Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
3.
Cell Death Dis ; 4: e771, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23949220

RESUMO

Global cerebral ischemia/reperfusion (I/R) facilitates the activation of procaspase-3 and promotes apoptosis in hippocampus. But the mechanisms have remained uncharacterized. Protein S-nitrosylation and denitrosylation is an important reversible posttranslational modification, which is a common mechanism in signal transduction and affects numerous physiological and pathophysiological events. However, it is not known whether S-nitrosylation/denitrosylation modification of procaspase-3 serves as a component of apoptosis and cell death induced by cerebral I/R. Here we show that procaspase-3 is significantly denitrosylated and activated after I/R in rat hippocampus. NS102, a glutamate receptor 6 (GluR6) antagonist, can inhibit the denitrosylation of procaspase-3 and diminish the increased Fas ligand (FasL) and thioredoxin (Trx)-2 expression induced by cerebral I/R. Moreover, downregulation of FasL expression by antisense oligodeoxynucleotides inhibits the denitrosylation and activation of procaspase-3. Auranofin, a TrxR inhibitor or TrxR2 antisense oligodeoxynucleotide, has similar effects. In primary hippocampal cultures, Lentiviral-mediated knockdown of FasL and TrxR2 before the oxygen and glucose deprivation/reoxygenation further verifies that FasL and TrxR2 are involved in the denitrosylation of procaspase-3. In situ TUNEL staining and cresyl violet staining validate that inhibiting denitrosylation of procaspase-3 may exert neuroprotective effect on apoptosis and cell death induced by cerebral I/R in hippocampal CA1 pyramidal neurons. This is the first evidence that cerebral I/R mediates procaspase-3 denitrosylation and activation through GluR6-FasL-Trx2 pathway, which leads to neuronal apoptosis and cell death.


Assuntos
Isquemia Encefálica/enzimologia , Caspase 3/metabolismo , Proteína Ligante Fas/metabolismo , Receptores de Ácido Caínico/metabolismo , Traumatismo por Reperfusão/enzimologia , Tiorredoxinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glucose/deficiência , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Hipocampo/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Nitrosação/efeitos dos fármacos , Oxigênio/farmacologia , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Tiorredoxina Redutase 1/metabolismo , Receptor de GluK2 Cainato
4.
Neuroscience ; 229: 36-48, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23137546

RESUMO

Apoptosis signal-regulating kinase 1 (ASK1) is a general mediator of cell death in response to a variety of stimuli, including reactive oxygen species, tumor necrosis factor α, lipopolysaccharide, endoplasmic reticulum stress, calcium influx and ischemia. Here we reported ASK1 was activated by nitric oxide (NO) through S-nitrosylation during cerebral ischemia-reperfusion. The reagents that abrogate neuronal nitric oxide synthase (nNOS) activity such as nNOS inhibitor 7NI and N-methyl-D-aspartate receptor antagonist MK801 prevented ASK1 activation via decreasing ASK1 S-nitrosylation. In HEK293 cells, over-expressed ASK1 could be S-nitrosylated by both exogenous and endogenous NO and Cys869 was identified as the site of ASK1 S-nitrosylation. S-nitrosylation increased the level of ASK1 phosphorylation at Thr845, which represents ASK1 activation. Our results further confirmed that S-nitrosylation led to the increment of ASK1 dimerization. S-nitrosylation of ASK1 also activated the downstream JNK signaling and JNK-mediated nucleic pathway. The exogenous NO (SNP and GSNO) reversed the effect of endogenous NO by suppressing S-nitrosylation of ASK1 and exerted neuroprotection during ischemia-reperfusion. These results suggest that inhibiting ASK1 S-nitrosylation may be a novel approach for stroke therapy.


Assuntos
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Óxido Nítrico/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Isquemia Encefálica/patologia , Inibidores Enzimáticos/farmacologia , Células HEK293 , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , MAP Quinase Quinase Quinase 5/genética , Masculino , Mutagênese Sítio-Dirigida , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/metabolismo , Ratos , Ratos Sprague-Dawley , Reperfusão , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Neuroscience ; 156(3): 483-97, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18773943

RESUMO

It is well documented that heat-shock protein (hsp90) plays an essential role in maintaining stability and activity of its clients. Recent studies have shown that geldanamycin (GA), an inhibitor of hsp90, could decrease the protein of mixed-lineage kinase (MLK) 3 and activate Akt; our previous research documented that MLK3 and Akt and subsequent c-Jun N-terminal kinase (JNK) were involved in neuronal cell death in ischemic brain injury. Here, we investigated whether GA could decrease the protein of MLK3 and activate Akt in rat four-vessel occlusion ischemic model. Our results showed that global cerebral ischemia followed by reperfusion could enhance the association of hsp90 with MLK3, the association of hsp90 with Src, and JNK3 activation. As a result, GA decreased the protein of MLK3 and down-regulated JNK activation. On the other hand, Src kinase was activated and phosphorylated Cbl, which then recruited the p85 subunit of phosphatidylinositol 3-kinase (PI-3K), resulting in PI-3K activation, and as a consequence increased Akt activation, which inhibited ASK1 activation and down-regulated JNK3 activation. In summary, our results indicated that GA showed a dual inhibitory role on JNK3 activation and exerted strong neuroprotection in vivo and in vitro, which provides a new possible approach for stroke therapy.


Assuntos
Benzoquinonas/farmacologia , Lesões Encefálicas/enzimologia , Inibidores Enzimáticos/farmacologia , Lactamas Macrocíclicas/farmacologia , MAP Quinase Quinase Quinase 5/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Oncogênica v-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Células Cultivadas , Cromonas/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Glucose/deficiência , Hipocampo/citologia , Infarto da Artéria Cerebral Média/complicações , Masculino , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Oxigênio/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Neuroscience ; 139(2): 609-27, 2006 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-16504411

RESUMO

Our previous studies and the others have strongly suggested that c-Jun N-terminal kinase (JNK) signaling pathway plays a critical role in ischemic brain injury. Here we reported that Tat-JNK binding domain (JBD) of JNK-interacting protein-1 (JIP-1), a smaller 11-mer peptide corresponding to residues 153-163 of murine JIP-1 conjugated to Tat peptide, perturbed the assembly of JIP-1-JNK3 complexes, thus inhibiting the activation of JNK3 induced by ischemia/reperfusion in the vulnerable hippocampal CA1 subregion. As a result, Tat-JBD diminished the increased phosphorylation of c-Jun (a nuclear substrate of JNK) and the increased expression of Fas ligand induced by ischemia/reperfusion in the vulnerable hippocampal CA1 subregion. At the same time, through inhibiting phosphorylation of Bcl-2 (a cytosolic target of JNK) and the release of Bax from Bcl-2/Bax dimers, Tat-JBD attenuated Bax translocation to mitochondria and the release of cytochrome c induced by ischemia/reperfusion. Furthermore, the activation of caspase3 and hydrolyzation of poly-ADP-ribose-polymerase induced by brain ischemia/reperfusion were also significantly suppressed by preinfusion of the peptide Tat-JBD. Importantly, Tat-JBD showed neuroprotective effects on ischemic brain damage in vivo, and administration of the peptide after ischemia also achieved the same effects as preinfusion of the peptide did. Thus, our findings imply that Tat-JBD induced neuroprotection against ischemia/reperfusion in rat hippocampal CA1 region via inhibiting nuclear and non-nuclear pathways of JNK signaling. Taken together, these results indicate that Tat-JBD peptide provides a promising therapeutic approach for ischemic brain injury.


Assuntos
Lesões Encefálicas/prevenção & controle , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Peptídeos/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Lesões Encefálicas/etiologia , Isquemia Encefálica/complicações , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Proteína Ligante Fas , Expressão Gênica/efeitos dos fármacos , Produtos do Gene tat/genética , Produtos do Gene tat/metabolismo , Hipocampo/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/química , Masculino , Glicoproteínas de Membrana/metabolismo , Neurônios/efeitos dos fármacos , Peptídeos/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Transdução de Sinais/fisiologia , Frações Subcelulares/efeitos dos fármacos , Fatores de Necrose Tumoral/metabolismo , Proteína X Associada a bcl-2/metabolismo , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA