Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Intern Med ; 285(4): 398-406, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30289186

RESUMEN

OBJECTIVES: With the emergence of targeted cell transplantation and gene therapy, there is a need for minimally invasive tissue access to facilitate delivery of therapeutic substrate. The objective of this study was to demonstrate the suitability of an endovascular device which is able to directly access tissue and deliver therapeutic agent to the heart, kidney and pancreas without need to seal the penetration site. METHODS: In vivo experiments were performed in 30 swine, including subgroups with follow-up to evaluate complications. The previously described trans-vessel wall (VW) device was modified to be sharper and not require tip detachment to seal the VW. Injections into targets in the heart (n = 13, 24-h follow-up n = 5, 72-h follow-up n = 3), kidney (n = 8, 14-day follow-up n = 3) and pancreas (n = 5) were performed. Some animals were used for multiple organ injections. Follow-up consisted of clinical monitoring, angiography and necropsy. Transvenous (in heart) and transarterial approaches (in heart, kidney and pancreas) were used. Injections were targeted towards the subepicardium, endomyocardium, pancreas head and tail, and kidney subcapsular space and cortex. RESULTS: Injections were successful in target organs, visualized by intraparenchymal contrast on fluoroscopy and by necropsy. No serious complications (defined as heart failure or persistent arrhythmia, haemorrhage requiring treatment or acute kidney injury) were encountered over a total of 157 injections. CONCLUSIONS: The trans-VW device can achieve superselective injections to the heart, pancreas and kidney for delivery of therapeutic substances without tip detachment. All parts of these organs including the subepicardium, pancreas tail and renal subcapsular space can be efficiently reached.


Asunto(s)
Trasplante de Células/métodos , Sistemas de Liberación de Medicamentos/métodos , Procedimientos Endovasculares/métodos , Corazón , Riñón , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Páncreas , Animales , Estudios de Factibilidad , Inyecciones/métodos , Porcinos
2.
Nat Med ; 7(11): 1236-40, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11689889

RESUMEN

Myocardial hypertrophy is an adaptational response of the heart to increased work load, but it is also associated with a high risk of cardiac mortality due to its established role in the development of cardiac failure, one of the leading causes of death in developed countries. Multiple growth factors and various downstream signaling pathways involving, for example, ras, gp-130 (ref. 4), JNK/p38 (refs. 5,6) and calcineurin/NFAT/CaM-kinase have been implicated in the hypertrophic response. However, there is evidence that the initial phase in the development of myocardial hypertrophy involves the formation of cardiac para- and/or autocrine factors like endothelin-1, norepinephrine or angiotensin II (refs. 7,8), the receptors of which are coupled to G-proteins of the Gq/11-, G12/13- and Gi/o-families. Cardiomyocyte-specific transgenic overexpression of alpha1-adrenergic or angiotensin (AT1)-receptors as well as of the Gq alpha-subunit, Galphaq, results in myocardial hypertrophy. These data demonstrate that chronic activation of the Gq/G11-family is sufficient to induce myocardial hypertrophy. In order to test whether Gq/G11 mediate the physiological hypertrophy response to pressure overload, we generated a mouse line lacking both Galphaq and Galpha11 in cardiomyocytes. These mice showed no detectable ventricular hypertrophy in response to pressure-overload induced by aortic constriction. The complete lack of a hypertrophic response proves that the Gq/G11-mediated pathway is essential for cardiac hypertrophy induced by pressure overload and makes this signaling process an interesting target for interventions to prevent myocardial hypertrophy.


Asunto(s)
Cardiomiopatía Hipertrófica/prevención & control , Proteínas de Unión al GTP Heterotriméricas/antagonistas & inhibidores , Animales , Secuencia de Bases , Presión Sanguínea , Cardiomiopatía Hipertrófica/etiología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , ADN Complementario/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes
3.
Nat Med ; 7(5): 591-7, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11329061

RESUMEN

Although cytoskeletal mutations are known causes of genetically based forms of dilated cardiomyopathy, the pathways that link these defects with cardiomyopathy are unclear. Here we report that the alpha-actinin-associated LIM protein (ALP; Alp in mice) has an essential role in the embryonic development of the right ventricular (RV) chamber during its exposure to high biomechanical workloads in utero. Disruption of the gene encoding Alp (Alp) is associated with RV chamber dilation and dysfunction, directly implicating alpha-actinin-associated proteins in the onset of cardiomyopathy. In vitro assays showed that Alp directly enhances the capacity of alpha-actinin to cross-link actin filaments, indicating that the loss of Alp function contributes to destabilization of actin anchorage sites in cardiac muscle. Alp also colocalizes at the intercalated disc with alpha-actinin and gamma-catenin, the latter being a known disease gene for human RV dysplasia. Taken together, these studies point to a novel developmental pathway for RV dilated cardiomyopathy via instability of alpha-actinin complexes.


Asunto(s)
Actinina/genética , Cardiomiopatías/etiología , Ventrículos Cardíacos/patología , Proteínas de Homeodominio/fisiología , Animales , Cardiomiopatías/genética , Proteínas del Citoesqueleto/metabolismo , Desmoplaquinas , Ventrículos Cardíacos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , gamma Catenina
4.
J Cell Biol ; 137(1): 131-40, 1997 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-9105042

RESUMEN

Hypertrophic cardiomyopathy is a human heart disease characterized by increased ventricular mass, focal areas of fibrosis, myocyte, and myofibrillar disorganization. This genetically dominant disease can be caused by mutations in any one of several contractile proteins, including beta cardiac myosin heavy chain (beta MHC). To determine whether point mutations in human beta MHC have direct effects on interfering with filament assembly and sarcomeric structure, full-length wild-type and mutant human beta MHC cDNAs were cloned and expressed in primary cultures of neonatal rat ventricular cardiomyocytes (NRC) under conditions that promote myofibrillogenesis. A lysine to arginine change at amino acid 184 in the consensus ATP binding sequence of human beta MHC resulted in abnormal subcellular localization and disrupted both thick and thin filament structure in transfected NRC. Diffuse beta MHC K184R protein appeared to colocalize with actin throughout the myocyte, suggesting a tight interaction of these two proteins. Human beta MHC with S472V mutation assembled normally into thick filaments and did not affect sarcomeric structure. Two mutant myosins previously described as causing human hypertrophic cardiomyopathy, R249Q and R403Q, were competent to assemble into thick filaments producing myofibrils with well defined I bands, A bands, and H zones. Coexpression and detection of wild-type beta MHC and either R249Q or R403Q proteins in the same myocyte showed these proteins are equally able to assemble into the sarcomere and provided no discernible differences in subcellular localization. Thus, human beta MHC R249Q and R403Q mutant proteins were readily incorporated into NRC sarcomeres and did not disrupt myofilament formation. This study indicates that the phenotype of myofibrillar disarray seen in HCM patients which harbor either of these two mutations may not be directly due to the failure of the mutant myosin heavy chain protein to assemble and form normal sarcomeres, but may rather be a secondary effect possibly resulting from the chronic stress of decreased beta MHC function.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cadenas Pesadas de Miosina/genética , Mutación Puntual/fisiología , Sarcómeros/química , Actinas/química , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Aminoácidos/genética , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Clonación Molecular , ADN Complementario , Epítopos/análisis , Expresión Génica/fisiología , Ventrículos Cardíacos , Humanos , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/fisiología , Fibras Musculares Esqueléticas/ultraestructura , Ratas , Sarcómeros/metabolismo , Transfección
5.
Science ; 264(5158): 582-6, 1994 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-8160017

RESUMEN

Transgenic mice were created with cardiac-specific overexpression of the beta 2-adrenergic receptor. This resulted in increased basal myocardial adenylyl cyclase activity, enhanced atrial contractility, and increased left ventricular function in vivo; these parameters at baseline in the transgenic animals were equal to those observed in control animals maximally stimulated with isoproterenol. These results illustrate a useful approach for studying the effect of gene expression on cardiac contractility. Because chronic heart failure in humans is accompanied by a reduction in the number of myocardial beta-adrenergic receptors and in inotropic responsiveness, these results suggest a potential gene therapy approach to this disease state.


Asunto(s)
Adenilil Ciclasas/metabolismo , Contracción Miocárdica , Miocardio/metabolismo , Receptores Adrenérgicos beta/genética , Función Ventricular Izquierda , Animales , Técnicas de Transferencia de Gen , Terapia Genética , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Frecuencia Cardíaca , Humanos , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Miosinas/genética , Fenotipo , Regiones Promotoras Genéticas , Receptores Adrenérgicos beta/biosíntesis , Receptores Adrenérgicos beta/fisiología
6.
J Clin Invest ; 91(4): 1713-20, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8386193

RESUMEN

Infection with the Ad5-SVR4 virus was used to introduce the large T antigen encoding region of the SV40 virus into bovine and human corneal endothelial cells. Expression of large T antigen occurred in 40% of bovine corneal endothelial cells after a 24-h incubation time versus 12% after 8 h of incubation. By 48 h after infection, almost all (92.8%) bovine corneal endothelial cells expressed large T antigen. Bovine and human corneal endothelial cells which expressed large T antigen proliferated and the characteristic morphologic features of corneal endothelium were maintained. This method may enable growth of enough corneal endothelium to perform studies to elucidate the biochemical mechanisms involved in regulating endothelial cell function.


Asunto(s)
Adenoviridae/inmunología , Antígenos Transformadores de Poliomavirus/fisiología , Endotelio Corneal/inmunología , Virus 40 de los Simios/inmunología , Proteínas Virales/inmunología , Animales , Bovinos , División Celular , Células Cultivadas , Endotelio Corneal/citología , Endotelio Corneal/microbiología , Ingeniería Genética , Humanos , Proteínas Recombinantes/inmunología , Virosis/metabolismo
7.
J Clin Invest ; 82(4): 1333-8, 1988 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-3139713

RESUMEN

Previous studies have suggested that phospholipid degradation is closely associated with the development of sarcolemmal membrane injury. This study was initiated to characterize the effects of synthetic inhibitors of phospholipase activities using a cultured myocardial cell model in which arachidonic acid is liberated after treatment with the metabolic inhibitor, iodoacetate. Pretreatment with a steroidal diamine (U26,384) blocked the degradation of labeled phosphatidylcholine and the release of arachidonic acid in cultured myocardial cells during ATP depletion. Inhibition of phospholipid degradation by U26,384 prevented the development of sarcolemmal membrane defects and the release of creatine kinase from the cultured myocardial cells during ATP depletion. Pretreatment with U26,384 had no significant effect on the extent of ATP depletion after iodoacetate treatment, which indicates that the activity of this compound could not be simply ascribed to a sparing effect on ATP concentration. These results support the hypothesis that the development of sarcolemmal membrane injury and the associated loss of cell viability are causally related to progressive phospholipid degradation. In addition, these studies indicate that the release of arachidonic acid during ATP depletion is associated with the net loss of the phosphatidylcholine molecule.


Asunto(s)
Adenosina Trifosfato/deficiencia , Ácidos Araquidónicos/metabolismo , Estrenos/farmacología , Membranas Intracelulares/patología , Miocardio/ultraestructura , Sarcolema/patología , Animales , Ácido Araquidónico , Células Cultivadas , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/ultraestructura , Miocardio/metabolismo , Fosfatidilcolinas/metabolismo , Fosfolipasas/antagonistas & inhibidores , Ratas , Sarcolema/efectos de los fármacos , Sarcolema/ultraestructura
8.
J Clin Invest ; 84(1): 331-6, 1989 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2544627

RESUMEN

This study examined the diversity of Na+ channel gene expression in intact cardiac tissue and purified myocardial cells. The screening of neonatal rat myocardial cell cDNA libraries with a conserved rat brain Na+ channel cDNA probe, resulted in the isolation and characterization of a putative rat cardiac Na+ channel cDNA probe (pCSC-1). The deduced amino acid sequence of pCSC-1 displayed a striking degree of homology with the eel, rat brain-1, and rat brain-2 Na+ channel, thereby identifying pCSC-1 as a related member of the family of Na+ channel genes. Northern blot analysis revealed the expression of a 7-kb CSC-1 transcript in rat cardiac tissue and purified myocardial cells, but little or no detectable expression of CSC-1 in rat brain, skeletal muscle, denervated skeletal muscle, or liver. Using RNase protection and Northern blot hybridization with specific rat brain Na+ channel gene probes, expression of the rat brain-1 Na+ channel was observed in rat myocardium, but no detectable expression of the rat brain-2 gene was found. This study provides evidence for the expression of diverse Na+ channel mRNAs in rat myocardium and presents the initial characterization of a new, related member of the family of Na+ channel genes, which appears to be expressed in a cardiac-specific manner.


Asunto(s)
Regulación de la Expresión Génica , Miocardio/metabolismo , ARN Mensajero/genética , Canales de Sodio/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Encéfalo/enzimología , Células Cultivadas , Datos de Secuencia Molecular , Músculos/metabolismo , Hibridación de Ácido Nucleico , Fosfotransferasas/metabolismo , Ratas
9.
J Clin Invest ; 96(3): 1311-8, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7657806

RESUMEN

To determine whether similar or divergent pathways mediate atrial natriuretic factor (ANF) induction in neonatal and hypertrophied adult ventricular myocardium, and to assess whether studies using an in vitro model system of hypertrophy have fidelity to the in vivo context during pressure overload hypertrophy, we generated transgenic mice which harbor either 638 or 3,003 bp of the rat ANF 5' flanking region ligated upstream from a luciferase reporter. Luciferase activity in the ventricles of day 1 transgenic neonates was 8-24-fold higher than the levels expressed in the ventricles of adult mice. Adult mice expressed the luciferase reporter in an appropriate tissue-specific manner. Transverse aortic constriction of adult mice harboring ANF reporter transgenes demonstrated no significant increase in reporter activity in the ventricle. These findings demonstrate that distinct regions of the ANF 5'-flanking region are required for inducible expression of the ANF gene in the hypertrophic adult ventricle compared with those required for atrial-specific and developmentally appropriate expression in the intact neonatal heart. Furthermore, the cis regulatory elements necessary for induction of ANF expression in endothelin-1 or alpha 1-adrenergically stimulated cultured neonatal ventricular myocytes are not sufficient for induction in the in vivo context of pressure overload hypertrophy.


Asunto(s)
Factor Natriurético Atrial/biosíntesis , Factor Natriurético Atrial/genética , Cardiomegalia/metabolismo , Expresión Génica , Miocardio/metabolismo , Animales , Animales Recién Nacidos , Cloranfenicol O-Acetiltransferasa/biosíntesis , Femenino , Corazón/crecimiento & desarrollo , Ventrículos Cardíacos , Luciferasas/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
10.
J Clin Invest ; 98(6): 1332-43, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8823298

RESUMEN

Retinoid-dependent pathways play a central role in regulating cardiac morphogenesis. Recently, we characterized gene-targeted RXR alpha -/- embryos, which display an atrial-like ventricular phenotype with the development of heart failure and lethality at embryonic day 14.5. To quantitate the frequency and complexity of cardiac morphogenic defects, we now use microdissection and scanning electron microscopy to examine 107 wild-type, heterozygous, and homozygous embryos at embryonic day 13.5, 14.5, and 15.5. RXR alpha -/- embryos display complex defects, including ventricular septal, atrioventricular cushion, and conotruncal ridge defects, with double outlet right ventricle, aorticopulmonary window, and persistent truncus arteriosus. In addition, heterozygous RXR alpha embryos display a predisposition for trabecular and papillary muscle defects, ventricular septal defects, conotruncal ridge defects, atrioventricular cushion defects, and pulmonic stenosis. Lastly, we show that the intermediate anatomic phenotype displayed by heterozygous embryos is mirrored in the molecular marker MLC-2a. The intermediate phenotype of RXR alpha heterozygous embryos documents a gene dosage effect for RXR alpha in maintaining normal cardiac morphogenesis. In addition, some defects in RXR alpha mutant mice are phenocopies of human congenital heart defects, thereby suggesting that a relative deficiency in RXR alpha or molecules downstream in its signaling pathway may represent congenital heart disease-susceptibility genes.


Asunto(s)
Corazón Fetal/crecimiento & desarrollo , Cardiopatías Congénitas/genética , Péptidos , Receptores de Ácido Retinoico/genética , Animales , Defectos de la Almohadilla Endocárdica/genética , Corazón Fetal/ultraestructura , Defectos del Tabique Interventricular/genética , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Electrónica de Rastreo , Modelos Biológicos , Músculos/anomalías , Biosíntesis de Péptidos , Tronco Arterial Persistente/genética
11.
J Clin Invest ; 95(2): 619-27, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7860746

RESUMEN

To determine whether additional hypertrophy would be beneficial or maladaptive in cardiac failure, the effects of insulin-like growth factor (IGF-1) were investigated in rats with left ventricular (LV) dysfunction. In normal rats, 3 mg/kg per d of recombinant human IGF-1 for 14 d augmented LV wt (32%) and increased LV/body wt ratio (P < 0.01). 2 d after coronary occlusion, rats were randomized to IGF-1 (3 mg/kg per d) or placebo. After 2 wk, IGF-1-treated rats showed significant increases in LV wt (13%) and LV wt/tibial length ratio, but LV/body wt ratio was unchanged. By microangiography, compared with controls (n = 12) IGF-1-treated rats (n = 16) showed increased LV end-diastolic volume (19%) and stroke volume (31%) (both significant normalized to tibial length, but not to body wt). Average infarct size did not differ between groups. The LV ejection fraction (EF) was not significantly different between groups, but estimated cardiac output was higher in treated rats; there was a significant interaction for the EF between infarct size and treatment (P = 0.029) and a trend for EF to be higher in treated rats with large infarctions (EF 33.4 vs 25.1% in controls). Myocyte cross-sectional areas in noninfarcted LV zones tended to be larger in treated rats (232.1 vs 205.4 microns 2; P = 0.10), but there was no difference in capillary density and collagen content did not differ between groups. In conclusion, IGF-1 administration caused hypertrophy of the normal heart in vivo. When stimulated by IGF-1, the severely dysfunctional heart in evolving myocardial infarction is capable of undergoing additional hypertrophy with evidence of improved function, suggesting a beneficial effect. Further investigation of the potential role of growth factor therapy in heart failure appears warranted.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/fisiopatología , Factor I del Crecimiento Similar a la Insulina/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Angiografía Coronaria , Femenino , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Humanos , Hipertrofia Ventricular Izquierda/patología , Tamaño de los Órganos/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Valores de Referencia
12.
J Clin Invest ; 98(11): 2648-55, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8958230

RESUMEN

To circumvent the embryonic lethality of a complete deficiency in insulin-like growth factor 1 (IGF-1), we generated mice homozygous for a site-specific insertional event that created a mutant IGF-1 allele (igf1m). These mice have IGF-1 levels 30% of wild type yet survive to adulthood, thereby allowing physiological analysis of the phenotype. Miniaturized catheterization technology revealed elevated conscious blood pressure in IGF-1(m/m) mice, and measurements of left ventricular contractility were increased. Adenylyl cyclase activity was enhanced in IGF-1(m/m) hearts, without an increase in beta-adrenergic receptor density, suggesting that crosstalk between IGF-1 and beta-adrenergic signaling pathways may mediate the increased contractility. The hypertrophic response of the left ventricular myocardium in response to aortic constriction, however, was preserved in IGF-1(m/m) mice. We conclude that chronic alterations in IGF-1 levels can selectively modulate blood pressure and left ventricular function, while not affecting adaptive myocardial hypertrophy in vivo.


Asunto(s)
Adenilil Ciclasas/metabolismo , Presión Sanguínea , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/genética , Mutagénesis Insercional , Contracción Miocárdica , Hipófisis/metabolismo , Receptores Adrenérgicos beta/metabolismo , Alelos , Animales , Glucemia/metabolismo , Cartilla de ADN , Femenino , Hormona del Crecimiento/sangre , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Masculino , Ratones , Ratones Mutantes , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa , Caracteres Sexuales
13.
J Clin Invest ; 75(6): 1770-80, 1985 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3924955

RESUMEN

The present study utilized a cultured myocardial cell model to evaluate the relationship between the release of arachidonate from membrane phospholipids, and the progression of cell injury during ATP depletion. High-energy phosphate depletion was induced by incubating cultured neonatal rat myocardial cells with various combinations of metabolic inhibitors (deoxyglucose, oligomycin, cyanide, and iodoacetate). Phospholipid degradation was assessed by the release of radiolabeled arachidonate from membrane phospholipids. In this model, the current study demonstrates that (a) cultured myocardial cells display a time-dependent progression of cell injury during ATP depletion; (b) the morphologic patterns of mild and severe cell injury in the cultured cells are similar to those found in intact ischemic canine myocardial models; (c) cultured myocardial cells release arachidonate from membrane phospholipids during ATP depletion; and (d) using two separate combinations of metabolic inhibitors, there is a correlation between the release of arachidonate, the development of severe cellular and sarcolemmal damage, the release of creatine kinase into the extracellular medium, and the loss of the ability of the myocardial cells to regenerate ATP when the metabolic inhibitors are removed. Thus, the present results suggest that during ATP depletion, in cultured neonatal rat myocardial cells, the release of arachidonate from myocardial membrane phospholipids is linked to the development of membrane defects and the associated loss of cell viability.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ácidos Araquidónicos/metabolismo , Enfermedad Coronaria/metabolismo , Miocardio/metabolismo , Animales , Ácido Araquidónico , Células Cultivadas , Creatina Quinasa/metabolismo , Cianuros/farmacología , Desoxiglucosa/farmacología , Yodoacetatos/farmacología , Ácido Yodoacético , Cinética , Lípidos de la Membrana/metabolismo , Microscopía Electrónica , Miocardio/citología , Oligomicinas/farmacología , Fosfolípidos/metabolismo , Ratas
14.
J Clin Invest ; 103(12): 1627-34, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10377168

RESUMEN

RhoA is a low-molecular-weight GTPase that has been implicated in the regulation of hypertrophic cardiac muscle cell growth. To study the role of RhoA in control of cardiac function in vivo, transgenic mice expressing wild-type and constitutively activated forms of RhoA under the control of the cardiac-specific alpha-myosin heavy chain promoter were generated. Transgene-positive mice expressing high levels of either wild-type or activated RhoA showed pronounced atrial enlargement and manifested a lethal phenotype, often preceded by generalized edema, with most animals dying over the course of a few weeks. Echocardiographic analysis of visibly healthy wild-type RhoA transgenic mice revealed no significant change in left ventricular function. As their condition deteriorated, significant dilation of the left ventricular chamber and associated decreases in left ventricular contractility were detected. Heart rate was grossly depressed in both wild-type and activated RhoA-expressing mice, even prior to the onset of ventricular failure. Electrocardiography showed evidence of atrial fibrillation and atrioventricular block. Interestingly, muscarinic receptor blockade with atropine did not elicit a positive chronotropic response in the transgenic mice. We suggest that RhoA regulates cardiac sinus and atrioventricular nodal function and that its overexpression results in bradycardia and development of ventricular failure.


Asunto(s)
Nodo Atrioventricular/fisiopatología , Cardiomiopatía Dilatada/enzimología , Contracción Miocárdica , Miocardio/enzimología , Proteínas Serina-Treonina Quinasas/biosíntesis , Nodo Sinoatrial/fisiopatología , Animales , Factor Natriurético Atrial/biosíntesis , Factor Natriurético Atrial/genética , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Regulación de la Expresión Génica , Atrios Cardíacos/fisiopatología , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/fisiopatología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica/genética , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Pesadas de Miosina/genética , Tamaño de los Órganos/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Quinasas Asociadas a rho
15.
J Clin Invest ; 108(10): 1459-67, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11714737

RESUMEN

The gp130 cytokine receptor activates a cardiomyocyte survival pathway during the transition to heart failure following the biomechanical stress of pressure overload. Although gp130 activation is observed transiently during transverse aortic constriction (TAC), its mechanism of inactivation is largely unknown in cardiomyocytes. We show here that suppressor of cytokine signaling 3 (SOCS3), an intrinsic inhibitor of JAK, shows biphasic induction in response to TAC. The induction of SOCS3 was closely correlated with STAT3 phosphorylation, as well as the activation of an embryonic gene program, suggesting that cardiac gp130-JAK signaling is precisely controlled by this endogenous suppressor. In addition to its cytoprotective action, gp130-dependent signaling induces cardiomyocyte hypertrophy. Adenovirus-mediated gene transfer of SOCS3 to ventricular cardiomyocytes completely suppressed both hypertrophy and antiapoptotic phenotypes induced by leukemia inhibitory factor (LIF). To our knowledge, this is the first clear evidence that these two separate cardiomyocyte phenotypes induced by gp130 activation lie downstream of JAK. Three independent signaling pathways, STAT3, MEK1-ERK1/2, and AKT activation, that are coinduced by LIF stimulation were completely suppressed by SOCS3 overexpression. We conclude that SOCS3 is a mechanical stress-inducible gene in cardiac muscle cells and that it directly modulates stress-induced gp130 cytokine receptor signaling as the key molecular switch for a negative feedback circuit for both myocyte hypertrophy and survival.


Asunto(s)
Antígenos CD/fisiología , Cardiomegalia , Supervivencia Celular/fisiología , Glicoproteínas de Membrana/fisiología , Miocardio/patología , Proteínas/metabolismo , Proteínas Represoras , Transducción de Señal , Factores de Transcripción , Animales , Antígenos CD/metabolismo , Receptor gp130 de Citocinas , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas
16.
Mol Cell Biol ; 15(6): 2972-82, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7760795

RESUMEN

Transient assays in cultured ventricular muscle cells and studies in transgenic mice have identified two adjacent regulatory elements (HF-1a and HF-1b/MEF-2) as required to maintain ventricular chamber-specific expression of the myosin light-chain 2v (MLC-2v) gene. A rat neonatal heart cDNA library was screened with an HF-1a binding site, resulting in the isolation of EFIA, the rat homolog of human YB-1. Purified recombinant EFIA/YB-1 protein binds to the HF-1a site in a sequence-specific manner and contacts a subset of the HF-1a contact points made by the cardiac nuclear factor(s). The HF-1a sequence contains AGTGG, which is highly homologous to the inverted CCAAT core of the EFIA/YB-1 binding sites and is found to be essential for binding of the recombinant EFIA/YB-1. Antiserum against Xenopus YB-3 (100% identical in the DNA binding domain and 89% identical in overall amino acid sequence to rat EFIA) can specifically abolish a component of the endogenous HF-1a complex in the rat cardiac myocyte nuclear extracts. In cotransfection assays, EFIA/YB-1 increased 250-bp MLC-2v promoter activity by 3.4-fold specifically in the cardiac cell context and in an HF-1a site-dependent manner. EFIA/YB-1 complexes with an unknown protein in cardiac myocyte nuclear extracts to form the endogenous HF-1a binding activity. Immunocoprecipitation revealed that EFIA/YB-1 has a major associated protein of approximately 30 kDa (p30) in cardiac muscle cells. This study suggests that EFIA/YB-1, together with the partner p30, binds to the HF-1a site and, in conjunction with HF-1b/MEF-2, mediates ventricular chamber-specific expression of the MLC-2v gene.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Proteínas de Unión al ADN/metabolismo , Ventrículos Cardíacos/metabolismo , Miosinas/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Células Cultivadas , ADN Complementario , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Miosinas/metabolismo , Factores de Transcripción NFI , Proteínas Nucleares , Ratas , Transcripción Genética , Xenopus , Proteína 1 de Unión a la Caja Y
17.
Mol Cell Biol ; 12(4): 1469-79, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1532229

RESUMEN

Recent studies have identified a conserved 28-bp element (HF-1) within the rat cardiac MLC-2 gene which confers cardiac muscle-specific and inducible expression during myocardial cell hypertrophy. Utilizing a combination of independent experimental approaches, this study characterizes two cardiac nuclear factors which bind to HF-1, a ubiquitous factor (HF-1a), and an A + T-rich binding factor (HF-1b) which is preferentially expressed in differentiated cardiac and skeletal muscle cells. The HF-1a binding site is located in a core region of the 28-bp conserved element, immediately upstream from the A + T-rich HF-1b site, which is homologous to the MEF-2 site found in a number of muscle genes. By a number of separate criteria (gel mobility shift, competition, and mutagenesis studies), HF-1b and MEF-2 appear to be indistinguishable and thus are either identical or closely related muscle factors. Transient assays of luciferase reporter genes containing point mutations throughout the 28-bp HF-1 regulatory element document the importance of both the HF-1a and HF-1b sites in transient assays in ventricular muscle cells. In the native 250-bp MLC-2 promoter fragment, mutations in the single E box had little effect on cardiac muscle specificity, while point mutations in either the HF-1a or HF-1b binding site significantly reduced promoter activity, underscoring the importance of both the HF-1a and HF-1b sites in the transcriptional activation of this cardiac muscle gene. Thus, this study provides evidence that a novel, ubiquitous factor (HF-1a) and a muscle factor (HF-1b/MEF-2) can form a novel, E-box-independent pathway for muscle-specific expression in ventricular cardiac muscle cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Miocardio/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Secuencia de Bases , Cardiomegalia/metabolismo , Diferenciación Celular , Análisis Mutacional de ADN , Regulación de la Expresión Génica , Datos de Secuencia Molecular , Miocardio/patología , Ratas , Proteínas Recombinantes de Fusión , Distribución Tisular
18.
Mol Cell Biol ; 14(2): 1220-9, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8289802

RESUMEN

The cardiac myosin light-chain 2v (MLC-2v) gene has served as a model system to identify the pathways which restrict the expression of cardiac muscle genes to particular chambers of the heart during cardiogenesis. To identify the critical cis regulatory elements which mediate ventricular chamber-specific expression of the MLC-2v gene in the in vivo context, a series of transgenic mice which harbor mutations in putative MLC-2 cis regulatory elements in a 250-bp MLC-2-luciferase fusion gene which is expressed in a ventricular chamber-specific fashion in transgenic mice were generated. These studies demonstrate that both components of HF-1 (HF-1a and HF-1b/MEF-2) are required to maintain ventricular chamber-specific expression and function as positive regulatory elements. Mutations in another conserved element (HF-2) are without statistically significant effect on ventricular chamber expression. Transgenics harboring mutations in the E-box site also displayed significant upregulation of reporter activity in the soleus, gastrocnemius, and uterus, with a borderline effect on expression in liver. Mutations in another conserved element (HF-3) result in a marked (> 75-fold) upregulation of the luciferase reporter activity in the soleus muscle of multiple independent or transgenic founders. Since the HF-3 mutations appeared to have only a marginal effect on luciferase reporter activity in liver tissue, HF-3 appears to function as a novel negative regulatory element to primarily suppress expression in muscle tissues. Thus, a combination of positive (HF-1a/HF-1b) and negative (E-box and HF-3) regulatory elements appear to be required to maintain ventricular chamber-specific expression in the in vivo context.


Asunto(s)
Luciferasas/biosíntesis , Miocardio/metabolismo , Miosinas/biosíntesis , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Secuencia de Bases , Secuencia Conservada , Femenino , Expresión Génica , Ventrículos Cardíacos/metabolismo , Hígado/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Músculos/metabolismo , Especificidad de Órganos , Proteínas Recombinantes de Fusión/biosíntesis , Útero/metabolismo
19.
Mol Cell Biol ; 14(11): 7331-9, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7935447

RESUMEN

Previous studies have documented that 250 bp of the rat cardiac ventricular myosin light-chain 2 (MLC-2v) promoter is sufficient to confer cardiac muscle-specific expression on a luciferase reporter gene in both transgenic mice and primary cultured neonatal rat myocardial cells. Utilizing ligation-mediated PCR to perform in vivo dimethyl sulfate footprinting, the present study has identified protein-DNA interaction within the position from -176 to -165. This region, identified as MLE1, contains a core sequence, CACGTG, which conforms to the consensus E-box site and is identical to the upstream stimulating factor (USF)-binding site of the adenovirus major late promoter. Transient assays of luciferase reporter genes containing point mutations of the site demonstrate the importance of this cis regulatory element in the transcriptional activation of this cardiac muscle gene in ventricular muscle cells. The protein complex that occupies this site is capable of binding to HF-1a and PRE B sites which are known to be required for cardiac muscle-specific expression of rat MLC-2v and alpha-myosin heavy-chain genes, respectively. This study provides direct evidence that USF, a member of the basic helix-loop-helix leucine zipper family, binds to MLE1, HF-1a, and PRE B sites and suggests that it is a component of protein complexes that may coordinately control the expression of MLC-2v and alpha-myosin heavy-chain genes. The current study also provides evidence that USF can positively and negatively regulate the MLC-2v gene via independent cis regulatory elements.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Genes Reguladores , Secuencias Hélice-Asa-Hélice/genética , Miocardio/metabolismo , Miosinas/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Núcleo Celular/metabolismo , Células Cultivadas , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Ratas , Factores de Transcripción/genética , Factores Estimuladores hacia 5'
20.
Mol Cell Biol ; 14(6): 4269-79, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8196663

RESUMEN

The molecular characterization of a cardiac determination gene has been an elusive goal for the past several years. Prior to cloning of the skeletal muscle determination factor MyoD, the presence of a dominantly acting skeletal muscle determination factor had been inferred from the observation that the skeletal muscle phenotype was dominant in skeletal muscle-fibroblast heterokaryons (H. M. Blau, G. K. Pavlath, E. C. Hardeman, C.-P. Chiu, L. Siberstein, S. G. Webster, S. C. Miller, and D. Webster, Science 230:758-766, 1985). In these experiments, we have examined cardiac-fibroblast heterokaryons to investigate the existence of a dominantly acting cardiac determination factor. We have employed a novel experimental approach using primary embryonic fibroblasts from transgenic mice as a means of assaying for the activation of a cardiac promoter-luciferase reporter transgene within fibroblast nuclei. This approach provides a potential means of genetic selection for a dominantly acting positive factor and can be generalized to other systems. We have examined the expression of three markers of the cardiac lineage: a myofibrillar protein promoter (MLC2), a secreted protein (ANF), and a transcription factor (MEF2). MEF2 is specific to both cardiac and skeletal muscle cells. Our results indicate that in a majority of heterokaryons with an equal ratio of cardiac to fibroblast nuclei, none of these cardiac markers are expressed, indicating that the cardiac phenotype is not dominant over the embryonic fibroblast phenotype. The distinction from previous results with skeletal muscle is emphasized by our results with MEF2, which is dominantly expressed in skeletal muscle-fibroblast but not cardiac-fibroblast heterokaryons, supporting its divergent regulation in the two cell types.


Asunto(s)
Proteína MioD/biosíntesis , Miocardio/citología , Animales , Animales Recién Nacidos , Factor Natriurético Atrial/análisis , Factor Natriurético Atrial/biosíntesis , Línea Celular , Separación Celular , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Corazón/embriología , Corazón/fisiología , Luciferasas/biosíntesis , Luciferasas/metabolismo , Ratones , Proteína MioD/genética , Miocardio/metabolismo , Fenotipo , Ratas , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA