Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 288(14): 9696-9709, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23400779

RESUMO

Excitotoxicity resulting from overstimulation of glutamate receptors is a major cause of neuronal death in cerebral ischemic stroke. The overstimulated ionotropic glutamate receptors exert their neurotoxic effects in part by overactivation of calpains, which induce neuronal death by catalyzing limited proteolysis of specific cellular proteins. Here, we report that in cultured cortical neurons and in vivo in a rat model of focal ischemic stroke, the tyrosine kinase Src is cleaved by calpains at a site in the N-terminal unique domain. This generates a truncated Src fragment of ~52 kDa, which we localized predominantly to the cytosol. A cell membrane-permeable fusion peptide derived from the unique domain of Src prevents calpain from cleaving Src in neurons and protects against excitotoxic neuronal death. To explore the role of the truncated Src fragment in neuronal death, we expressed a recombinant truncated Src fragment in cultured neurons and examined how it affects neuronal survival. Expression of this fragment, which lacks the myristoylation motif and unique domain, was sufficient to induce neuronal death. Furthermore, inactivation of the prosurvival kinase Akt is a key step in its neurotoxic signaling pathway. Because Src maintains neuronal survival, our results implicate calpain cleavage as a molecular switch converting Src from a promoter of cell survival to a mediator of neuronal death in excitotoxicity. Besides unveiling a new pathological action of Src, our discovery of the neurotoxic action of the truncated Src fragment suggests new therapeutic strategies with the potential to minimize brain damage in ischemic stroke.


Assuntos
Calpaína/química , Regulação Enzimológica da Expressão Gênica , Neurônios/metabolismo , Quinases da Família src/química , Animais , Isquemia Encefálica/patologia , Calpaína/metabolismo , Morte Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Lentivirus/genética , Masculino , Modelos Biológicos , Mutação , Peptídeos/química , Ratos , Ratos Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/patologia , Quinases da Família src/metabolismo
2.
J Neurochem ; 109 Suppl 1: 198-206, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19393028

RESUMO

GABAergic striatal neurons are compromised in basal ganglia pathologies and we analysed how insult nature determined their patterns of injury and recruitment of the intrinsic mitochondrial pathway during programmed cell death (PCD). Stressors affecting targets implicated in striatal neurodegeneration [3-morpholinylsydnoneimine (SIN-1), 3-nitropropionic acid (3-NP), NMDA, 3,5-dihydroxyphenylglycine (DHPG), and staurosporine (STS)] were compared in cultured GABAergic neurons from murine striatum by analyzing the progression of injury and its correlation with mitochondrial involvement, the redistribution of intermembrane space (IMS) proteins, and patterns of protease activation. Stressors produced PCD exhibiting slow-onset kinetics with time-dependent annexin-V labeling and eventual DNA fragmentation. IMS proteins including cytochrome c were differentially distributed, although stressors except STS produced early redistribution of apoptosis-inducing factor and Omi, suggestive of early recruitment of both caspase-dependent and caspase-independent signaling. In general, Bax mobilization to mitochondria appeared to promote IMS protein redistribution. Caspase 3 activation was prominent after STS, whereas NMDA and SIN-1 produced mainly calpain activation, and 3-NP and DHPG elicited a mixed profile of protease activation. PCD and redistribution of IMS proteins in striatal GABAergic neurons were canonical and insult-dependent, reflecting differential interplay between the caspase cascade and alternate cell death pathways.


Assuntos
Apoptose/fisiologia , Caspases/fisiologia , Mitocôndrias/fisiologia , Neostriado/fisiologia , Neurônios/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Calpaína/metabolismo , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Fragmentação do DNA , Ativação Enzimática/fisiologia , Feminino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mitocôndrias/efeitos dos fármacos , Neostriado/citologia , Neostriado/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Gravidez
3.
Brain Res ; 1594: 1-14, 2015 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-25451123

RESUMO

BACKGROUND: Src-family kinases (SFKs) are involved in neuronal survival and their aberrant regulation contributes to neuronal death. However, how they control neuronal survival and death remains unclear. OBJECTIVE: To define the effect of inhibition of Src activity and expression on neuronal survival. RESULTS: In agreement with our previous findings, we demonstrated that Src was cleaved by calpain to form a 52-kDa truncated fragment in neurons undergoing excitotoxic cell death, and expression of the recombinant truncated Src fragment induced neuronal death. The data confirm that the neurotoxic signaling pathways are intact in the neurons we used for our study. To define the functional role of neuronal SFKs, we treated these neurons with SFK inhibitors and discovered that the treatment induced cell death, suggesting that the catalytic activity of one or more of the neuronal SFKs is critical to neuronal survival. Using small hairpin RNAs that suppress Src expression, we demonstrated that Src is indispensable to neuronal survival. Additionally, we found that neuronal death induced by expression of the neurotoxic truncated Src mutant, treatment of SFK inhibitors or knock-down of Src expression caused inhibition of the neuroprotective protein kinases Erk1/2, or Akt. CONCLUSIONS: Src is critical to both neuronal survival and death. Intact Src sustains neuronal survival. However, in the excitotoxic condition, calpain cleavage of Src generates a neurotoxic truncated Src fragment. Both intact Src and the neurotoxic truncated Src fragment exert their biological actions by controlling the activities of neuroprotective protein kinases.


Assuntos
Neurônios/enzimologia , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo , Animais , Western Blotting , Calpaína/metabolismo , Sobrevivência Celular , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/metabolismo
4.
Eur J Pharmacol ; 633(1-3): 24-32, 2010 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-20153315

RESUMO

Tolerance to brain injury involves hypoxia-inducible factor-1 (HIF-1) and its target genes as the key pathway mediating a cascade of events including cell survival, energetics, and angiogenesis. In this study, we established the treatment paradigms for an in vitro model of tolerance to oxidative injury in primary astrocytic cultures and further examined the roles for the HIF-1 signalling cascade. Isolated murine astrocytes were preconditioned with sub-toxic concentrations of HIF-1 inducers and subsequently exposed to a H(2)O(2) insult, where changes in cell viability and protein expression were determined. Preconditioning with non-damaging concentrations of desferrioxamine (DFO) and ethyl-3,4-dihydroxybenzoate (EDHB) significantly improved cellular viability after H(2)O(2) injury treatment. Time course studies revealed that DFO and EDHB treatments alone induced sequential activation of HIF-1 signal transduction where nuclear HIF-1alpha protein accumulation was detected as early as 2h, followed by downstream upregulation of intracellular and released VEGF from 4h and 8h onwards, respectively. The protective effects of DFO and EDHB preconditioning against H(2)O(2) injury were abolished by co-treatment with cycloheximide, an inhibitor of protein synthesis. Importantly, when the anti-HIF-1 compound, 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) was used, the cytoprotection and VEGF accumulation produced by DFO and EDHB preconditioning were diminished. These results indicate the essential role of the HIF-1 pathway in our model of tolerance against oxidative injury in cultured astrocytes, and suggest roles for astrocytic HIF-1 expression and VEGF release which may influence the function of surrounding cells and vasculature during oxidative stress-related brain diseases.


Assuntos
Astrócitos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fator 1 Induzível por Hipóxia/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Cicloeximida/farmacologia , Citoproteção/efeitos dos fármacos , Desferroxamina/antagonistas & inibidores , Desferroxamina/farmacologia , Interações Medicamentosas , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/toxicidade , Hidroxibenzoatos/antagonistas & inibidores , Hidroxibenzoatos/farmacologia , Indazóis/farmacologia , Camundongos , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA