Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(15): 5969-74, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23530233

RESUMEN

Mitochondrial morphological dynamics affect the outcome of ischemic heart damage and pathogenesis. Recently, mitochondrial fission protein dynamin-related protein 1 (Drp1) has been identified as a mediator of mitochondrial morphological changes and cell death during cardiac ischemic injury. In this study, we report a unique relationship between Pim-1 activity and Drp1 regulation of mitochondrial morphology in cardiomyocytes challenged by ischemic stress. Transgenic hearts overexpressing cardiac Pim-1 display reduction of total Drp1 protein levels, increased phosphorylation of Drp1-(S637), and inhibition of Drp1 localization to the mitochondria. Consistent with these findings, adenoviral-induced Pim-1 neonatal rat cardiomyocytes (NRCMs) retain a reticular mitochondrial phenotype after simulated ischemia (sI) and decreased Drp1 mitochondrial sequestration. Interestingly, adenovirus Pim-dominant negative NRCMs show increased expression of Bcl-2 homology 3 (BH3)-only protein p53 up-regulated modulator of apoptosis (PUMA), which has been previously shown to induce Drp1 accumulation at mitochondria and increase sensitivity to apoptotic stimuli. Overexpression of the p53 up-regulated modulator of apoptosis-dominant negative adenovirus attenuates localization of Drp1 to mitochondria in adenovirus Pim-dominant negative NRCMs promotes reticular mitochondrial morphology and inhibits cell death during sI. Therefore, Pim-1 activity prevents Drp1 compartmentalization to the mitochondria and preserves reticular mitochondrial morphology in response to sI.


Asunto(s)
Dinaminas/metabolismo , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/fisiología , Adenoviridae/genética , Animales , Ratones , Ratones Transgénicos , Miocitos Cardíacos/citología , Fosforilación , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Ratas
2.
Circulation ; 124(1): 31-9, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21670227

RESUMEN

BACKGROUND: Puma (p53-upregulated modulator of apoptosis) is a proapoptotic Bcl-2 family protein that serves as a general sensor in response to pathological apoptotic stimuli. In previous work, we demonstrated that puma ablation protects the heart from reperfusion injury in a Langendorff setting. Consistent with this, downregulation of Puma in isolated cardiac myocytes prevented apoptosis induced by different proapoptotic agents. Here, we extended our research to investigate the role of Puma, a downstream mediator of p53, in the development of heart failure using Puma(-/-) mice. METHODS AND RESULTS: Mice underwent transverse aortic constriction, and the characteristics of cardiac remodeling were analyzed by echocardiography, histology, and gene expression at multiple time points after surgery. Four weeks after the operation, puma deletion attenuated pressure overload-induced apoptosis and fibrosis; however, it did not affect hypertrophy and angiogenesis and maintained functional performance (fractional shortening, 39% versus 25.2% in Puma(-/-) versus WT mice, respectively). Even at 12 weeks after transverse aortic constriction, Puma(-/-) mice displayed only slightly reduced contractility. In addition, transverse aortic constriction induced puma expression in a partially p53-dependent manner. To corroborate these findings, we studied another heart failure model in which heart-specific mdm4 deletion leads to p53 activation and dilated cardiomyopathy. In these mice, Puma was upregulated and its deletion rescued the cardiomyopathy phenotype. CONCLUSIONS: Our data indicate that Puma might be a critical component of the apoptotic signaling pathways that contribute to ventricular remodeling and heart failure. Therefore, Puma inactivation may serve as a preferential target to prevent heart failure induced by cellular stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/fisiología , Progresión de la Enfermedad , Insuficiencia Cardíaca/prevención & control , Corazón/fisiopatología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Animales , Aorta/fisiopatología , Proteínas Reguladoras de la Apoptosis/fisiología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/prevención & control , Modelos Animales de Enfermedad , Fibrosis/patología , Fibrosis/prevención & control , Eliminación de Gen , Insuficiencia Cardíaca/fisiopatología , Ratones , Ratones Noqueados , Miocardio/patología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/fisiología , Vasoconstricción
3.
Biochem Biophys Res Commun ; 380(3): 493-7, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19284993

RESUMEN

Recent studies on cardiac hypertrophy animal model suggest that inter-domain interactions within the ryanodine receptor (RyR2) become defective concomitant with the development of hypertrophy (e.g. de-stabilization of the interaction between N-terminal and central domains of RyR2; T. Oda, M. Yano, T. Yamamoto, T. Tokuhisa, S. Okuda, M. Doi, T. Ohkusa, Y. Ikeda, S. Kobayashi, N. Ikemoto, M. Matsuzaki, Defective regulation of inter-domain interactions within the ryanodine receptor plays a key role in the pathogenesis of heart failure, Circulation 111 (2005) 3400-3410). To determine if de-stabilization of the inter-domain interaction in fact causes hypertrophy, we introduced DPc10 (a peptide corresponding to the G(2460)-P(2495) region of RyR2, which is known to de-stabilize the N-terminal/central domain interaction) into rat neonatal cardiomyocytes by mediation of peptide carrier BioPORTER. After incubation for 24h the peptide induced hypertrophy, as evidenced by significant increase in cell size and [(3)H]leucine uptake. K201 or dantrolene, the reagents known to correct the de-stabilized inter-domain interaction to a normal mode, prevented the DPc10-induced hypertrophy. These results suggest that disruption of the normal N-terminal/central inter-domain interaction within the RyR2 is a causative mechanism of cardiomyocyte hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/patología , Células Cultivadas , Endotelina-1/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fragmentos de Péptidos/farmacología , Ratas , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/farmacología
4.
Methods Mol Biol ; 559: 293-311, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19609765

RESUMEN

Acute myocardial infarction represents the leading cause of morbidity and mortality in the western societies. Importantly, both apoptosis and necrosis of cardiomyocytes have been implicated in the pathomechanism of myocardial infarction. The simplest way to analyze apoptosis in cardiac cells is the application of isolated neonatal primary cardiac myocytes, in which ischemia/reperfusion can be mimicked in vitro by exposing them to hypoxia and serum starvation, followed by restored oxygen and serum conditions, referred to as hypoxia/reoxygenation. In this chapter, we describe protocols routinely applied in our lab for investigating cardiomyocyte apoptosis. In summary, a better understanding of the apoptotic pathways and their regulation in the heart will potentially yield novel therapeutic targets for cardiac infarction.


Asunto(s)
Apoptosis , Técnicas Citológicas , Miocitos Cardíacos/citología , Animales , Western Blotting/métodos , Caspasa 3/análisis , Caspasa 3/metabolismo , Fraccionamiento Celular/métodos , Hipoxia de la Célula , Membrana Celular/metabolismo , Células Cultivadas , Citocromos c/análisis , Citocromos c/metabolismo , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ/métodos , Ratones , Oxígeno/metabolismo , Ratas
5.
Cardiovasc Res ; 73(1): 48-56, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17107669

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Proteínas Reguladoras de la Apoptosis , Biomarcadores/análisis , Western Blotting/métodos , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Caspasa 3/análisis , Línea Celular , Células Cultivadas , Citocromos c/análisis , Retículo Endoplásmico/efectos de los fármacos , Eliminación de Gen , Expresión Génica , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-bcl-2 , Interferencia de ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tapsigargina/farmacología , Transfección , Proteína p53 Supresora de Tumor , Proteínas Supresoras de Tumor/genética , Tunicamicina/farmacología
6.
Artículo en Inglés | MEDLINE | ID: mdl-17896961

RESUMEN

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.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Fármacos Cardiovasculares/uso terapéutico , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/fisiología , Cardiopatías/tratamiento farmacológico , Estrés Fisiológico/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Retículo Endoplásmico/química , Cardiopatías/fisiopatología , Humanos , Pliegue de Proteína , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/fisiopatología
7.
J Biol Chem ; 281(6): 3679-89, 2006 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-16339144

RESUMEN

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.


Asunto(s)
Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Adenoviridae/genética , Animales , Animales Recién Nacidos , Antineoplásicos/farmacología , Apoptosis , Línea Celular Transformada , Proliferación Celular , Supervivencia Celular , Endotelina-1/metabolismo , Hipoxia , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Miocardio/patología , Péptidos/química , Perfusión , Fenilefrina/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
8.
Am J Physiol Heart Circ Physiol ; 291(1): H52-60, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16399862

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Marcación de Gen/métodos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas/metabolismo , Daño por Reperfusión/fisiopatología , Disfunción Ventricular Izquierda/prevención & control , Disfunción Ventricular Izquierda/fisiopatología , Animales , Animales Recién Nacidos , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Células Cultivadas , Sistemas de Liberación de Medicamentos/métodos , Eliminación de Gen , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Daño por Reperfusión/terapia , Resultado del Tratamiento , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/patología
9.
Mol Cell ; 20(4): 539-50, 2005 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-16307918

RESUMEN

The phosphoinositide 3-kinase (PI 3-K) signaling axis is intimately associated with deregulated cancer cell growth, primarily by promoting increased survival through Akt/PKB (protein kinase B). However, there is relatively little information on the role of Akt in cancer cell motility, a key phenotype of invasive carcinomas. Here we report that activation of Akt inhibits carcinoma migration and invasion of breast cancer cells. Conversely, downregulation of Akt using RNA interference increased migration and invasion. Akt blunts invasion by inhibiting the transcriptional activity of NFAT (nuclear factor of activated T cells). Specifically, signaling through Akt reduces NFAT expression levels due to ubiquitination and proteasomal degradation, mediated by the E3 ubiquitin ligase HDM2. These results indicate that while Akt can promote tumor progression through increased cell survival mechanisms, it can block breast cancer cell motility and invasion by a mechanism that depends, at least in part, on the NFAT transcription factor.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Inhibición de Migración Celular , Movimiento Celular/fisiología , Factores de Transcripción NFATC/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Células 3T3 NIH , Invasividad Neoplásica , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA