YAP1 ratio in vibration-treated immobilized soleus muscle. The results indicate that vibration stimulus is effective to restore immobilization-induced inactivation of YAP1 pathway. Phosphorylation of ERK 1/2, but not AKT, was enhanced in vibration-treated immobilized soleus muscle. Furthermore, vibration stimuli restored immobilization-induced downregulation of the paired box transcription factor, PAX7, a critical factor for regenerative myogenesis in muscle satellite cells. Our results indicate that cyclic vibration stimuli are effective in activating satellite cells and facilitate recovery from immobilization-induced oxidative myofiber atrophy through upregulation of MGF and YAP1.

18.
Neurotoxicology ; 29(1): 22-30, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17920127

RESUMO

Cellular stress responses following exposure to methylmercury (MeHg) were investigated using myogenic cell lines that showed different susceptibilities to MeHg. The susceptible cell line showed apoptosis within 24h after exposure to low levels of MeHg. The activation of caspase 12, 9, and 3 was detected in the apoptotic cells at 14-16 h after MeHg exposure, suggesting that MeHg causes apoptosis via both mitochondria- and endoplasmic reticulum (ER)-generated processes. An early increase in the level of intracellular reactive oxygen species (ROS) was quantitatively recognized since 2-3h after exposure to MeHg in both MeHg-susceptible and non-susceptible cell lines; however, the increase was lower in the latter cell line. The phosphorylation of apoptosis signal-regulating kinase 1 (ASK1) was also recognized in both cell lines, with the increase in intracellular ROS. However, the activation of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) pathways was observed only in the MeHg-susceptible cell line. In contrast, the non-susceptible cell line exhibited activation of the cell survival ERK pathway. Up-regulation of metallothioneine I and Hic-5 mRNAs encoding proteins induced by oxidative stress was recognized during the early stage of MeHg cytotoxicity in the MeHg-susceptible cell line. Quantitative real-time PCR and western blot analyses confirmed that ER stress is a late event during MeHg cytotoxicity. Coaddition of the antioxidant Trolox dramatically suppressed the increase in the level of ROS, activation of caspases and, finally, apoptosis. However, later treatment with Trolox attenuated its protective effect against MeHg cytotoxicity. The results indicate that failure to protect cells against the early oxidative stress triggers ER stress and apoptosis processes. Combined treatment with protective factors against oxidative and ER stresses is necessary, especially in the later stages of MeHg cytotoxicity.


Assuntos
Apoptose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , MAP Quinase Quinase Quinase 5/fisiologia , Compostos de Metilmercúrio/toxicidade , Mitocôndrias/efeitos dos fármacos , Mioblastos , Transdução de Sinais/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Caspases/metabolismo , Linhagem Celular Transformada , Cromanos/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Camundongos , Mutação/fisiologia , Mioblastos/efeitos dos fármacos , Mioblastos/fisiologia , Mioblastos/ultraestrutura , Miotonina Proteína Quinase , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Transfecção/métodos , Expansão das Repetições de Trinucleotídeos/genética , Regulação para Cima/efeitos dos fármacos
19.
Neurotoxicology ; 67: 226-233, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29913201

RESUMO

Methylmercury (MeHg) induces site-specific cerebrocortical neuronal cell death. In our previous study using an in vivo mouse model, we reported that MeHg-induced cerebrocortical neuronal cell death may be due to neural hyperactivity triggered by activation of kinase pathways. However, the detailed molecular mechanism remained to be completely understood. In this study, we analyzed detailed signaling pathways for MeHg-induced neuronal cell death using all-trans-retinoic acid (RA) differentiated SH-SY5Y cells, which show neuron-like morphological changes and express neuron/synapse markers for cerebrocortical neurons. Time course studies revealed that MeHg-induced upregulation of c-fos, a marker of neural activation, preceded neuronal cell death. These results were similar to those observed in a MeHg-intoxicated mouse model. We observed early expression of the oxidative stress marker thymidine glycol followed by activation of p44/42 mitogen-activated protein kinase (MAPK) and p38 MAPK, and an increase in cAMP response element binding protein (CREB). Investigation of the effects of specific kinase inhibitors revealed that SB203580, a specific inhibitor for p38 MAPK, significantly blocked the upregulation of c-fos and the subsequent neuronal cell death. In contrast, PD98059 and U0126, specific inhibitors for p44/p42 MAPK, showed no effects on MeHg-induced neurotoxicity. Furthermore, the antioxidants Trolox and edaravone significantly suppressed MeHg-induced thymidine glycol expression, p38 MAPK-CREB pathway activation, and neurotoxicity. Altogether, these results suggest that MeHg-induced oxidative stress and subsequent activation of the p38 MAPK-CREB pathway contribute to cerebrocortical neuronal hyperactivity and subsequent neuronal cell death.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Compostos de Metilmercúrio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Humanos , Estresse Oxidativo/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
20.
Toxicol Lett ; 271: 66-73, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28267559

RESUMO

Methylmercury (MeHg) induces site-specific neurotoxicity in the adult brain. In this study, we investigated the site-specific expression of the signaling cascade related to neural activity in a mouse model of MeHg intoxication showing neurodegeneration only in the deep layer of the cerebral cortex, especially layer IV. We performed time course studies of c-fos and brain-derived neurotrophic factor (BDNF) expression levels which are proper markers of neural activity. We showed that upregulation of both markers preceded the neuronal degeneration in the cerebral cortex. Immunohistochemical analysis revealed the site-specific upregulation of c-fos in the deep layer of the cerebral cortex. Western blot analysis showed that c-fos and BDNF expression was associated with CREB phosphorylation, which was triggered by the activation of the p44/42 MAPK, p38 MAPK and PKA pathways. However, we did not detect any changes in the expression levels of c-fos and BDNF proteins and no signs of neuronal degeneration in the hippocampus and cerebellum, despite the fact that we could detect accumulation of MeHg in these two brain regions. These results suggested an intriguing possibility that MeHg-induced neuronal degeneration was caused by site-specific neural hyperactivity triggered by the activation of MAPK and PKA/CREB pathways followed by c-fos and BDNF upregulation.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Intoxicação do Sistema Nervoso por Mercúrio/prevenção & controle , Compostos de Metilmercúrio , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Degeneração Neural , Neurônios/efeitos dos fármacos , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Ativação Enzimática , Imuno-Histoquímica , Masculino , Intoxicação do Sistema Nervoso por Mercúrio/enzimologia , Intoxicação do Sistema Nervoso por Mercúrio/patologia , Intoxicação do Sistema Nervoso por Mercúrio/fisiopatologia , Camundongos Endogâmicos ICR , Neurônios/enzimologia , Neurônios/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transdução de Sinais , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA