Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Dent Res ; 97(12): 1374-1382, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29863952

RESUMEN

Sclerostin (Scl) negatively regulates bone formation and favors bone resorption. Osteocytes, the cells responsible for mechanosensing, are known as the primary source of Scl and are a key regulator of bone remodeling through the induction of receptor activator of NF-κB ligand (RANKL). However, the spatiotemporal patterns of Scl in response to mechanical stimuli and their regulatory mechanisms remain unknown. We investigated the regulatory dynamics of the SOST/Scl expression generated by orthodontic tooth movement (OTM) in vivo and in vitro. In 8-wk-old male mice, coil springs were used to move the first molar mesially for 0, 1, 5, or 10 d. A regional histogram and the distribution patterns of the Scl expression showed that the Scl expression in the alveolar bone was increased on the compression side and peaked on day 5, with a gradual increase in the degree of significance. On day 10, the expression around the periodontal ligament (PDL)-alveolar bone boundary returned to the control level. Conversely, the expression of Scl on the tension side was only significantly decreased on day 1. Compressive force biphasically modulated the SOST/Scl expression in the isolated human PDL and thereby upregulated osteocytic SOST via paracrine activation in an osteocyte-PDL co-culture system designed to mimic OTM. This system did not affect the RANKL or OPG expression in osteocytes, suggesting that the bone resorption pathways are acted upon in a PDL-dependent and osteocyte-independent manner through RANKL/OPG signaling. Moreover, sclerostin neutralizing antibody significantly attenuated the upregulation of SOST that was induced by compressive force. In conclusion, our results provide evidence to support that factors secreted by the PDL, including SOST/Scl, control alveolar bone remodeling through osteocytic SOST/Scl in OTM.


Asunto(s)
Resorción Ósea/metabolismo , Glicoproteínas/metabolismo , Mecanotransducción Celular/fisiología , Osteocitos/metabolismo , Comunicación Paracrina/fisiología , Ligamento Periodontal/citología , Técnicas de Movimiento Dental , Proteínas Adaptadoras Transductoras de Señales , Animales , Remodelación Ósea , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ligando RANK/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 314(2): H370-H379, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29127239

RESUMEN

The small size of the mouse heart frequently imparts technical challenges when applying conventional in vivo imaging methods for assessing heart function. Here, we describe the use of high-frequency ultrasound imaging in conjunction with a size-tuned blood pool contrast agent for quantitatively assessing myocardial perfusion in living mice. A perflurocarbon microbubble formulation exhibiting a narrow size distribution was developed, and echogenicity was assessed at 18 MHz in vitro. Adult mice were subjected to permanent ligation of the left anterior descending artery. Ultrasound imaging was performed on day 7, and a cohort of intact mice was used as a control. Parasternal long-axis cine clips were acquired at 18 MHz before and after contrast administration. Reduced ejection fraction and increased end-systolic volume were observed in infarcted compared with control mice. In control animals, washin of the contrast agent was visible in all myocardial segments. Reduced contrast enhancement was observed in apical-posterolateral regions of all infarcted mice. A novel method for reslicing of the imaging data through the time domain provided a two-dimensional presentation of regional contrast agent washin, enabling convenient identification of locations exhibiting altered perfusion. Myocardial segments exhibiting diminished contractility were observed to have correspondingly low relative myocardial perfusion. The contrast agent formulation and methods demonstrated here provide the basis for simplifying routine in vivo estimation of infarct size in mice and may be particularly useful in longitudinal evaluation of revascularization interventions and assessment of peri-infarct ischemia. NEW & NOTEWORTHY Murine myocardial contrast echocardiography frequently suffers from poor sensitivity to contrast. Here, we formulated a novel size-tuned microbubble contrast agent and validated it for use with ultra-high-frequency ultrasound. A novel data method for evaluating myocardial perfusion based on reslicing the imaging data through the time domain is presented.


Asunto(s)
Medios de Contraste/administración & dosificación , Ecocardiografía/métodos , Infarto del Miocardio/diagnóstico por imagen , Imagen de Perfusión Miocárdica/métodos , Animales , Circulación Coronaria , Modelos Animales de Enfermedad , Interpretación de Imagen Asistida por Computador , Masculino , Ratones Endogámicos C57BL , Microburbujas , Contracción Miocárdica , Infarto del Miocardio/fisiopatología , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Factores de Tiempo
3.
Microsc Res Tech ; 70(1): 50-4, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17094078

RESUMEN

A paracrystalline structure was observed within left ventricular cardiomyocyte nuclei of MLP(-/-) mice. The paracrystal possessed cross lines, approximately 8.0 micro m long and 0.3 micro m wide, with a slender spindle shape and a periodicity of 13 nm. Paracrystals were best observed along the longitudinal orientation of myofibrils and were detected in less than 10% of the nuclei observed. One dimension of the protein unit forming the paracrystal was 8.5 nm long. The electron density of the paracrystal appeared to be slightly higher than that of heterochromatin, suggesting that RNA-associated proteins are constituents of the paracrystal. This is the first report of intranuclear paracrystals in cardiomyocytes, which appear to be unique to MLP(-/-) mice.


Asunto(s)
Cardiomiopatía Dilatada/patología , Núcleo Celular/ultraestructura , Proteínas Musculares/deficiencia , Miocitos Cardíacos/ultraestructura , Animales , Cristalización , Ventrículos Cardíacos/patología , Proteínas con Dominio LIM , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología
4.
Gene Ther ; 10(21): 1814-20, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12960971

RESUMEN

High-efficiency somatic gene transfer in adult mouse heart has not yet been achieved in vivo. Here, we demonstrate high-efficiency in vivo transcoronary gene delivery to the adult murine myocardium using a catheter-based technique with recombinant adenovirus (AdV) and adeno-associated virus (AAV) vectors in normal and genetically engineered mice. The method involves immersion hypothermia followed by transient aortic and pulmonary artery occlusion with proximal intra-aortic segmental injection of cardioplegic solution containing substance P and viral vectors. Gene expression measured using a LacZ marker gene was observed throughout both ventricles. The expression efficiency of a cytoplasmic LacZ marker gene in the left ventricular myocardium was 56.4+/-14.5% (mean+/-s.d.) at 4 days with an AdV vector, and with an AAV vector it was 81.0+/-5.9% at 4 weeks. Following AAV gene transfer, no gene expression was found in kidney, brain, lung, and spleen, but there was slight expression in liver. In addition, we demonstrate temporally controlled genetic manipulation in the heart with an efficiency of 54.6+/-5.2%, by transferring an AdV vector carrying Cre recombinase in ROSA26 flox-LacZ reporter mice. Procedure-related mortality was 16% for AdV and zero for AAV transfer. Thus, this method provides efficient, relatively homogeneous gene expression in both ventricles of the adult mouse heart, and offers a novel approach for conditional gene rescue or ablation in genetically engineered mouse models.


Asunto(s)
Terapia Genética/métodos , Insuficiencia Cardíaca/terapia , Integrasas/genética , Miocardio/metabolismo , Transducción Genética/métodos , Proteínas Virales/genética , Adenoviridae/genética , Animales , Vasos Coronarios , Dependovirus/genética , Expresión Génica , Vectores Genéticos/administración & dosificación , Hipotermia , Inyecciones Intraarteriales , Operón Lac , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
6.
Cell ; 107(6): 801-13, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11747815

RESUMEN

KChIP2, a gene encoding three auxiliary subunits of Kv4.2 and Kv4.3, is preferentially expressed in the adult heart, and its expression is downregulated in cardiac hypertrophy. Mice deficient for KChIP2 exhibit normal cardiac structure and function but display a prolonged elevation in the ST segment on the electrocardiogram. The KChIP2(-/-) mice are highly susceptible to the induction of cardiac arrhythmias. Single-cell analysis revealed a substrate for arrhythmogenesis, including a complete absence of transient outward potassium current, I(to), and a marked increase in action potential duration. These studies demonstrate that a defect in KChIP2 is sufficient to confer a marked genetic susceptibility to arrhythmias, establishing a novel genetic pathway for ventricular tachycardia via a loss of the transmural gradient of I(to).


Asunto(s)
Proteínas de Unión al Calcio/genética , Predisposición Genética a la Enfermedad , Miocardio/metabolismo , Canales de Potasio con Entrada de Voltaje , Potasio/metabolismo , Taquicardia Ventricular/genética , Potenciales de Acción/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Electrocardiografía , Embrión de Mamíferos/metabolismo , Marcación de Gen , Humanos , Hibridación in Situ , Proteínas de Interacción con los Canales Kv , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Datos de Secuencia Molecular , Miocardio/citología , Técnicas de Placa-Clamp , Canales de Potasio/genética , Canales de Potasio/metabolismo , Isoformas de Proteínas , Canales de Potasio Shal , Taquicardia Ventricular/etiología , Taquicardia Ventricular/fisiopatología
7.
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
8.
Arterioscler Thromb Vasc Biol ; 21(4): 548-54, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11304471

RESUMEN

Small GTPase Rho and its target Rho-kinase/ROK/ROCK play an important role in various cellular functions, including smooth muscle contraction, actin cytoskeleton organization, and cell adhesion and migration, all of which may be involved in the pathogenesis of arteriosclerosis. Here, we show that adenovirus-mediated transfer of dominant-negative Rho-kinase (DNRhoK) induces a marked regression of coronary constrictive remodeling and abolishes coronary vasospastic activity in vivo. Porcine coronary segments were chronically treated with interleukin-1beta, which resulted in the development of constrictive remodeling and vasospastic responses to serotonin, as previously reported. Adenovirus-mediated transfer of DNRhoK, but not that of beta-galactosidase, into the interleukin-1beta-treated coronary segment caused a marked regression of the constrictive remodeling and abolished the vasospastic activity in 3 weeks. Western blot analysis showed that the phosphorylation of adducin and the ezrin/radixin/moesin family, the target proteins of Rho-kinase, were upregulated at the coronary lesions and were significantly suppressed by the transfer of DNRHOK: These results indicate that Rho-kinase is substantially involved in coronary constrictive remodeling and vasospastic responses, both of which can be reversed by the selective inhibition of the molecule in our porcine model in vivo.


Asunto(s)
Enfermedad Coronaria/terapia , Terapia Genética/métodos , Proteínas Serina-Treonina Quinasas/genética , Adenoviridae/genética , Animales , Proteínas Sanguíneas/farmacología , Western Blotting , Enfermedad Coronaria/genética , Vasoespasmo Coronario/fisiopatología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/enzimología , Vasos Coronarios/fisiopatología , Proteínas del Citoesqueleto/farmacología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Interleucina-1/farmacología , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/farmacología , Proteínas de Microfilamentos/farmacología , Fosfoproteínas/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Serotonina/farmacología , Porcinos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/genética , beta-Galactosidasa/genética , Quinasas Asociadas a rho
9.
Biochem Biophys Res Commun ; 282(1): 278-83, 2001 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-11264004

RESUMEN

Osteoclast differentiation factor (ODF) induces differentiation of mouse RAW264 cells to mature osteoclasts. To understand the mechanism controlling a coupling between withdrawal from the cell cycle and differentiation, we examined cell cycle progression and expression profiles of cell cycle regulatory genes at the initial phase in committed cells. ODF rapidly converted the hyperphosphorylated form of the retinoblastoma protein (pRb) into the hypophosphorylated form. The p21 protein was induced by ODF treatment in the same time course with that of dephosphorylation of pRb, followed by a sharp decline. After this period, a delayed entry of the S phase started accompanying the induction of CycD3 and cdk6 in differentiating cells. Hydroxyurea treatment indicated that the S phase entry was a prerequisite for osteoclast formation. Thus, ODF induces pleiotropic effects on cell cycle regulatory genes in RAW264 cells during the initial phase of the differentiation process to osteoclasts.


Asunto(s)
Proteínas Portadoras/fisiología , Ciclo Celular/fisiología , Glicoproteínas de Membrana/fisiología , Osteoclastos/citología , Fase S , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular , División Celular/efectos de los fármacos , Línea Celular , Perfilación de la Expresión Génica , Hidroxiurea/farmacología , Ratones , Osteoclastos/efectos de los fármacos , Fosforilación , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Ann N Y Acad Sci ; 947: 407-11, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11795302

RESUMEN

Small GTPase Rho and its target Rho-kinase play an important role in various cellular functions that may be involved in the pathogenesis of arteriosclerosis. Here we show that adenovirus-mediated transfer of dominant-negative Rho-kinase (AdDNRhoK) induces a regression of coronary constrictive remodeling and abolishes coronary vasospastic activity in vivo. Porcine coronary segments were chronically treated with interleukin-1,beta which resulted in the development of constrictive remodeling and vasospastic responses to serotonin in vivo. AdDNRhoK, but not that of beta-galactosidase, into the interleukin-1beta-treated coronary segment caused regression of constrictive remodeling and abolished vasospastic activity in 3 weeks. The unregulated phosphorylation of the target proteins of Rho-kinase at the coronary lesion was significantly suppressed by AdDNRhoK. These results indicate that Rho-kinase is substantially involved in the mechanism of coronary arteriosclerosis, which can be reversed by selective inhibition of the molecule in our porcine model in vivo.


Asunto(s)
Enfermedad de la Arteria Coronaria/terapia , Proteínas Serina-Treonina Quinasas/genética , Animales , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Terapia Genética , Péptidos y Proteínas de Señalización Intracelular , Radiografía , Porcinos , Transfección , Vasoconstricción/fisiología , Quinasas Asociadas a rho
11.
Biochem Biophys Res Commun ; 272(2): 505-12, 2000 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-10833443

RESUMEN

The Enigma homologue protein (ENH), containing an N-terminal PDZ domain and three C-terminal LIM domains, is a heart and skeletal muscle-specific protein that has been shown to preferentially interact with protein kinase C beta (PKCbeta) through the LIM domains (Kuroda et al., J. Biol. Chem. 271, 31029-31032, 1996). We here demonstrate that ENH is colocalized with a cytoskeletal protein alpha-actinin in the Z-disk region of rat neonatal cardiomyocytes. Pull-down assays using the glutathione-S-transferase-fusion system also showed the interaction of the PDZ domain of ENH with actin and alpha-actinin. Furthermore, by combined use of the in silico and conventional cDNA cloning methods, we have isolated three ENH-related clones from a mouse heart-derived cDNA library: mENH1 (591 amino acid residues) corresponding to rat ENH, mENH2 (337 residues), and mENH3 (239 residues); the latter two containing only a single PDZ domain. Deciphering their cDNA sequences, these mENH1-3 mRNAs appear to be generated from a single mENH gene by alternative splicing. Northern blot analyses using human cancer cells and mouse embryos have shown expression of each mENH mRNA to vary considerably among the cell types and during the developmental stage. Together with a recent finding that PKCbeta is markedly activated in the cardiac hypertrophic signaling, these results suggest that ENH1 plays an important role in the heart development by scaffolding PKCbeta to the Z-disk region and that ENH2 and ENH3 negatively modulate the scaffolding activity of ENH1.


Asunto(s)
Actinina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Músculo Esquelético/química , Miocardio/química , Actinas/metabolismo , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Células Cultivadas , Clonación Molecular , Proteínas del Citoesqueleto , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Humanos , Inmunohistoquímica , Proteínas con Dominio LIM , Ratones , Proteínas de Microfilamentos , Datos de Secuencia Molecular , Músculo Esquelético/citología , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Miocardio/citología , Miocardio/metabolismo , Especificidad de Órganos , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia
12.
Am J Physiol Heart Circ Physiol ; 278(6): H1744-50, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10843868

RESUMEN

Restenosis after angioplasty still remains a major problem for which neointimal formation appears to play an important role. Recent studies in vitro suggested that Rho kinase, a target protein of Rho, is important in various cellular functions. We thus examined whether Rho kinase is involved in the restenotic changes after balloon injury. In vivo gene transfer was performed immediately after balloon injury in both sides of the porcine femoral arteries with adenoviral vector encoding either a dominant negative form of Rho kinase (AdDNRhoK) or beta-galactosidase (AdLacZ) as a control. One week after the transfer, immunohistochemistry confirmed the successful gene expression in the vessel wall, whereas 2 wk after the transfer, Western blotting showed the functional upregulation of Rho kinase at the AdLacZ site and its suppression at the AdDNRhoK site. Angiography showed the development of a stenotic lesion at the AdLacZ site where histological neointimal formation was noted, whereas those changes were significantly suppressed at the AdDNRhoK site. These results indicate that Rho kinase is involved in the pathogenesis of neointimal formation after balloon injury in vivo.


Asunto(s)
Cateterismo/efectos adversos , Arteria Femoral/lesiones , Arteria Femoral/fisiopatología , Proteínas Serina-Treonina Quinasas/farmacología , Túnica Íntima/lesiones , Túnica Íntima/fisiopatología , Angiografía , Animales , Western Blotting , Arteria Femoral/diagnóstico por imagen , Arteria Femoral/patología , Técnicas de Transferencia de Gen , Genes Dominantes , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas Serina-Treonina Quinasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Túnica Íntima/diagnóstico por imagen , Túnica Íntima/patología , Quinasas Asociadas a rho
13.
Am J Physiol Heart Circ Physiol ; 278(4): H1362-70, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10749734

RESUMEN

A mutation in the delta-sarcoglycan (SG) gene with absence of delta-SG protein in the heart has been identified in the BIO14.6 cardiomyopathic (CM) hamster, but how the defective gene leads to myocardial degeneration and dysfunction is unknown. We correlated left ventricular (LV) function with increased sarcolemmal membrane permeability and investigated the LV distribution of the dystrophin-dystroglycan complex in BIO14.6 CM hamsters. On echocardiography at 5 wk of age, the CM hamsters showed a mildly enlarged diastolic dimension (LVDD) with decreased LV percent fractional shortening (%FS), and at 9 wk further enlargement of LVDD with reduction of %FS was observed. The percent area of myocardium exhibiting increased membrane permeability or membrane rupture, assessed by Evans blue dye (EBD) staining and wheat germ agglutinin, was greater at 9 than at 5 wk. In areas not stained by EBD, immunostaining of dystrophin was detected in CM hamsters at sarcolemma and T tubules, as expected, but it was also abnormally expressed at the intercalated discs; in addition, the expression of beta-dystroglycan was significantly reduced compared with control hearts. As previously described, alpha-SG was completely deficient in CM hearts compared with control hearts. In myocardial areas showing increased sarcolemmal permeability, neither dystrophin nor beta-dystroglycan could be identified by immunolabeling. Thus, together with the known loss of delta-SG and other SGs, abnormal distribution of dystrophin and reduction of beta-dystroglycan are associated with increased sarcolemmal permeability followed by cell rupture, which correlates with early progressive cardiac dysfunction in the BIO14.6 CM hamster.


Asunto(s)
Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Proteínas del Citoesqueleto/metabolismo , Distrofina/metabolismo , Glicoproteínas de Membrana/metabolismo , Animales , Cardiomiopatías/diagnóstico por imagen , Permeabilidad de la Membrana Celular/fisiología , Colorantes/farmacocinética , Cricetinae , Proteínas del Citoesqueleto/análisis , Modelos Animales de Enfermedad , Distroglicanos , Distrofina/análisis , Ecocardiografía , Azul de Evans/farmacocinética , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Immunoblotting , Masculino , Glicoproteínas de Membrana/análisis , Microscopía Inmunoelectrónica , Miocardio/química , Miocardio/metabolismo , Miocardio/ultraestructura , Sarcoglicanos
14.
Cell ; 99(3): 313-22, 1999 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-10555147

RESUMEN

Dilated cardiomyopathy and end-stage heart failure result in multiple defects in cardiac excitation-contraction coupling. Via complementation of a genetically based mouse model of dilated cardiomyopathy, we now provide evidence that progressive chamber dilation and heart failure are dependent on a Ca2+ cycling defect in the cardiac sarcoplasmic reticulum. The ablation of a muscle-specific sarcoplasmic reticulum Ca2+ ATPase (SERCA2a) inhibitor, phospholamban, rescued the spectrum of phenotypes that resemble human heart failure. Inhibition of phospholamban-SERCA2a interaction via in vivo expression of a phospholamban point mutant dominantly activated the contractility of ventricular muscle cells. Thus, interfering with phospholamban-SERCA2a interaction may provide a novel therapeutic approach for preventing the progression of dilated cardiomyopathy.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Calcio/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Corazón/fisiopatología , Hemodinámica/fisiología , Retículo Sarcoplasmático/enzimología , Animales , Proteínas de Unión al Calcio/deficiencia , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Corazón/fisiología , Hemodinámica/genética , Homeostasis , Humanos , Ratones , Ratones Noqueados , Contracción Miocárdica , Miocardio/patología , Función Ventricular Izquierda
15.
J Biol Chem ; 273(13): 7725-30, 1998 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-9516480

RESUMEN

The assembly of contractile proteins into organized sarcomeric units is one of the most distinctive features of cardiac myocyte hypertrophy. In a well characterized in vitro model system using cultured neonatal rat ventricular myocytes, a subset of G protein-coupled receptor agonists has been shown to induce actin-myosin filament organization. Pretreatment of myocytes with C3 exoenzyme ADP-ribosylated Rho and inhibited the characteristic alpha1-adrenergic receptor agonist-induced myofibrillar organization, suggesting involvement of the Rho GTPase in cardiac myofibrillogenesis. We used adenoviral mediated gene transfer to examine the effects of activated Rho and inhibitory mutants of one of its effectors, Rho kinase, in myocytes. Rho immunoreactivity was increased in the particulate fraction of myocytes infected with a recombinant adenovirus expressing constitutively activated Rho. Rho-infected cells demonstrated a striking increase in the assembly and organization of sarcomeric units and in the expression of the atrial natriuretic factor protein. These Rho-induced responses were markedly inhibited by co-infection with adenoviruses expressing putative dominant negative forms of Rho kinase. A parallel pathway involving Ras-induced myofibrillar organization and atrial natriuretic factor expression was only minimally affected. alpha1-Adrenergic receptor agonist-induced myofibrillogenesis was inhibited by some but not all of the Rho kinase mutants. Our data demonstrate that activated Rho has profound effects on myofibrillar organization in cardiac myocytes and suggest that Rho kinase mediates Rho-induced hypertrophic responses.


Asunto(s)
Toxinas Botulínicas , GTP Fosfohidrolasas/metabolismo , Miocardio/enzimología , Miofibrillas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , ADP Ribosa Transferasas/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Células Cultivadas , Proteínas Activadoras de GTPasa , Péptidos y Proteínas de Señalización Intracelular , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal , Proteínas Activadoras de ras GTPasa , Quinasas Asociadas a rho
16.
J Biol Chem ; 272(9): 5783-91, 1997 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-9038192

RESUMEN

Cardiac myocyte survival is of central importance in the maintenance of the function of heart, as well as in the development of a variety of cardiac diseases. To understand the molecular mechanisms that govern this function, we characterized apoptosis in cardiac muscle cells following serum deprivation. Cardiotrophin 1 (CT-1), a potent cardiac survival factor (Sheng, Z., Pennica, D., Wood, W. I., and Chien, K. R. (1996) Development (Camb.) 122, 419-428), is capable of inhibiting apoptosis in cardiac myocytes. To explore the potential downstream pathways that might be responsible for this effect, we documented that CT-1 activated both signal transducer and activator of transcription 3 (STAT3)- and mitogen-activated protein (MAP) kinase-dependent pathways. The transfection of a MAP kinase kinase 1 (MEK1) dominant negative mutant cDNA into myocardial cells blocked the antiapoptotic effects of CT-1, indicating a requirement of the MAP kinase pathway for the survival effect of CT-1. A MEK-specific inhibitor (PD098059) (Dudley, D. T., Pang, L., Decker, S.-J., Bridges, A. J., and Saltiel, A. R. (1995) Proc. Natl. Acad. Sci. USA 92, 7686-7689) is capable of blocking the activation of MAP kinase, as well as the survival effect of CT-1. In contrast, this inhibitor did not block the activation of STAT3, nor did it have any effect on the hypertrophic response elicited following stimulation of CT-1. Therefore, CT-1 promotes cardiac myocyte survival via the activation of an antiapoptotic signaling pathway that requires MAP kinases, whereas the hypertrophy induced by CT-1 may be mediated by alternative pathways, e.g. Janus kinase/STAT or MEK kinase/c-Jun NH2-terminal protein kinase.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Citocinas/farmacología , Corazón/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Miocardio/citología , Animales , Fragmentación del ADN , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Interleucina-6/farmacología , MAP Quinasa Quinasa 1 , Ratones , Proteína Quinasa 1 Activada por Mitógenos , Mutagénesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Ratas , Factor de Transcripción STAT3 , Transactivadores/metabolismo
17.
Jpn Circ J ; 61(2): 91-118, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9070966

RESUMEN

The past two decades of cardiovascular biology and medicine have been based largely upon the consideration of the heart and vasculature as an integrated physiological system, a view that has resulted in major therapeutic advances. With the advent of developments of gene transfer, mouse and human genetics, genetic engineering of intact animals, and molecular and cellular technology, cardiovascular medicine is now on the threshold of a molecular therapeutic era. Major steps have been taken toward unraveling the molecular determinants of complex, integrative, and polygenic cardiovascular disease states, including atherogenesis, hypertension, cardiac hypertrophy and failure, congenital heart disease, and coronary restenosis following balloon angioplasty. Our improved understanding of the fundamental basis of these important cardiovascular disease processes has established a scientific foundation for diagnostic, prognostic, and therapeutic advances in the mainstream of cardiovascular medicine.


Asunto(s)
Cardiopatías/genética , Animales , Predicción , Terapia Genética , Cardiopatías/terapia , Humanos , Ratones
18.
J Biol Chem ; 271(49): 31185-90, 1996 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-8940118

RESUMEN

G protein-coupled receptor agonists initiate a cascade of signaling events in neonatal rat ventricular myocytes that culminates in changes in gene expression and cell growth characteristic of hypertrophy. These responses have been previously shown to be dependent on Gq and Ras. Rho, a member of the Ras superfamily of GTPases, regulates cytoskeletal rearrangement and transcriptional activation of the c-fos serum response element. Immunofluorescence staining of cardiomyocytes shows that Rho is present and predominantly cytosolic. We used two inhibitors of Rho function, dominant negative N19RhoA and Clostridium botulinum C3 transferase, to examine the possible requirement for Rho in alpha1-adrenergic receptor-mediated hypertrophy. Both inhibitors markedly attenuated atrial natriuretic factor (ANF) reporter gene expression induced by alpha1-adrenergic receptor stimulation with phenylephrine, and virtually abolished the increase in ANF reporter gene expression induced by GTPase-deficient Galphaq. These effects were reproduced with the myosin light chain-2 reporter gene. Notably, N19RhoA did not block the ability of activated Ras to induce ANF and myosin light chain-2 reporter gene expression. Furthermore, activation of the extracellular signal-regulated kinase by phenylephrine was not blocked by N19RhoA, nor was it stimulated by an activated mutant of RhoA. Since activated RhoA and Ras produce a large synergistic effect on ANF-luciferase gene expression, we conclude that Rho functions in a pathway separate from but complementary to Ras. Our results provide direct evidence that Rho is an effector of Galphaq signaling and suggest for the first time that a low molecular weight GTPase other than Ras is involved in regulating myocardial cell growth and gene expression in response to heterotrimeric G protein-linked receptor activation.


Asunto(s)
Antígenos/metabolismo , Toxinas Botulínicas , Proteínas de Unión al GTP/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Miocardio/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , ADP Ribosa Transferasas/metabolismo , Actinas/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Clostridium botulinum/enzimología , Citosol/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Fenilefrina/farmacología , Ratas , Receptores del Factor Natriurético Atrial/genética , Receptores del Factor Natriurético Atrial/metabolismo
19.
FEBS Lett ; 368(2): 343-7, 1995 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-7628634

RESUMEN

In cultured vascular smooth muscle cells (VSMCs), angiotensin II (Ang II) stimulated tyrosine phosphorylation of several proteins including a cluster of 70-80-kDa proteins as assessed by anti-phosphotyrosine immunoblotting. These 70-80-kDa proteins were identified as a focal adhesion-associated protein, paxillin, by anti-paxillin immunoprecipitation. Ang II-stimulated tyrosine phosphorylation of paxillin was detectable within 1 min and maximal at around 10 min and was concentration dependent (half-maximal effect at around 1 nM). Ang II also stimulated tyrosine phosphorylation of focal adhesion kinase in a time- and concentration-dependent manner. The Ang II type 1 (AT1) receptor antagonist, CV-11974, but not the Ang II type 2 receptor antagonist, PD123319, inhibited these reactions. These results indicate that Ang II transduces its signal to focal adhesions via AT1 receptors in cultured VSMCs.


Asunto(s)
Angiotensina II/farmacología , Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Músculo Liso Vascular/fisiología , Fosfoproteínas/metabolismo , Receptores de Angiotensina/fisiología , Transducción de Señal/efectos de los fármacos , Antagonistas de Receptores de Angiotensina , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo , Adhesión Celular/fisiología , Células Cultivadas , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Imidazoles/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Paxillin , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Piridinas/farmacología , Ratas , Tetrazoles/farmacología
20.
Am J Hypertens ; 3(8 Pt 2): 220S-223S, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2121166

RESUMEN

GTP-binding proteins with molecular weights of approximately 20,000 are designated small Mr G proteins. We purified four small Mr G proteins to near homogeneity from bovine brain crude membranes. Properties of these proteins, 20K G, 21K G, 22K G, and 24K G, were then assessed. The cDNA of 22K G and 24K G were cloned, and their nucleotide and amino acid sequences were determined. Three types of both C-terminal sequences and amino acid sequences were identified. We conclude that small Mr G proteins and large Mr G proteins are components of large intracellular regulatory and messenger systems with specific roles in cell function regulation.


Asunto(s)
Proteínas de Unión al GTP/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Química Encefálica , Bovinos , ADN/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/fisiología , Peso Molecular , Poli(ADP-Ribosa) Polimerasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...