Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Cardiovasc Res ; 73(1): 48-56, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17107669

RESUMO

OBJECTIVE: Puma (p53-upregulated modulator of apoptosis), a proapoptotic BH3-only member of the Bcl-2 protein family, has been implicated in the pathomechanism of several diseases, including cancer, AIDS, and ischemic brain disease. We have recently shown that Puma is required for cardiac cell death upon ischemia/reperfusion of mouse hearts. Since ischemia/reperfusion is also associated with endoplasmic reticulum (ER) stress, in the present study we investigated whether Puma contributes to the ER stress-dependent component of cardiomyocyte apoptosis. METHODS: Primary cultures of rat and mouse neonatal cardiomyocytes were treated with 3 muM thapsigargin or 100 ng mL(-1) tunicamycin. Puma levels were suppressed by adenoviral delivery of shRNA or targeted deletion of the puma gene. Puma expression was detected by RT-PCR and Western blotting. Apoptosis was assessed by TUNEL assay, caspase-3 cleavage, and cytochrome c release. RESULTS: We have shown that in rat neonatal cardiac myocytes, thapsigargin or tunicamycin treatment led to ER-stress, transcriptional upregulation of Puma, and apoptosis. Most importantly, cardiac myocytes acquired resistance to ER stress-induced apoptosis if Puma expression was downregulated by adenoviral delivery of shRNA or eliminated by targeted deletion in knockout mice. CONCLUSION: Taken together, our data indicate that Puma is a critical component of ER stress-induced apoptosis in cardiac myocytes, and inhibition of Puma activity may be used to treat cardiac infarcts or prevent heart failure by blocking ER stress-induced apoptosis.


Assuntos
Retículo Endoplasmático/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Proteínas Reguladoras de Apoptose , Biomarcadores/análise , Western Blotting/métodos , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Caspase 3/análise , Linhagem Celular , Células Cultivadas , Citocromos c/análise , Retículo Endoplasmático/efeitos dos fármacos , Deleção de Genes , Expressão Gênica , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas c-bcl-2 , Interferência de RNA , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tapsigargina/farmacologia , Transfecção , Proteína Supressora de Tumor p53 , Proteínas Supressoras de Tumor/genética , Tunicamicina/farmacologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-17896961

RESUMO

The endoplasmic reticulum (ER) is a multifunctional organelle responsible for the synthesis and folding of proteins as well as calcium storage and signaling. Perturbations of ER function cause ER stress leading to the unfolded protein response (UPR), which includes inhibition of protein synthesis, protein refolding and clearance of misfolded proteins. The UPR aims at restoring cellular homeostasis, however, prolonged ER stress can trigger apoptosis. ER stress-induced apoptosis has been implicated in the pathogenesis of various diseases such as brain ischemia/reperfusion, neurodegeneration, diabetes and, most recently, myocardial infarction and heart failure. Initial events leading to UPR and apoptosis in the heart include protein oxidation and disturbed calcium handling upon ischemia/reperfusion, and forced protein synthesis during cardiac hypertrophy. While XBP-1 and ATF6-mediated induction of ER chaperones seems to protect the heart from ischemia/reperfusion injury, the PERK/ATF4/CHOP branch of the UPR might transmit proapoptotic signals. The precise mechanism of ER stress-induced cardiomyocyte apoptosis remains elusive, however, recent data suggest that the mitochondrial apoptotic machinery is recruited through the upregulation of Puma, a proapoptotic member of the Bcl-2 family. Importantly, suppression of Puma activity prevented both ER stress and ischemia/reperfusion-induced cardiomyocyte loss, highlighting the ER stress pathways as potential therapeutic targets in cardiovascular diseases.


Assuntos
Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Cardiopatias/tratamento farmacológico , Estresse Fisiológico/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Retículo Endoplasmático/química , Cardiopatias/fisiopatologia , Humanos , Dobramento de Proteína , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico/fisiopatologia
3.
J Biol Chem ; 281(6): 3679-89, 2006 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-16339144

RESUMO

MDM2 is an E3 ubiquitin ligase that regulates the proteasomal degradation and activity of proteins involved in cell growth and apoptosis, including the tumor suppressors p53 and retinoblastoma and the transcription factor E2F1. Although the effect of several MDM2 targets on cardiomyocyte survival and hypertrophy has already been investigated, the role of MDM2 in these processes has not yet been established. We have, therefore, analyzed the effect of overexpression as well as inhibition of MDM2 on cardiac ischemia/reperfusion injury and hypertrophy. Here we show that isolated cardiac myocytes overexpressing MDM2 acquired resistance to hypoxia/reoxygenation-induced cell death. Conversely, inactivation of MDM2 by a peptide inhibitor resulted in elevated p53 levels and promoted hypoxia/reoxygenation-induced apoptosis. Consistent with this, decreased expression of MDM2 in a genetic mouse model was accompanied by reduced functional recovery of the left ventricles determined with the Langendorff ex vivo model of ischemia/reperfusion. In contrast to cell survival, cell hypertrophy induced by the alpha-agonists phenylephrine or endothelin-1 was inhibited by MDM2 overexpression. Collectively, our studies indicate that MDM2 promotes survival and attenuates hypertrophy of cardiac myocytes. This differential regulation of cell growth and cell survival is unique, because most other survival factors are prohypertrophic. MDM2, therefore, might be a potential therapeutic target to down-regulate both cell death and pathologic hypertrophy during remodeling upon cardiac infarction. In addition, our data also suggest that cancer treatments with MDM2 inhibitors to reactivate p53 may have adverse cardiac side effects by promoting cardiomyocyte death.


Assuntos
Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Transformada , Proliferação de Células , Sobrevivência Celular , Endotelina-1/metabolismo , Hipóxia , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/patologia , Peptídeos/química , Perfusão , Fenilefrina/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo
4.
Am J Physiol Heart Circ Physiol ; 291(1): H52-60, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16399862

RESUMO

The p53-upregulated modulator of apoptosis (Puma), a BH3-only member of the Bcl-2 protein family, is required for p53-dependent and -independent forms of apoptosis and has been implicated in the pathomechanism of several diseases, including cancer, acquired immunodeficiency syndrome, and ischemic brain disease. The role of Puma in cardiomyocyte death, however, has not been analyzed. On the basis of the ability of Puma to integrate diverse cell death stimuli, we hypothesized that Puma might be critical for cardiomyocyte death upon ischemia-reperfusion (I/R) of the heart. Here we show that hypoxia-reoxygenation of isolated cardiomyocytes led to an increase in Puma mRNA and protein levels. Moreover, if Puma was delivered by an adenoviral construct, cardiomyocytes died by apoptosis. Under ATP-depleted conditions, however, Puma overexpression primarily induced necrosis, suggesting that Puma is involved in the development of both types of cell death. Consistent with these findings, targeted deletion of Puma in a mouse model attenuated both apoptosis and necrosis. When the Langendorff ex vivo I/R model was used, infarcts were approximately 50% smaller in Puma(-/-) than in wild-type mice. As a result, after I/R, cardiac function was significantly better preserved in Puma(-/-) mice than in their wild-type littermates. Our study thus establishes Puma as an essential mediator of cardiomyocyte death upon I/R injury and offers a novel therapeutic target to limit cell loss in ischemic heart disease.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Marcação de Genes/métodos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Disfunção Ventricular Esquerda/prevenção & controle , Disfunção Ventricular Esquerda/fisiopatologia , Animais , Animais Recém-Nascidos , Apoptose , Proteínas Reguladoras de Apoptose/genética , Células Cultivadas , Sistemas de Liberação de Medicamentos/métodos , Deleção de Genes , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/terapia , Resultado do Tratamento , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA