Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Glia ; 60(7): 1024-36, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22488924

RESUMO

Glutamate transporter-1 (GLT-1) plays a central role in preventing excitotoxicity by removing excess glutamate from the synaptic clefts. 17ß-Estradiol (E2) and tamoxifen (TX), a selective estrogen receptor (ER) modulator, afford neuroprotection in a range of experimental models. However, the mechanisms that mediate E2 and TX neuroprotection have yet to be elucidated. We tested the hypothesis that E2 and TX enhance GLT-1 function by increasing transforming growth factor (TGF)-α expression and, thus, attenuate manganese (Mn)-induced impairment in astrocytic GLT-1 expression and glutamate uptake in rat neonatal primary astrocytes. The results showed that E2 (10 nM) and TX (1 µM) increased GLT-1 expression and reversed the Mn-induced reduction in GLT-1, both at the mRNA and protein levels. E2/TX also concomitantly reversed the Mn-induced inhibition of astrocytic glutamate uptake. E2/TX activated the GLT-1 promoter and attenuated the Mn-induced repression of the GLT-1 promoter in astrocytes. TGF-α knockdown (siRNA) abolished the E2/TX effect on GLT-1 expression, and inhibition of epidermal growth factor receptor (TGF-α receptor) suppressed the effect of E2/TX on GLT-1 expression and GLT-1 promoter activity. E2/TX also increased TGF-α mRNA and protein levels with a concomitant increase in astrocytic glutamate uptake. All ERs (ER-α, ER-ß, and G protein-coupled receptor 30) were involved in mediating E2 effects on the regulation of TGF-α, GLT-1, and glutamate uptake. These results indicate that E2/TX increases GLT-1 expression in astrocytes via TGF-α signaling, thus offering an important putative target for the development of novel therapeutics for neurological disorders.


Assuntos
Astrócitos/efeitos dos fármacos , Estradiol/farmacologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Transportador 2 de Aminoácido Excitatório/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno , Ratos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Fator de Crescimento Transformador alfa/genética , Regulação para Cima/fisiologia
2.
Free Radic Biol Med ; 52(6): 1067-74, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22245093

RESUMO

Excessive exposure to manganese (Mn) increases levels of oxidative stressors and proinflammatory mediators, such as cyclooxygenase-2 and prostaglandin E(2). Mn also activates nuclear factor-κB (NF-κB), an important mediator of inflammation. The signaling molecule 15-deoxy-Δ12,14-prostaglandin J(2) (15 d-PGJ(2)) is an anti-inflammatory prostaglandin. Here, we tested the hypothesis that 15 d-PGJ(2) modulates Mn-induced activation of astrocytic intracellular signaling, including NF-κB and nuclear factor erythroid 2-related factor (Nrf2), a master regulator of antioxidant transcriptional responses. The results establish that 15 d-PGJ(2) suppresses Mn-induced NF-κB activation by interacting with several signaling pathways. The PI3K/Akt pathway, which is upstream of NF-κB, plays a role in this activation, because (i) pretreatment with 15 d-PGJ(2) (10 µM for 1h) significantly (p<0.01) inhibited Mn (500 µM)-induced PI3K/Akt activation and (ii) inhibition of the PI3K/Akt pathway with LY29004 significantly (p<0.05) decreased NF-κB activation. 15 d-PGJ(2) also significantly (p<0.05) attenuated Mn-induced astrocytic NF-κB activation by inhibiting the Mn-induced phosphorylation of IκB kinase and subsequent IκB-α degradation. Because Mn-induced oxidative stress is also associated with Nrf2 activation, additional studies addressed the ability of 15 d-PGJ(2) to modulate the Nrf2 pathway. 15 d-PGJ(2) significantly (p<0.01) increased Nrf2 expression in whole-cell lysates. Consistent with its pro-oxidant properties, Mn also increased Nrf2 expression. Nevertheless, cotreatment of whole-cell lysates with both Mn and 15 d-PGJ(2) partially suppressed (p<0.01) the 15 d-PGJ(2)-induced increase in astrocytic Nrf2 protein expression. Mn treatment also decreased (p<0.001) expression of DJ-1, a Parkinson disease-associated protein and a stabilizer of Nrf2, and 15 d-PGJ(2) attenuated Mn-induced astrocytic inhibition of DJ-1 expression. Collectively, these results demonstrate that 15d-PGJ(2) exerts a protective effect in astrocytes against Mn-induced inflammation and oxidative stress by modulating the activation of the NF-κB and Nrf2 signaling pathways.


Assuntos
Astrócitos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Oncogênicas/metabolismo , Doença de Parkinson/metabolismo , Prostaglandina D2/análogos & derivados , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Células Cultivadas , Cromonas/farmacologia , Citoproteção/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Manganês/metabolismo , Morfolinas/farmacologia , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/metabolismo , Proteínas Oncogênicas/genética , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Prostaglandina D2/farmacologia , Proteína Desglicase DJ-1 , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
3.
Neurotoxicology ; 32(3): 291-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21300091

RESUMO

Methylmercury (MeHg) preferentially accumulates in glia of the central nervous system (CNS), but its toxic mechanisms have yet to be fully recognized. In the present study, we tested the hypothesis that MeHg induces neurotoxicity via oxidative stress mechanisms, and that these effects are attenuated by the antioxidant, ebselen. Rat neonatal primary cortical astrocytes were pretreated with or without 10 µM ebselen for 2h followed by MeHg (0, 1, 5, and 10 µM) treatments. MeHg-induced changes in astrocytic [(3)H]-glutamine uptake were assessed along with changes in mitochondrial membrane potential (ΔΨ(m)), using the potentiometric dye tetramethylrhodamine ethyl ester (TMRE). Western blot analysis was used to detect MeHg-induced ERK (extracellular-signal related kinase) phosphorylation and caspase-3 activation. MeHg treatment significantly decreased (p<0.05) astrocytic [(3)H]-glutamine uptake at all time points and concentrations. Ebselen fully reversed MeHg's (1 µM) effect on [(3)H]-glutamine uptake at 1 min. At higher MeHg concentrations, ebselen partially reversed the MeHg-induced astrocytic inhibition of [(3)H]-glutamine uptake [at 1 min (5 and 10 µM) (p<0.05); 5 min (1, 5 and 10 µM) (p<0.05)]. MeHg treatment (1h) significantly (p<0.05) dissipated the ΔΨ(m) in astrocytes as evidenced by a decrease in mitochondrial TMRE fluorescence. Ebselen fully reversed the effect of 1 µM MeHg treatment for 1h on astrocytic ΔΨ(m) and partially reversed the effect of 5 and 10 µM MeHg treatments for 1h on ΔΨ(m). In addition, ebselen inhibited MeHg-induced phosphorylation of ERK (p<0.05) and blocked MeHg-induced activation of caspase-3 (p<0.05-0.01). These results are consistent with the hypothesis that MeHg exerts its toxic effects via oxidative stress and that the phosphorylation of ERK and the dissipation of the astrocytic mitochondrial membrane potential are involved in MeHg toxicity. In addition, the protective effects elicited by ebselen reinforce the idea that organic selenocompounds represent promising strategies to counteract MeHg-induced neurotoxicity.


Assuntos
Antioxidantes/farmacologia , Astrócitos/efeitos dos fármacos , Azóis/farmacologia , Poluentes Ambientais/toxicidade , Intoxicação do Sistema Nervoso por Mercúrio/etiologia , Compostos de Metilmercúrio/toxicidade , Fármacos Neuroprotetores/farmacologia , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Astrócitos/patologia , Western Blotting , Caspase 3/metabolismo , Células Cultivadas , Citoproteção , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glutamina/metabolismo , Isoindóis , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Intoxicação do Sistema Nervoso por Mercúrio/metabolismo , Intoxicação do Sistema Nervoso por Mercúrio/patologia , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
4.
Glia ; 59(5): 810-20, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21351162

RESUMO

As the two major glial cell types in the brain, astrocytes and microglia play pivotal but different roles in maintaining optimal brain function. Although both cell types have been implicated as major targets of methylmercury (MeHg), their sensitivities and adaptive responses to this metal can vary given their distinctive properties and physiological functions. This study was carried out to compare the responses of astrocytes and microglia following MeHg treatment, specifically addressing the effects of MeHg on cell viability, reactive oxygen species (ROS) generation and glutathione (GSH) levels, as well as mercury (Hg) uptake and the expression of NF-E2-related factor 2 (Nrf2). Results showed that microglia are more sensitive to MeHg than astrocytes, a finding that is consistent with their higher Hg uptake and lower basal GSH levels. Microglia also demonstrated higher ROS generation compared with astrocytes. Nrf2 and its downstream genes were upregulated in both cell types, but with different kinetics (much faster in microglia). In summary, microglia and astrocytes each exhibit a distinct sensitivity to MeHg, resulting in their differential temporal adaptive responses. These unique sensitivities appear to be dependent on the cellular thiol status of the particular cell type.


Assuntos
Astrócitos/efeitos dos fármacos , Compostos de Metilmercúrio/farmacologia , Microglia/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Glutationa/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Imuno-Histoquímica , Compostos de Metilmercúrio/metabolismo , Microglia/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
5.
Toxicol Sci ; 116(2): 590-603, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20421342

RESUMO

The neurotoxicity of methylmercury (MeHg) is well documented in both humans and animals. MeHg causes acute and chronic damage to multiple organs, most profoundly the central nervous system (CNS). Microglial cells are derived from macrophage cell lineage, making up approximately 12% of cells in the CNS, yet their role in MeHg-induced neurotoxicity is not well defined. The purpose of the present study was to characterize microglial vulnerability to MeHg and their potential adaptive response to acute MeHg exposure. We examined the effects of MeHg on microglial viability, reactive oxygen species (ROS) generation, glutathione (GSH) level, redox homeostasis, and Nrf2 protein expression. Our data showed that MeHg (1-5 microM) treatment caused a rapid (within 1 min) concentration- and time-dependent increase in ROS generation, accompanied by a statistically significant decrease in the ratio of GSH and its oxidized form glutathione disulfide (GSSG) (GSH:GSSG ratio). MeHg increased the cytosolic Nrf2 protein level within 1 min of exposure, followed by its nuclear translocation after 10 min of treatment. Consistent with the nuclear translocation of Nrf2, quantitative real-time PCR revealed a concentration-dependent increase in the messenger RNA level of Ho-1, Nqo1, and xCT 30 min post MeHg exposure, whereas Nrf2 knockdown greatly reduced the upregulation of these genes. Furthermore, we observed increased microglial death upon Nrf2 knockdown by the small hairpin RNA approach. Taken together, our study has demonstrated that microglial cells are exquisitely sensitive to MeHg and respond rapidly to MeHg by upregulating the Nrf2-mediated antioxidant response.


Assuntos
Compostos de Metilmercúrio/toxicidade , Microglia/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/análise , Estresse Oxidativo/efeitos dos fármacos , Sistema y+ de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Acídicos , Animais , Células Cultivadas , Glutationa/metabolismo , Heme Oxigenase (Desciclizante)/genética , Interleucina-6/biossíntese , Microglia/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Fator 2 Relacionado a NF-E2/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
6.
Brain Res ; 1326: 152-61, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20170646

RESUMO

Manganese (Mn) is an essential trace metal; however, exposure to high Mn levels can result in neurodegenerative changes resembling Parkinson's disease (PD). Information on Mn's effects on endothelial cells of the blood-brain barrier (BBB) is lacking. Accordingly, we tested the hypothesis that BBB endothelial cells are a primary target for Mn-induced neurotoxicity. The studies were conducted in an in vitro BBB model of immortalized rat brain endothelial (RBE4) cells. ROS production was determined by F(2)-isoprostane (F(2)-IsoPs) measurement. The relationship between Mn toxicity and redox status was investigated upon intracellular glutathione (GSH) depletion with diethylmaleate (DEM) or L-buthionine sulfoximine (BSO). Mn exposure (200 or 800 microM MnCl(2) or MnSO(4)) for 4 or 24h led to significant decrease in cell viability vs. controls. DEM or BSO pre-treatment led to further enhancement in cytotoxicity vs. exposure to Mn alone, with more pronounced cell death after 24-h DEM pre-treatment. F(2)-IsoPs levels in cells exposed to MnCl(2) (200 or 800 microM) were significantly increased after 4h and remained elevated 24h after exposure compared with controls. Consistent with the effects on cell viability and F(2)-IsoPs, treatment with MnCl(2) (200 or 800 microM) was also associated with a significant decrease in membrane potential. This effect was more pronounced in cells exposed to DEM plus MnCl(2) vs. cells exposed to Mn alone. We conclude that Mn induces direct injury to mitochondria in RBE4 cells. The ensuing impairment in energy metabolism and redox status may modify the restrictive properties of the BBB compromising its function.


Assuntos
Barreira Hematoencefálica/citologia , Células Endoteliais/efeitos dos fármacos , Manganês/toxicidade , Oligoelementos/toxicidade , Animais , Butionina Sulfoximina/farmacologia , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Glutationa/metabolismo , Maleatos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
7.
J Neurochem ; 112(5): 1190-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20002294

RESUMO

Although manganese (Mn) is an essential trace element for human development and growth, chronic exposure to excessive Mn levels can result in psychiatric and motor disturbances, referred to as manganism. However, there are no known mechanism(s) for efflux of excess Mn from mammalian cells. Here, we test the hypothesis that the cytoplasmic iron (Fe) exporter ferroportin (Fpn) may also function as a Mn exporter to attenuate Mn toxicity. Using an inducible human embryonic kidney (HEK293T) cell model, we examined the influence of Fpn expression on Mn-induced cytotoxicity and intracellular Mn concentrations. We found that induction of an Fpn-green fluorescent protein fusion protein in HEK293T cells was cytoprotective against several measures of Mn toxicity, including Mn-induced cell membrane leakage and Mn-induced reductions in glutamate uptake. Fpn-green fluorescent protein mediated cytoprotection correlated with decreased Mn accumulation following Mn exposure. Thus, Fpn expression reduces Mn toxicity concomitant with reduced Mn accumulation. To determine if mammalian cells may utilize Fpn in response to increased intracellular Mn concentrations and toxicity, we assessed endogenous Fpn levels in Mn-exposed HEK293T cells and in mouse brain in vivo. We find that 6 h of Mn exposure in HEK293T cells is associated with a significant increase in Fpn levels. Furthermore, mice exposed to Mn showed an increase in Fpn levels in both the cerebellum and cortex. Collectively, these results indicate that (i) Mn exposure promotes Fpn protein expression, (ii) Fpn expression reduces net Mn accumulation, and (iii) reduces cytotoxicity associated with exposure to this metal.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/uso terapêutico , Intoxicação por Manganês/tratamento farmacológico , Intoxicação por Manganês/metabolismo , Manganês/toxicidade , Análise de Variância , Animais , Proteínas de Transporte de Cátions/genética , Linhagem Celular Transformada , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ecdisterona/análogos & derivados , Ecdisterona/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Intoxicação por Manganês/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Transfecção
8.
J Neurochem ; 110(2): 530-44, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19453300

RESUMO

Chronic exposure to manganese (Mn) can cause manganism, a neurodegenerative disorder similar to Parkinson's disease. The toxicity of Mn includes impairment of astrocytic glutamate transporters. 17beta-Estradiol (E2) has been shown to be neuroprotective in various neurodegenerative diseases including Parkinson's disease and Alzheimer's disease, and some selective estrogen receptor modulators, including tamoxifen (TX), also possess neuroprotective properties. We have tested our hypothesis that E2 and TX reverse Mn-induced glutamate transporter impairment in astrocytes. The results established that E2 and TX increased glutamate transporter function and reversed Mn-induced glutamate uptake inhibition, primarily via the up-regulation of glutamate/aspartate transporter (GLAST). E2 and TX also increased astrocytic GLAST mRNA levels and attenuated the Mn-induced inhibition of GLAST mRNA expression. In addition, E2 and TX effectively increased the expression of transforming growth factor beta1, a potential modulator of the stimulatory effects of E2/TX on glutamate transporter function. This effect was mediated by the activation of MAPK/extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt signaling pathways. These novel findings suggest, for the first time, that E2 and TX enhance astrocytic glutamate transporter expression via increased transforming growth factor beta1 expression. Furthermore, the present study is the first to show that both E2 and TX effectively reverse Mn-induced glutamate transport inhibition by restoring its expression and activity, thus offering a potential therapeutic modality in neurodegenerative disorders characterized by altered glutamate homeostasis.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Astrócitos/metabolismo , Estrogênios/farmacologia , Manganês/toxicidade , Tamoxifeno/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Manganês/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
9.
Neurochem Int ; 55(1-3): 136-42, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19428818

RESUMO

Methylmercury (MeHg) is an environmental toxicant that induces enduring neuropsychological deficits in humans. Although the mechanisms associated with MeHg-induced neurotoxicity have not yet been fully elucidated, some lines of evidence point out to excitatory amino acids dyshomeostasis as an important outcome of MeHg exposure. The present study was designed to characterize the effects of MeHg on amino acid content in co-cultured astrocytes and neurons or in each cell type under solitary conditions. The results showed that glutamate concentrations significantly decreased in neurons, but not in astrocyte cultures exposed to 10 microM MeHg. The decrease in neurons was fully reversed when these cells were co-cultured with astrocytes. The content of other amino acids (aspartate, alanine, glycine and serine) decreased upon exposure to 10 microM MeHg in both neurons and astrocytes cultured in solitary conditions, although the effect was generally smaller in astrocytes than in neurons. However, the content of these amino acids in each of the cell types was indistinguishable from controls when co-cultures were treated with MeHg. Overall, the results indicate that astrocytes, which are more resistant to amino acid modulation by MeHg, can (i) mitigate the effects of MeHg that occur in neurons cultured in solitary conditions and (ii) become themselves more MeHg resistant in the presence of neurons. Delineating the mechanisms underlying the mutual neuroprotective effects of astrocytes and neurons in co-culture to MeHg-induced amino acid imbalance requires further investigation.


Assuntos
Aminoácidos/metabolismo , Astrócitos/metabolismo , Compostos de Metilmercúrio/toxicidade , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
10.
Toxicol Sci ; 110(1): 156-67, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19383943

RESUMO

Chronic exposure to manganese (Mn) leads to a neurological disorder, manganism, which shares multiple common features with idiopathic Parkinson disease (IPD). 17beta-Estradiol (E2) and some selective estrogen receptor modulators, including tamoxifen (TX), afford neuroprotection in various experimental models of neurodegeneration. However, the neuroprotective effects and mechanisms of E2/TX in Mn-induced toxicity have yet to be documented. Herein, we studied the ability of E2/TX to protect rat cortical primary neuronal and astroglial cultures from Mn-induced toxicity. Cell viability, Western blot, and reactive oxygen species (ROS) generation were assessed. Results established that both E2 (10nM) and TX (1 microM) attenuated Mn-induced toxicity. The protective effects of E2/TX were more pronounced in astrocytes versus neurons. The E2-mediated attenuation of Mn-induced ROS generation in astrocytes at 6-h treatment (where no cell death was detected) was mediated by a classical estrogen receptor (ER) pathway and the TX-mediated effect on Mn-induced ROS generation was not mediated via classical ER-dependent mechanisms and likely by its antioxidant properties. The phosphatidylinositol-3 kinase (PI3K)/Akt signaling pathway was involved in both E2- and TX-induced attenuation of Mn-induced ROS formation (6 h) in astrocytes. Treatments with Mn for a longer duration (24 h) led to significant cell death, and the protective effects of E2 and TX were (1) not mediated by a classical ER pathway and (2) associated with activation of both mitogen-activated protein kinase/extracellular signal-regulated kinase and PI3K/Akt signaling pathways. Taken together, the results suggest that both E2 and TX offer effective therapeutic means for neuroprotection against Mn-induced toxicity.


Assuntos
Astrócitos/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Estradiol/farmacologia , Intoxicação por Manganês/prevenção & controle , Manganês/antagonistas & inibidores , Manganês/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores , Tamoxifeno/farmacologia , Animais , Antioxidantes/farmacologia , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Oncogênica v-akt/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Toxicol Sci ; 107(1): 135-43, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18815141

RESUMO

Methylmercury (MeHg) is a potent neurotoxicant and preferentially induces oxidative injury in astrocytes. In neuronal tissues, nuclear factor erythroid 2-related factor 2 (Nrf2) is a key factor determining the protective antioxidant response against various environmental toxicants. Nrf2 is subjected to regulation by many other signaling pathways. The purpose of this study is to characterize its interaction with the phosphatidylinositol 3 (PI3) kinase in cultured rat neonatal primary astrocytes. The results showed that at pathologically relevant concentrations, exposure of primary astrocytes to MeHg led to Nrf2 activation and upregulation of its downstream antioxidant genes. Inhibition of the PI3 kinase resulted in decreased Nrf2 activity, decreased cellular glutathione, and increased cell death to high-dose MeHg. The functional interaction between the two signaling pathways underlined an important mechanism for astrocyte protection against MeHg toxicity. Modulation of Nrf2 by pharmacological modalities should afford a treatment to attenuate MeHg-induced neurotoxicity.


Assuntos
Astrócitos/metabolismo , Compostos de Metilmercúrio/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Cromonas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Compostos de Metilmercúrio/toxicidade , Morfolinas/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Interferência de RNA , Ratos , Regulação para Cima/efeitos dos fármacos
12.
J Neurochem ; 107(4): 1083-90, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18793329

RESUMO

Methylmercury (MeHg) is a potent neurotoxin. The mechanism(s) that governs MeHg transport across the blood-brain barrier and other biological membranes remains unclear. This study addressed the role of the L-type large neutral amino acid transporter, LAT1, in MeHg transport. Studies were carried out in CHO-k1 cells. Over-expression of LAT1 in these cells was associated with enhanced uptake of [(14)C]-MeHg when treated with L-cysteine, but not with the D-cysteine conjugate. In the presence of excess L-methionine, a substrate for LAT1, L-cysteine-conjugated [(14)C]-MeHg uptake was significantly attenuated. Treatment of LAT-1 over-expressing CHO-k1 cells with L-cysteine-conjugated MeHg was also associated with increased leakage of lactate dehydrogenase into the media as well as reduced cell viability measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction assay. In contrast, knock-down of LAT1 decreased the uptake of l-cysteine-conjugated MeHg and attenuated the effects of MeHg on lactate dehydrogenase leakage and CHO-k1 cell viability. These results indicate that the MeHg-L-cysteine conjugate is a substrate for the neutral amino acid transporter, LAT1, which actively transports MeHg across membranes.


Assuntos
Cisteína/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Compostos de Metilmercúrio/metabolismo , Análise de Variância , Animais , Células CHO , Isótopos de Carbono/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Cisteína/farmacologia , Humanos , L-Lactato Desidrogenase/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/genética , Compostos de Metilmercúrio/farmacologia , RNA Interferente Pequeno/farmacologia , Sais de Tetrazólio , Tiazóis , Transfecção/métodos
13.
Toxicol Sci ; 98(1): 198-205, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17468184

RESUMO

Excessive free radical formation has been implicated as a causative factor in neurotoxic damage associated with exposures to a variety of metals, including manganese (Mn). It is well established that Mn accumulates in astrocytes, affecting their ability to indirectly induce and/or exacerbate neuronal dysfunction. The present study examined the effects of Mn treatment on the following endpoints in primary astrocyte cultures: (1) oxidative injury, (2) alterations in high-energy phosphate (adenosine 5'-triphosphate, ATP) levels, (3) mitochondrial inner membrane potential, and (4) glutamine uptake and the expression of glutamine transporters. We quantified astrocyte cerebral oxidative damage by measuring F(2)-isoprostanes (F(2)-IsoPs) using stable isotope dilution methods followed by gas chromatography-mass spectrometry with selective ion monitoring. Our data showed a significant (p < 0.01) elevation in F(2)-IsoPs levels at 2 h following exposure to Mn (100 microM, 500 microM, or 1 mM). Consistent with this observation, Mn induced a concentration-dependent reduction in ATP and the inner mitochondrial membrane potential (DeltaPsi(m)), measured by the high pressure liquid chromatography method and the potentiometric dye, tetramethyl rhodamine ethyl ester, respectively. Moreover, 30 min of pretreatment with Mn (100 microM, 500 microM, or 1 mM) inhibited the net uptake of glutamine (GLN) ((3)H-glutamine) measured at 1 and 5 min. Expression of the messenger RNA coding the GLN transporters, SNAT3/SN1 and SNAT1, was inhibited after 100 and 500 microM Mn treatment for 24 h. Our results demonstrate that induction of oxidative stress, associated mitochondrial dysfunction, and alterations in GLN/glutamate cycling in astrocytes represent key mechanisms by which Mn exerts its neurotoxicity.


Assuntos
Astrócitos/patologia , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/patologia , Estresse Oxidativo/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Sistema ASC de Transporte de Aminoácidos/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Espaço Extracelular/efeitos dos fármacos , F2-Isoprostanos/metabolismo , Glutamina/metabolismo , L-Lactato Desidrogenase/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Antígenos de Histocompatibilidade Menor , Membranas Mitocondriais/efeitos dos fármacos , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo
14.
Brain Res ; 1131(1): 1-10, 2007 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-17182013

RESUMO

The neurotoxicity of high levels of methylmercury (MeHg) is well established both in humans and experimental animals. Astrocytes accumulate MeHg and play a prominent role in mediating MeHg toxicity in the central nervous system (CNS). Although the precise mechanisms of MeHg neurotoxicity are ill-defined, oxidative stress and altered mitochondrial and cell membrane permeability appear to be critical factors in its pathogenesis. The present study examined the effects of MeHg treatment on oxidative injury, mitochondrial inner membrane potential, glutamine uptake and expression of glutamine transporters in primary astrocyte cultures. MeHg caused a significant increase in F(2)-isoprostanes (F(2)-IsoPs), lipid peroxidation biomarkers of oxidative damage, in astrocyte cultures treated with 5 or 10 microM MeHg for 1 or 6 h. Consistent with this observation, MeHg induced a concentration-dependant reduction in the inner mitochondrial membrane potential (DeltaPsi(m)), as assessed by the potentiometric dye, tetramethylrhodamine ethyl ester (TMRE). Our results demonstrate that DeltaPsi(m) is a very sensitive endpoint for MeHg toxicity, since significant reductions were observed after only 1 h exposure to concentrations of MeHg as low as 1 microM. MeHg pretreatment (1, 5 and 10 microM) for 30 min also inhibited the net uptake of glutamine ((3)H-glutamine) measured at 1 min and 5 min. Expression of the mRNA coding the glutamine transporters, SNAT3/SN1 and ASCT2, was inhibited only at the highest (10 microM) MeHg concentration, suggesting that the reduction in glutamine uptake observed after 30 min treatment with lower concentrations of MeHg (1 and 5 microM) was not due to inhibition of transcription. Taken together, these studies demonstrate that MeHg exposure is associated with increased mitochondrial membrane permeability, alterations in glutamine/glutamate cycling, increased ROS formation and consequent oxidative injury. Ultimately, MeHg initiates multiple additive or synergistic disruptive mechanisms that lead to cellular dysfunction and cell death.


Assuntos
Astrócitos/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Glutamina/metabolismo , Intoxicação do Sistema Nervoso por Mercúrio/metabolismo , Compostos de Metilmercúrio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Astrócitos/patologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Relação Dose-Resposta a Droga , Ácido Glutâmico/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Intoxicação do Sistema Nervoso por Mercúrio/fisiopatologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/patologia , Estresse Oxidativo/fisiologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-12058187

RESUMO

Previous studies showed that the gene argS encoding the arginyl-tRNA synthetase(ArgRS) from Escherichia coli(E.coli), was overexpressed 1 000 folds in the E.coli transformant TG1/pUC-argS, while the gene leuS, encoding the leucyl-tRNA synthetase(LeuRS) from E.coli, was only overproduced 35-fold in the same case. To investigate why the expression of these two aminoacyl-tRNA synthetase genes is so different, a fused gene (termed parg-leuS) was constructed by replacement of the 5' flanking region of leuS to 5' flanking region of argS. In the E.coli transformant TG1/pUC-parg-leuS, the activity of LeuRS was only improved 8.5-fold, which was much lower than that of the transformant harboring the recombinant plasmid pUC18-leuS or pKK-leuS. However, by RNA dot hybridization the amount of mRNA produced in the transcription of parg-leuS was about 5 times than that of the wild type leuS, and was similar to that of pKK-leuS, suggesting that the promoter of argS is very strong. Analysis of the secondary structure around the initiation codon among three mRNAs showed that the secondary structure of the mRNA from parg-leuS was the strongest of the three mRNAs. From the results, it could be deduced that expression of the fused gene parg-leuS might be controlled at the translational level and the strong secondary structure of this mRNA may hinder translation initiation and result in a low translation efficiency.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...