Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 30(50): 16938-48, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159964

RESUMO

An increasing body of evidence points to a key role of endoplasmic reticulum (ER) stress in acute and chronic neurodegenerative conditions. Extensive ER stress can trigger neuronal apoptosis, but the signaling pathways that regulate this cell death remain unclear. In the present study, we demonstrate that PUMA, a Bcl-2 homology 3 (BH3)-only member of the Bcl-2 family, is transcriptionally activated in cortical neurons by ER stress and is essential for ER-stress-induced cell death. PUMA is known to be a key transcriptional target of p53, but we have found that ER stress triggers PUMA induction and cell death through a p53-independent mechanism mediated by the ER-stress-inducible transcription factor ATF4 (activating transcription factor 4). Specifically, we demonstrate that ectopic expression of ATF4 sensitizes mouse cortical neurons to ER-stress-induced apoptosis and that ATF4-deficient neurons exhibit markedly reduced levels of PUMA expression and cell death. However, chromatin immunoprecipitation experiments suggest that ATF4 does not directly regulate the PUMA promoter. Rather, we found that ATF4 induces expression of the transcription factor CHOP (C/EBP homologous protein) and that CHOP in turn activates PUMA induction. Specifically, we demonstrate that CHOP binds to the PUMA promoter during ER stress and that CHOP knockdown attenuates PUMA induction and neuronal apoptosis. In summary, we have identified a key signaling pathway in ER-stress-induced neuronal death involving ATF4-CHOP-mediated transactivation of the proapoptotic Bcl-2 family member PUMA. We propose that this pathway may be an important therapeutic target relevant to a number of neurodegenerative conditions.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/genética , Retículo Endoplasmático/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Estresse Fisiológico/genética , Fator de Transcrição CHOP/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Fator 4 Ativador da Transcrição/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/genética , Técnicas de Cultura de Células , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estresse Fisiológico/fisiologia , Tapsigargina/farmacologia , Fator de Transcrição CHOP/genética , Transfecção/métodos , Proteínas Supressoras de Tumor/genética , Tunicamicina/farmacologia
2.
J Neurosci ; 26(34): 8819-28, 2006 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-16928871

RESUMO

Cell cycle regulators appear to play a paradoxical role in neuronal death. We have shown previously that cyclin-dependent kinases (CDKs), along with their downstream effectors, Rb (retinoblastoma) and E2F/DP1 (E2 promoter binding factor/deleted in polyposis 1), regulate neuronal death evoked by the DNA damaging agent camptothecin. However, the mechanism by which CDKs are activated in this model is unclear. The cell division cycle 25A (Cdc25A) phosphatase is a critical regulator of cell cycle CDKs in proliferating cells. In cortical neurons, we presently show that expression of Cdc25A promotes death even in the absence of DNA damage. Importantly, Cdc25A activity is rapidly increased during DNA damage treatment. Inhibition of Cdc25A blocks death and reduces cyclin D1-associated kinase activity and Rb phosphorylation. This indicates that endogenous Cdc25A activity is important for regulation of cell cycle-mediated neuronal death. We also examined how Cdc25A activity is regulated after DNA damage. Cultured embryonic cortical neurons have a significant basal activity of checkpoint kinase 1 (Chk1), a kinase that regulates cell cycle arrest. During camptothecin treatment of neurons, this activity is rapidly downregulated with a concomitant increase in Cdc25A activity. Importantly, expression of wild-type Chk1, but not kinase-dead Chk1, inhibits the camptothecin-induced increase in Cdc25A activity. In addition, Chk1 expression also promotes survival in the presence of the DNA-damaging agent. Together, our data suggest that a Chk1/Cdc25A activity participates in activation of a cell cycle pathway-mediated death signal in neurons. These data also define how a proliferative signal may be abnormally activated in a postmitotic environment.


Assuntos
Apoptose/fisiologia , Camptotecina/farmacologia , Ciclo Celular/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Proteínas Quinases/metabolismo , Fosfatases cdc25/metabolismo , Animais , Células Cultivadas , Quinase 1 do Ponto de Checagem , Quinases Ciclina-Dependentes/metabolismo , Dano ao DNA , Camundongos , Camundongos Endogâmicos , Neurônios/efeitos dos fármacos , Proteínas Quinases/fisiologia , Interferência de RNA , Fosfatases cdc25/antagonistas & inibidores , Fosfatases cdc25/genética
3.
J Neurochem ; 96(2): 489-99, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16336220

RESUMO

Mechanical transection of the nigrostriatal dopamine pathway at the medial forebrain bundle (MFB) results in the delayed degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). We have previously demonstrated that c-Jun activation is an obligate component of neuronal death in this model. Here we identified the small GTPase, cdc42, and mixed lineage kinases (MLKs) as upstream factors regulating neuronal loss and activation of c-Jun following MFB axotomy. Adenovirus-mediated expression of a dominant-negative form of cdc42 in nigral neurons blocked MFB axotomy-induced activation (phosphorylation) of MAP kinase kinase 4 (MKK4) and c-Jun, resulting in attenuation of SNpc neuronal death. Pharmacological inhibition of MLKs, MKK4-activating kinases, significantly reduced the phosphorylation of c-Jun and abrogated dopaminergic neuronal degeneration following MFB axotomy. Taken together, these findings suggest that death of nigral dopaminergic neurons following axotomy can be attenuated by targeting cell signaling events upstream of c-Jun N-terminal mitogen-activated protein kinase/c-Jun.


Assuntos
Axotomia , Dopamina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , Feixe Prosencefálico Mediano/fisiopatologia , Neurônios/fisiologia , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Animais , Morte Celular , Inibidores Enzimáticos/farmacologia , Marcação de Genes , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase Quinases/genética , Masculino , Feixe Prosencefálico Mediano/patologia , Degeneração Neural/fisiopatologia , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Substância Negra/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
4.
Proc Natl Acad Sci U S A ; 102(41): 14611-6, 2005 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-16195385

RESUMO

IFN-gamma induction of the CIITA (class II transactivator) promoter (pIV) requires Brahma-related gene 1 (BRG1), a chromatin-remodeling enzyme. However, the events that lead to pIV activation are only partially understood, and the point at which BRG1 acts is unknown. The first IFN-gamma-induced event triggers nuclear translocation of STAT1 (signal transducer and activator of transcription 1), which binds IFN-gamma-responsive promoters. BRG1 is recruited after activator binding at several other inducible loci, and STAT family members are known to bind BRG1, suggesting that BRG1 might act downstream of STAT1. Here, we delineate a comprehensive view of factor assembly and detailed histone modifications at pIV and show that all events, even STAT1 binding, require BRG1 at CIITA pIV and other IFN-gamma target promoters. Recruitment of IFN-stimulated gene factor-3 (ISGF3) [STAT1/STAT2/IFN regulatory factor 9 (IRF9)] to several IFN-alpha-responsive promoters is also BRG1-dependent. In contrast, constitutive BRG1 association at IFN targets is STAT1-independent. Furthermore, BRG1 is required for IFN-induced restriction enzyme and DNase I accessibility at promoters. Thus, BRG1 has an apical role in cytokine-induced promoter assembly, acting upstream of STAT complexes at multiple IFN target loci.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Transativadores/genética , Fatores de Transcrição/metabolismo , Adenoviridae , Western Blotting , Montagem e Desmontagem da Cromatina/genética , Imunoprecipitação da Cromatina , DNA Helicases , Vetores Genéticos , Células HeLa , Humanos , Fator Gênico 3 Estimulado por Interferon/metabolismo , Interferon gama/metabolismo , Reação em Cadeia da Polimerase , Mapeamento por Restrição , Fator de Transcrição STAT1/metabolismo
5.
J Neurosci ; 25(39): 8954-66, 2005 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-16192386

RESUMO

Cyclin-dependent kinase 5 (cdk5) is a member of the cyclin-dependent kinase family whose activity is localized mainly to postmitotic neurons attributable to the selective expression of its activating partners p35 and p39. Deregulation of cdk5, as a result of calpain cleavage of p35 to a smaller p25 form, has been suggested to be a central component of neuronal death underlying numerous neurodegenerative diseases. However, the relevance of cdk5 in apoptotic death that relies on the mitochondrial pathway is unknown. Furthermore, evidence that cdk5 can also promote neuronal survival has necessitated a more complex understanding of cdk5 in the control of neuronal fate. Here we explore each of these issues using apoptotic and excitotoxic death models. We find that apoptotic death induced by the DNA-damaging agent camptothecin is associated with early transcription-mediated loss of p35 and with late production of p25 that is dependent on Bax, Apaf1, and caspases. In contrast, during excitotoxic death induced by glutamate, neurons rapidly produce p25 independent of the mitochondrial pathway. Analysis of the localization of p35 and p25 revealed that p35 is mainly cytoplasmic, whereas p25 accumulates selectively in the nucleus. By targeting a dominant-negative cdk5 to either the cytoplasm or nucleus, we show that cdk5 has a death-promoting activity within the nucleus and that this activity is required in excitotoxic death but not apoptotic death. Moreover, we also find that cdk5 contributes to pro-survival signaling selectively within the cytoplasm, and manipulation of this signal can modify death induced by both excitotoxicity and DNA damage.


Assuntos
Apoptose/fisiologia , Núcleo Celular/metabolismo , Quinase 5 Dependente de Ciclina/fisiologia , Citoplasma/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurotoxinas/farmacologia , Animais , Caspases/metabolismo , Morte Celular/fisiologia , Linhagem Celular , Dano ao DNA/fisiologia , Ativação Enzimática , Ácido Glutâmico/farmacologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Fosfotransferases
6.
J Neurosci ; 24(44): 10003-12, 2004 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-15525786

RESUMO

The p53 tumor suppressor gene has been implicated in the regulation of apoptosis in a number of different neuronal death paradigms. Because of the importance of p53 in neuronal injury, we questioned the mechanism underlying p53-mediated apoptosis in neurons. Using adenoviral-mediated gene delivery, reconstitution experiments, and mice carrying a knock-in mutation in the endogenous p53 gene, we show that the transactivation function of p53 is essential to induce neuronal cell death. Although p53 possesses two transactivation domains that can activate p53 targets independently, we demonstrate that the first activation domain (ADI) is required to drive apoptosis after neuronal injury. Furthermore, the BH3-only proteins Noxa and PUMA exhibit differential regulation by the two transactivation domains. Here, we show that Noxa can be induced by either activation domain, whereas PUMA induction requires both activation domains to be intact. Unlike Noxa, the upregulation of PUMA alone is sufficient to induce neuronal cell death. We demonstrate, therefore, that the first transactivation domain of p53 is indispensable for the induction of neuronal cell death.


Assuntos
Apoptose/fisiologia , Neurônios/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Motivos de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose , Células Cultivadas , Dano ao DNA/fisiologia , Camundongos , Camundongos Knockout , Mutação Puntual , Estrutura Terciária de Proteína , Ativação Transcricional/fisiologia , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/fisiologia , Regulação para Cima
7.
J Biol Chem ; 279(27): 28706-14, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15105421

RESUMO

The p53 tumor suppressor gene is believed to play an important role in neuronal cell death in acute neurological disease and in neurodegeneration. The p53 signaling cascade is complex, and the mechanism by which p53 induces apoptosis is cell type-dependent. Using DNA microarray analysis, we have found a striking induction of the proapoptotic gene, SIVA. SIVA is a proapoptotic protein containing a death domain and interacts with members of the tumor necrosis factor receptor family as well as anti-apoptotic Bcl-2 family proteins. SIVA is induced following direct p53 gene delivery, treatment with a DNA-damaging agent camptothecin, and stroke injury in vivo. SIVA up-regulation is sufficient to initiate the apoptotic cascade in neurons. Through isolation and analysis of the SIVA promoter, we have identified response elements for both p53 and E2F1. Like p53, E2F1 is another tumor suppressor gene involved in the regulation of apoptosis, including neuronal injury models. We have identified E2F consensus sites in the promoter region, whereas p53 recognition sequences were found in intron1. Sequence analysis has shown that these consensus sites are also conserved between mouse and human SIVA genes. Electrophoretic mobility shift assays reveal that both transcription factors are capable of binding to putative consensus sites, and luciferase reporter assays reveal that E2F1 and p53 can activate transcription from the SIVA promoter. Here, we report that the proapoptotic gene, SIVA, which functions in a broad spectrum of cell types, is a direct transcriptional target for both tumor suppressors, p53 and E2F1.


Assuntos
Apoptose , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adenoviridae/genética , Animais , Proteínas Reguladoras de Apoptose , Sequência de Bases , Sítios de Ligação , Western Blotting , Camptotecina/farmacologia , Células Cultivadas , Dano ao DNA , DNA Complementar/metabolismo , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Genes Reporter , Humanos , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Dados de Sequência Molecular , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Regulação para Cima
8.
J Cell Biol ; 158(3): 507-17, 2002 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12147675

RESUMO

Caspase-independent death mechanisms have been shown to execute apoptosis in many types of neuronal injury. P53 has been identified as a key regulator of neuronal cell death after acute injury such as DNA damage, ischemia, and excitotoxicity. Here, we demonstrate that p53 can induce neuronal cell death via a caspase-mediated process activated by apoptotic activating factor-1 (Apaf1) and via a delayed onset caspase-independent mechanism. In contrast to wild-type cells, Apaf1-deficient neurons exhibit delayed DNA fragmentation and only peripheral chromatin condensation. More importantly, we demonstrate that apoptosis-inducing factor (AIF) is an important factor involved in the regulation of this caspase-independent neuronal cell death. Immunofluorescence studies demonstrate that AIF is released from the mitochondria by a mechanism distinct from that of cytochrome-c in neurons undergoing p53-mediated cell death. The Bcl-2 family regulates this release of AIF and subsequent caspase-independent cell death. In addition, we show that enforced expression of AIF can induce neuronal cell death in a Bax- and caspase-independent manner. Microinjection of neutralizing antibodies against AIF significantly decreased injury-induced neuronal cell death in Apaf1-deficient neurons, indicating its importance in caspase-independent apoptosis. Taken together, our results suggest that AIF may be an important therapeutic target for the treatment of neuronal injury.


Assuntos
Apoptose/fisiologia , Encéfalo/embriologia , Caspases/metabolismo , Flavoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Doenças Neurodegenerativas/enzimologia , Neurônios/enzimologia , Proteínas/metabolismo , Animais , Anticorpos/farmacologia , Fator de Indução de Apoptose , Fator Apoptótico 1 Ativador de Proteases , Encéfalo/citologia , Encéfalo/enzimologia , Camptotecina/farmacologia , Inibidores de Caspase , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Inibidores Enzimáticos/farmacologia , Flavoproteínas/genética , Frequência do Gene/fisiologia , Marcação In Situ das Extremidades Cortadas , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/fisiopatologia , Neurônios/citologia , Proteínas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transfecção , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA