Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Circulation ; 122(4): 361-9, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20625112

RESUMEN

BACKGROUND: Apoptosis may contribute to the development of heart failure, but the role of apoptotic signaling initiated by the endoplasmic reticulum in this condition has not been well clarified. METHODS AND RESULTS: In myocardial samples from patients with heart failure, quantitative real-time polymerase chain reaction revealed an increase in messenger RNA for C/EBP homologous protein (CHOP), a transcriptional factor that mediates endoplasmic reticulum-initiated apoptotic cell death. We performed transverse aortic constriction or sham operation on wild-type (WT) and CHOP-deficient mice. The CHOP-deficient mice showed less cardiac hypertrophy, fibrosis, and cardiac dysfunction compared with WT mice at 4 weeks after transverse aortic constriction, although the contractility of isolated cardiomyocytes from CHOP-deficient mice was not significantly different from that in the WT mice. In the hearts of CHOP-deficient mice, phosphorylation of eukaryotic translation initiation factor 2alpha, which may reduce protein translation, was enhanced compared with WT mice. In the hearts of WT mice, CHOP-increased apoptotic cell death with activation of caspase-3 was observed at 4 weeks after transverse aortic constriction. In contrast, CHOP-deficient mice had less apoptotic cell death and lower caspase-3 activation at 4 weeks after transverse aortic constriction. Furthermore, the Bcl2/Bax ratio was decreased in WT mice, whereas this change was significantly blunted in CHOP-deficient mice. Real-time polymerase chain reaction microarray analysis revealed that CHOP could regulate several Bcl2 family members in failing hearts. CONCLUSIONS: We propose the novel concept that CHOP, which may modify protein translation and mediate endoplasmic reticulum-initiated apoptotic cell death, contributes to development of cardiac hypertrophy and failure induced by pressure overload.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Insuficiencia Cardíaca/genética , Animales , Apoptosis/fisiología , Proteínas Potenciadoras de Unión a CCAAT/deficiencia , Cardiomiopatías/patología , Cardiomiopatías/cirugía , Cartilla de ADN , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Volumen Sistólico
2.
J Mol Cell Cardiol ; 48(6): 1280-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20170659

RESUMEN

The unfolded protein response (UPR) is triggered to assist protein folding when endoplasmic reticulum (ER) function is impaired. Recent studies demonstrated that ER stress can also induce cell-specific genes. In this study, we examined whether X-box binding protein 1 (XBP1), a major UPR-linked transcriptional factor, regulates the expression of brain natriuretic peptide (BNP) in cardiomyocytes. In samples from failing human hearts, extensive splicing of XBP1 was observed along with increased expression of glucose-regulated protein of 78 kDa (GRP78), a target of spliced XBP1 (sXBP1), suggesting that the UPR was induced in heart failure in humans. Interestingly, quantitative real-time PCR revealed a positive correlation between cardiac expression of GRP78 and BNP, leading us to test the hypothesis that sXBP1 regulates BNP as well as GRP78 in cardiomyocytes. A pharmacological ER stressor caused a dose-dependent increase in the expression of sXBP1 and BNP by cultured cardiomyocytes. Short interfering RNA targeting XBP1 suppressed the induction of BNP expression by a pharmacological ER stressor or norepinephrine, which was rescued by the adenovirus-mediated overexpression of sXBP1. The promoter assay with overexpression of sXBP1 or norepinephrine showed that the proximal AP1/CRE-like element in the promoter region of BNP was critical for transcriptional regulation of BNP by sXBP1. Direct binding of sXBP1 to this element was confirmed by the chromatin immunoprecipitation assay. These findings suggest that ER stress observed in failing hearts regulates cardiac BNP expression through a novel promoter region of the AP1/CRE-like element.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Miocitos Cardíacos/citología , Péptido Natriurético Encefálico/biosíntesis , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Animales , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Humanos , Microscopía Confocal/métodos , Miocardio/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Ratas , Factores de Transcripción del Factor Regulador X , Proteína 1 de Unión a la X-Box
3.
Cardiovasc Res ; 79(4): 600-10, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508854

RESUMEN

AIMS: Proteasome inhibitors are a novel class of anticancer agents that induce tumour cell death via endoplasmic reticulum (ER) stress. Since ER stress is involved in the development of heart failure, we investigated the role of ER-initiated cardiomyocyte death by proteasome inhibition. METHODS AND RESULTS: Rat neonatal cardiomyocytes were used in this study. Proteasome activity was assayed using proteasome peptidase substrates. Cell viability and apoptosis were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenol tetrazolium bromide and flow cytometry, respectively. Western blot analysis, real-time polymerase chain reaction (PCR) and reverse transcriptional PCR were used to detect the expression of protein and messenger ribonucleic acid (RNA). The location of overexpressed glucose-regulated protein (GRP) 78 was observed by confocal fluorescence microscopy. Proteasome inhibition induced cardiomyocyte death and activated ER stress-induced transcriptional factor ATF6, but not XBP1 (X-box binding protein 1), without up-regulating ER chaperones. ER-initiated apoptosis signalling, including cytosine-cytosine-adenine-adenine-thymine enhancer-binding protein (C/EBP) homologous protein (CHOP), c-Jun-N-terminal kinase (JNK), and caspase-12, was activated by proteasome inhibition. Short interference RNA targeting CHOP, but not the blockage of caspase-12 or JNK pathway, attenuated cardiomyocyte death. Overexpression of GRP78 suppressed both CHOP expression and cardiomyocyte death by proteasome inhibition. CONCLUSION: These findings demonstrate that proteasome inhibition induces ER-initiated cardiomyocyte death via CHOP-dependent pathways without compensatory up-regulation of ER chaperones. Supplement and/or pharmacological induction of GRP78 can attenuate cardiac damage by proteasome inhibition.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Retículo Endoplásmico/efectos de los fármacos , Chaperonas Moleculares/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteasoma , Factor de Transcripción Activador 6/metabolismo , Animales , Animales Recién Nacidos , Caspasa 12/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/patología , Proteínas de Choque Térmico/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leupeptinas/farmacología , Chaperonas Moleculares/genética , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Oligopéptidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , Tunicamicina/farmacología
4.
J Cardiol Cases ; 1(1): e25-e27, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30615758

RESUMEN

Percutaneous transluminal septal myocardial ablation (PTSMA) is appropriate in patients who are refractory to medical treatments for hypertrophic obstructive cardiomyopathy (HOCM). However, about 10% of patients show recurrence of left ventricular outflow tract pressure gradient (LVOG) after PTSMA and need repeat PTSMA. The authors describe two HOCM cases who underwent intravenous myocardial contrast echocardiography (IVMCE) at two weeks after PTSMA. IVMCE revealed the total defect of the PTSMA area in one patient, a 69-year-old woman. This patient has shown no recurrence of LVOG for 4 years. Faintly stained PTSMA area by IVMCE was seen in the other patient, an 83-year-old woman. This patient had a recurrence of LVOG 3 months later. Therefore, in conclusion, IVMCE might predict recurrence of LVOG after PTSMA.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA