Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Circulation ; 126(18): 2208-19, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23019294

RESUMO

BACKGROUND: Heart failure is a growing cause of morbidity and mortality. Cardiac phosphatidylinositol 3-kinase signaling promotes cardiomyocyte survival and function, but it is paradoxically activated in heart failure, suggesting that chronic activation of this pathway may become maladaptive. Here, we investigated the downstream phosphatidylinositol 3-kinase effector, serum- and glucocorticoid-regulated kinase-1 (SGK1), in heart failure and its complications. METHODS AND RESULTS: We found that cardiac SGK1 is activated in human and murine heart failure. We investigated the role of SGK1 in the heart by using cardiac-specific expression of constitutively active or dominant-negative SGK1. Cardiac-specific activation of SGK1 in mice increased mortality, cardiac dysfunction, and ventricular arrhythmias. The proarrhythmic effects of SGK1 were linked to biochemical and functional changes in the cardiac sodium channel and could be reversed by treatment with ranolazine, a blocker of the late sodium current. Conversely, cardiac-specific inhibition of SGK1 protected mice after hemodynamic stress from fibrosis, heart failure, and sodium channel alterations. CONCLUSIONS: SGK1 appears both necessary and sufficient for key features of adverse ventricular remodeling and may provide a novel therapeutic target in cardiac disease.


Assuntos
Cardiomiopatia Dilatada/enzimologia , Insuficiência Cardíaca/enzimologia , Proteínas Imediatamente Precoces/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Remodelação Ventricular/fisiologia , Acetanilidas/uso terapêutico , Animais , Cardiomegalia Induzida por Exercícios , Sequência Consenso , Modelos Animais de Doenças , Eletrocardiografia , Indução Enzimática , Humanos , Hipertensão/complicações , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Piperazinas/uso terapêutico , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ranolazina , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Taquicardia Ventricular/enzimologia , Taquicardia Ventricular/etiologia
2.
Am J Physiol Cell Physiol ; 297(5): C1124-32, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19759331

RESUMO

Myostatin is a highly conserved negative regulator of skeletal muscle growth. Loss of functional myostatin in cattle, mice, sheep, dogs, and humans results in increased muscle mass. The molecular mechanisms responsible for this increase in muscle growth are not fully understood. Previously, we have reported that phenylephrine-induced cardiac muscle growth and Akt activation are enhanced in myostatin knockout mice compared with controls. Here we report that skeletal muscle from myostatin knockout mice show increased Akt protein expression and overall activity at baseline secondary to an increase in Akt mRNA. We examined the functional role of myostatin modulation of Akt in C2C12 myotubes, a well-established in vitro model of skeletal muscle hypertrophy. Adenoviral overexpression of myostatin attenuated the insulin-like growth factor-I (IGF-I)-mediated increase in myotube diameter, as well as IGF-I-stimulated Akt phosphorylation. Inhibition of myostatin by overexpression of the NH(2)-terminal portion of myostatin was sufficient to increase myotube diameter and Akt phosphorylation. Coexpression of myostatin and constitutively active Akt (myr-Akt) restored the increase in myotube diameter. Conversely, expression of dominant negative Akt (dn-Akt) with the inhibitory myostatin propeptide blocked the increase in myotube diameter. Of note, ribosomal protein S6 phosphorylation and atrogin-1/muscle atrophy F box mRNA were increased in skeletal muscle from myostain knockout mice. Together, these data suggest myostatin regulates muscle growth at least in part through regulation of Akt.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Miostatina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Western Blotting , Linhagem Celular , Hipertrofia , Camundongos , Camundongos Knockout , Miostatina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Aging Cell ; 8(5): 573-83, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19663901

RESUMO

Inhibitors of myostatin, a negative regulator of skeletal muscle mass, are being developed to mitigate aging-related muscle loss. Knock-out (KO) mouse studies suggest myostatin also affects adiposity, glucose handling and cardiac growth. However, the cardiac consequences of inhibiting myostatin remain unclear. Myostatin inhibition can potentiate cardiac growth in specific settings (Morissette et al., 2006), a concern because of cardiac hypertrophy is associated with adverse clinical outcomes. Therefore, we examined the systemic and cardiac effects of myostatin deletion in aged mice (27-30 months old). Heart mass increased comparably in both wild-type (WT) and KO mice. Aged KO mice maintained twice as much quadriceps mass as aged WT; however, both groups lost the same percentage (36%) of adult muscle mass. Dual-energy X-ray absorptiometry revealed increased bone density, mineral content, and area in aged KO vs. aged WT mice. Serum insulin and glucose levels were lower in KO mice. Echocardiography showed preserved cardiac function with better fractional shortening (58.1% vs. 49.4%, P = 0.002) and smaller left ventricular diastolic diameters (3.41 vs. 2.71, P = 0.012) in KO vs. WT mice. Phospholamban phosphorylation was increased 3.3-fold in KO hearts (P < 0.05), without changes in total phospholamban, sarco(endo)plasmic reticulum calcium ATPase 2a or calsequestrin. Aged KO hearts showed less fibrosis by Masson's Trichrome staining. Thus, myostatin deletion does not affect aging-related increases in cardiac mass and appears beneficial for bone density, insulin sensitivity and heart function in senescent mice. These results suggest that clinical interventions designed to inhibit skeletal muscle mass loss with aging could have beneficial effects on other organ systems as well.


Assuntos
Envelhecimento/fisiologia , Deleção de Genes , Camundongos/genética , Miostatina/deficiência , Miostatina/genética , Absorciometria de Fóton , Adiponectina/sangue , Animais , Glicemia/metabolismo , Densidade Óssea , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/diagnóstico por imagem , Eletrocardiografia , Coração/anatomia & histologia , Coração/crescimento & desenvolvimento , Coração/fisiologia , Insulina/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos/crescimento & desenvolvimento , Camundongos Knockout , Miostatina/antagonistas & inibidores , Tíbia/anatomia & histologia , Tíbia/crescimento & desenvolvimento
4.
Circ Res ; 99(1): 15-24, 2006 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-16763166

RESUMO

Myostatin is a highly conserved, potent negative regulator of skeletal muscle hypertrophy in many species, from rodents to humans, although its mechanisms of action are incompletely understood. Transcript profiling of hearts from a genetic model of cardiac hypertrophy revealed dramatic upregulation of myostatin, not previously recognized to play a role in the heart. Here we show that myostatin abrogates the cardiomyocyte growth response to phenylephrine in vitro through inhibition of p38 and the serine-threonine kinase Akt, a critical determinant of cell size in many species from drosophila to mammals. Evaluation of male myostatin-null mice revealed that their cardiomyocytes and hearts overall were slightly smaller at baseline than littermate controls but exhibited more exuberant growth in response to chronic phenylephrine infusion. The increased cardiac growth in myostatin-null mice corresponded with increased p38 phosphorylation and Akt activation in vivo after phenylephrine treatment. Together, these data demonstrate that myostatin is dynamically regulated in the heart and acts more broadly than previously appreciated to regulate growth of multiple types of striated muscle.


Assuntos
Divisão Celular/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Camundongos , Camundongos Knockout , Miocárdio/citologia , Miocárdio/metabolismo , Miostatina , Fenilefrina/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
5.
Curr Opin Pharmacol ; 5(2): 165-70, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15780826

RESUMO

Accumulating evidence suggests that apoptosis is not only a common feature of diverse forms of heart failure but also contributes to disease pathogenesis and progression. This contribution of apoptotic signaling to heart failure could reflect not only loss of cardiomyocytes but also dysfunction of surviving cells. The convergence of signaling mechanisms controlling both cardiomyocyte survival and function provides an opportunity for therapeutic strategies that target these pathways. However, significant hurdles must be overcome before the clinical application of these insights becomes possible.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Fármacos Cardiovasculares/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Transdução de Sinais/efeitos dos fármacos
6.
J Mol Cell Cardiol ; 36(4): 481-93, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15081308

RESUMO

The effect of the lysophospholipid, lysophosphatidic acid (LPA), on signaling and hypertrophy of neonatal rat ventricular cardiomyocytes was examined. Myocytes express mRNA for all three G-protein-coupled LPA receptor subtypes (LPA(1)/Edg-2, LPA(2)/Edg-4, and LPA(3)/Edg-7) as indicated by RT-PCR analysis. LPA inhibits isoproterenol-stimulated cyclic AMP accumulation with an IC(50) approximately 40 nM and promotes phosphorylation of ERK-1/2. LPA also elicits a small, slow onset, and activation of phosphoinositide hydrolysis with EC(50) approximately 400 nM, and stimulates a marked increase in the extent of Rho activation. Longer-term treatment with LPA induces a hypertrophic response in myocytes as indicated by increases in cell size, actin organization, ANF staining of the perinuclear region and activation of ANF promoter-luciferase gene expression. Pretreatment of myocytes with pertussis toxin (PTX) not only blocks the capacity of LPA to inhibit cyclic AMP formation and stimulate ERK phosphorylation, but also inhibits hypertrophic changes in cell morphology and ANF-luciferase gene expression. Neither phospholipase C nor Rho activation is PTX sensitive. The hypertrophic effects of LPA on myocytes are also inhibited by treatment with C3 exoenzyme or by transfection of plasmids expressing either C3 exoenzyme or dominant-negative Rho to block Rho function. Inhibition of ERK activation with PD98059 blocks LPA-induced hypertrophy while inhibitors of phospholipase C (U73122), PKC (GF109203X), or p38MAPK (SB203580) do not. These data suggest that LPA induces cardiomyocyte hypertrophy via a pathway different from the conventional G(q) pathway utilized by phenylephrine, endothelin, and PGF2 alpha and involving activation of a PTX-sensitive G(i)/ERK pathway in conjunction with activation of Rho-mediated signals.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Miócitos Cardíacos/patologia , Proteínas rho de Ligação ao GTP/metabolismo , Adenilil Ciclases/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Flavonoides/farmacologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hidrólise , Concentração Inibidora 50 , Luciferases/metabolismo , Lisofosfolipídeos/química , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Toxina Pertussis/farmacologia , Fosfatidilinositóis/química , Biossíntese de Proteínas , Proteína Quinase C/antagonistas & inibidores , Proteínas/química , Pirrolidinonas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção , Fosfolipases Tipo C/farmacologia
7.
J Mol Cell Cardiol ; 35(10): 1217-27, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14519432

RESUMO

During hypertrophy the heart increases its utilization of glucose and decreases that of fatty acids, resuming a fetal pattern of substrate metabolism. As demonstrated here, GLUT1 protein expression is increased in association with in vivo pressure-overload-induced hypertrophy. The relationship of changes in GLUT1 to enhanced glucose uptake and to cardiomyocyte hypertrophy and survival is not known. To explore this question we first examined the effect of prostaglandin F2alpha (PGF2alpha), an established hypertrophic agonist, on GLUT1 expression and glucose uptake in neonatal rat ventricular myocytes (NRVMs). PGF2alpha treatment for 24 h led to a fivefold increase in GLUT1 expression and a sixfold increase in glucose uptake. However, NRVMs cultured in the absence of glucose or with 3-O-methyl glucose, a competitive inhibitor of glucose uptake, still exhibited PGF2alpha-induced hypertrophic growth. In addition, we determined that overexpression of GLUT1 using adenovirus was insufficient to cause an increase in cell size, myofibrillar organization, or atrial natriuretic factor (ANF) expression. On the other hand, adenoviral overexpression of antisense GLUT1 (which blocked PGF2alpha-induced increases in GLUT1 protein) prevented PGF2alpha-stimulated cell enlargement and increases in ANF transcription. Overexpression of GLUT1 or addition of PGF2alpha also protected cells against serum deprivation-induced apoptosis; this effect was blocked by antisense GLUT1 but, surprisingly, was not dependent on glucose. Together, these data suggest that upregulation of GLUT1 serves a role in agonist-induced hypertrophy and survival which can be dissociated from its role in glucose transport.


Assuntos
Proteínas de Transporte de Monossacarídeos/biossíntese , Miócitos Cardíacos/citologia , Regulação para Cima , Adenoviridae/genética , Animais , Aorta/patologia , Apoptose , Fator Natriurético Atrial/biossíntese , Transporte Biológico , Western Blotting , Sobrevivência Celular , Células Cultivadas , Dinoprosta/metabolismo , Ensaio de Imunoadsorção Enzimática , Glucose/metabolismo , Glucose/farmacocinética , Transportador de Glucose Tipo 1 , Hipertrofia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transporte de Monossacarídeos/metabolismo , Células Musculares/citologia , Oligonucleotídeos Antissenso/farmacologia , Pressão , Ratos , Ratos Sprague-Dawley , Frações Subcelulares , Fatores de Tempo
8.
J Biol Chem ; 277(2): 1261-7, 2002 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-11694533

RESUMO

The delta isoform of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) predominates in the heart. To investigate the role of CaMKII in cardiac function, we made transgenic (TG) mice that express the nuclear delta(B) isoform of CaMKII. The expressed CaMKIIdelta(B) transgene was restricted to the myocardium and highly concentrated in the nucleus. Cardiac hypertrophy was evidenced by an increased left ventricle to body weight ratio and up-regulation of embryonic and contractile protein genes including atrial natriuretic factor, beta-myosin heavy chain, and alpha-skeletal actin. Echocardiography revealed ventricular dilation and decreased cardiac function, which was also observed in hemodynamic measurements from CaMKIIdelta(B) TG mice. Surprisingly, phosphorylation of phospholamban at both Thr(17) and Ser(16) was significantly decreased in the basal state as well as upon adrenergic stimulation. This was associated with diminished sarcoplasmic reticulum Ca(2+) uptake in vitro and altered relaxation properties in vivo. The activity and expression of protein phosphatase 2A were both found to be increased in CaMKII TG mice, and immunoprecipitation studies indicated that protein phosphatase 2A directly associates with CaMKII. Our findings are the first to demonstrate that CaMKII can induce hypertrophy and dilation in vivo and indicate that compensatory increases in phosphatase activity contribute to the resultant phenotype.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cardiomegalia/fisiopatologia , Cardiomiopatia Dilatada/fisiopatologia , Miocárdio/enzimologia , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Cardiomegalia/patologia , Cardiomiopatia Dilatada/patologia , Dobutamina/farmacologia , Ecocardiografia , Coração/efeitos dos fármacos , Coração/fisiologia , Hemodinâmica , Imuno-Histoquímica , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Proteína Fosfatase 2 , Retículo Sarcoplasmático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...