Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
PLoS One ; 12(1): e0170623, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28118383

RESUMO

Clinical manifestations of methylmercury (MeHg) intoxication include cerebellar ataxia, concentric constriction of visual fields, and sensory and auditory disturbances. The symptoms depend on the site of MeHg damage, such as the cerebellum and occipital lobes. However, the underlying mechanism of MeHg-induced tissue vulnerability remains to be elucidated. In the present study, we used a rat model of subacute MeHg intoxication to investigate possible MeHg-induced blood-brain barrier (BBB) damage. The model was established by exposing the rats to 20-ppm MeHg for up to 4 weeks; the rats exhibited severe cerebellar pathological changes, although there were no significant differences in mercury content among the different brain regions. BBB damage in the cerebellum after MeHg exposure was confirmed based on extravasation of endogenous immunoglobulin G (IgG) and decreased expression of rat endothelial cell antigen-1. Furthermore, expression of vascular endothelial growth factor (VEGF), a potent angiogenic growth factor, increased markedly in the cerebellum and mildly in the occipital lobe following MeHg exposure. VEGF expression was detected mainly in astrocytes of the BBB. Intravenous administration of anti-VEGF neutralizing antibody mildly reduced the rate of hind-limb crossing signs observed in MeHg-exposed rats. In conclusion, we demonstrated for the first time that MeHg induces BBB damage via upregulation of VEGF expression at the BBB in vivo. Further studies are required in order to determine whether treatment targeted at VEGF can ameliorate MeHg-induced toxicity.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Ataxia Cerebelar/induzido quimicamente , Cerebelo/efeitos dos fármacos , Intoxicação do Sistema Nervoso por Mercúrio/genética , Compostos de Metilmercúrio/toxicidade , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Anticorpos Neutralizantes/uso terapêutico , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Química Encefálica , Ataxia Cerebelar/tratamento farmacológico , Ataxia Cerebelar/fisiopatologia , Cerebelo/metabolismo , Cerebelo/patologia , Masculino , Mercúrio/análise , Intoxicação do Sistema Nervoso por Mercúrio/metabolismo , Intoxicação do Sistema Nervoso por Mercúrio/patologia , Compostos de Metilmercúrio/farmacologia , Lobo Occipital/efeitos dos fármacos , Lobo Occipital/metabolismo , Lobo Occipital/patologia , Ratos , Ratos Wistar , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética
7.
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
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 ; 3: 2346, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23907635

RESUMO

We demonstrate that methylmercury (MeHg)-susceptible cells preconditioned with an inhibitor of endoplasmic reticulum (ER) Ca(2+)-ATPase, thapsigargin, showed resistance to MeHg cytotoxicity through favorable stress responses, which included phosphorylation of eukaryotic initiation factor 2 alpha (Eif2α), accumulation of activating transcription factor 4 (Atf4), upregulation of stress-related proteins, and activation of extracellular signal regulated kinase pathway. In addition, ER stress preconditioning induced suppression of nonsense-mediated mRNA decay (NMD) mainly through the phospho-Eif2α-mediated general suppression of translation initiation and possible combined effects of decreased several NMD components expression. Atf4 accumulation was not mediated by NMD inhibition but translation inhibition of its upstream open reading frame (uORF) and translation facilitation of its protein-coding ORF by the phospho-Eif2α. These results suggested that ER stress plays an important role in MeHg cytotoxicity and that the modulation of ER stress has therapeutic potential to attenuate MeHg cytotoxicity, the underlying mechanism being the induction of integrated stress responses.


Assuntos
Apoptose/fisiologia , Retículo Endoplasmático/fisiologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Compostos de Metilmercúrio/farmacologia , Mioblastos/fisiologia , Estresse Fisiológico/fisiologia , Tapsigargina/farmacologia , Adenosina Trifosfatases/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Retículo Endoplasmático/efeitos dos fármacos , Camundongos , Mioblastos/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA