Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Proc Natl Acad Sci U S A ; 105(34): 12457-62, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18711143

RESUMO

G protein-coupled receptor (GPCR) kinases (GRKs) are critical regulators of cellular signaling and function. In cardiomyocytes, GRK2 and GRK5 are two GRKs important for myocardial regulation, and both have been shown to be up-regulated in the dysfunctional heart. We report that increased levels and activity of GRK5 in failing myocardium may have unique significance due to its nuclear localization, a property not shared by GRK2. We find that transgenic mice with elevated cardiac GRK5 levels have exaggerated hypertrophy and early heart failure compared with control mice after pressure overload. This pathology is not present in cardiac GRK2-overexpressing mice or in mice with overexpression of a mutant GRK5 that is excluded from the nucleus. Nuclear accumulation of GRK5 is enhanced in myocytes after aortic banding in vivo and in vitro in myocytes after increased G alpha q activity, the trigger for pressure-overload hypertrophy. GRK5 enhances activation of MEF2 in concert with Gq signals, demonstrating that nuclear localized GRK5 regulates gene transcription via a pathway critically linked to myocardial hypertrophy. Mechanistically, we show that this is due to GRK5 acting, in a non-GPCR manner, as a class II histone deacetylase (HDAC) kinase because it can associate with and phosphorylate the myocyte enhancer factor-2 repressor, HDAC5. Moreover, significant HDAC activity can be found with GRK5 in the heart. Our data show that GRK5 is a nuclear HDAC kinase that plays a key role in maladaptive cardiac hypertrophy apparently independent of any action directly on GPCRs.


Assuntos
Núcleo Celular/enzimologia , Quinase 5 de Receptor Acoplado a Proteína G/fisiologia , Miócitos Cardíacos/enzimologia , Animais , Quinase 5 de Receptor Acoplado a Proteína G/análise , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/etiologia , Histona Desacetilases/metabolismo , Hipertrofia/enzimologia , Hipertrofia/etiologia , Fatores de Transcrição MEF2 , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/ultraestrutura , Fatores de Regulação Miogênica/metabolismo , Regulação para Cima
2.
Circulation ; 117(11): 1378-87, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18316484

RESUMO

BACKGROUND: A salient characteristic of dysfunctional myocardium progressing to heart failure is an upregulation of the adenylyl cyclase inhibitory guanine nucleotide (G) protein alpha subunit, G alpha(i2). It has not been determined conclusively whether increased Gi activity in the heart is beneficial or deleterious in vivo. Gi signaling has been implicated in the mechanism of cardioprotective agents; however, no in vivo evidence exists that any of the G alpha subunits are cardioprotective. We have created a novel molecular tool to specifically address the role of Gi proteins in normal and dysfunctional myocardium. METHODS AND RESULTS: We have developed a class-specific Gi inhibitor peptide, GiCT, composed of the region of G alpha(i2) that interacts specifically with G protein-coupled receptors. GiCT inhibits Gi signals specifically in vitro and in vivo, whereas Gs and Gq signals are not affected. In vivo expression of GiCT in transgenic mice effectively causes a "functional knockout" of cardiac G alpha(i2) signaling. Inducible, cardiac-specific GiCT transgenic mice display a baseline phenotype consistent with nontransgenic mice. However, when subjected to ischemia/reperfusion injury, GiCT transgenic mice demonstrate a significant increase in infarct size compared with nontransgenic mice (from 36.9+/-2.5% to 50.9+/-4.3%). Mechanistically, this post-ischemia/reperfusion phenotype includes increased myocardial apoptosis and resultant decreased contractile performance. CONCLUSIONS: Overall, our results demonstrate the in vivo utility of GiCT to dissect specific mechanisms attributed to Gi signaling in stressed myocardium. Our results with GiCT indicate that upregulation of G alpha(i2) is an adaptive protective response after ischemia to shield myocytes from apoptosis.


Assuntos
Apoptose/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Mitocôndrias Cardíacas/fisiologia , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Humanos , Isoproterenol/farmacologia , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Estresse Oxidativo , Fragmentos de Peptídeos/genética , Ratos , Receptores Acoplados a Proteínas G/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Transdução Genética
3.
Circulation ; 114(21): 2240-50, 2006 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-17088462

RESUMO

BACKGROUND: Both the A1- and A3-adenosine receptors (ARs) have been implicated in mediating the cardioprotective effects of adenosine. Paradoxically, overexpression of both A1-AR and A3-AR is associated with changes in the cardiac phenotype. To evaluate the temporal relationship between AR signaling and cardiac remodeling, we studied the effects of controlled overexpression of the A1-AR using a cardiac-specific and tetracycline-transactivating factor-regulated promoter. METHODS AND RESULTS: Constitutive A1-AR overexpression caused the development of cardiac dilatation and death within 6 to 12 weeks. These mice developed diminished ventricular function and decreased heart rate. In contrast, when A1-AR expression was delayed until 3 weeks of age, mice remained phenotypically normal at 6 weeks, and >90% of the mice survived at 30 weeks. However, late induction of A1-AR still caused mild cardiomyopathy at older ages (20 weeks) and accelerated cardiac hypertrophy and the development of dilatation after pressure overload. These changes were accompanied by gene expression changes associated with cardiomyopathy and fibrosis and by decreased Akt phosphorylation. Discontinuation of A1-AR induction mitigated cardiac dysfunction and significantly improved survival rate. CONCLUSIONS: These data suggest that robust constitutive myocardial A1-AR overexpression induces a dilated cardiomyopathy, whereas delaying A1-AR expression until adulthood ameliorated but did not eliminate the development of cardiac pathology. Thus, the inducible A1-AR transgenic mouse model provides novel insights into the role of adenosine signaling in heart failure and illustrates the potentially deleterious consequences of selective versus nonselective activation of adenosine-signaling pathways in the heart.


Assuntos
Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/fisiopatologia , Ecocardiografia , Coração/fisiopatologia , Miocárdio/patologia , Receptor A1 de Adenosina/metabolismo , Animais , Cardiomegalia/etiologia , Cardiomiopatia Dilatada/mortalidade , Cardiomiopatia Dilatada/patologia , Doxiciclina/farmacologia , Eletrocardiografia , Fibrose , Expressão Gênica , Frequência Cardíaca , Humanos , Hipertensão/etiologia , Hipertensão/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor A1 de Adenosina/genética , Análise de Sobrevida , Fatores de Tempo
4.
J Clin Invest ; 114(11): 1550-63, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15578088

RESUMO

Cardiac-restricted overexpression of the Ca2+-binding protein S100A1 has been shown to lead to increased myocardial contractile performance in vitro and in vivo. Since decreased cardiac expression of S100A1 is a characteristic of heart failure, we tested the hypothesis that S100A1 gene transfer could restore contractile function of failing myocardium. Adenoviral S100A1 gene delivery normalized S100A1 protein expression in a postinfarction rat heart failure model and reversed contractile dysfunction of failing myocardium in vivo and in vitro. S100A1 gene transfer to failing cardiomyocytes restored diminished intracellular Ca2+ transients and sarcoplasmic reticulum (SR) Ca2+ load mechanistically due to increased SR Ca2+ uptake and reduced SR Ca2+ leak. Moreover, S100A1 gene transfer decreased elevated intracellular Na+ concentrations to levels detected in nonfailing cardiomyocytes, reversed reactivated fetal gene expression, and restored energy supply in failing cardiomyocytes. Intracoronary adenovirus-mediated S100A1 gene delivery in vivo to the postinfarcted failing rat heart normalized myocardial contractile function and Ca2+ handling, which provided support in a physiological context for results found in myocytes. Thus, the present study demonstrates that restoration of S100A1 protein levels in failing myocardium by gene transfer may be a novel therapeutic strategy for the treatment of heart failure.


Assuntos
Adenoviridae/genética , Proteínas de Ligação ao Cálcio , Baixo Débito Cardíaco/terapia , Terapia Genética/métodos , Coração/fisiologia , Miocárdio/metabolismo , Adenoviridae/metabolismo , Animais , Células COS , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Chlorocebus aethiops , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Transferência de Genes , Vetores Genéticos , Coração/anatomia & histologia , Hemodinâmica , Humanos , Masculino , Contração Miocárdica , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miocárdio/citologia , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas S100 , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
5.
PLoS One ; 8(3): e57324, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23472081

RESUMO

G protein-Coupled Receptors (GPCRs) kinases (GRKs) play a crucial role in regulating cardiac hypertrophy. Recent data from our lab has shown that, following ventricular pressure overload, GRK5, a primary cardiac GRK, facilitates maladaptive myocyte growth via novel nuclear localization. In the nucleus, GRK5's newly discovered kinase activity on histone deacetylase 5 induces hypertrophic gene transcription. The mechanisms governing the nuclear targeting of GRK5 are unknown. We report here that GRK5 nuclear accumulation is dependent on Ca(2+)/calmodulin (CaM) binding to a specific site within the amino terminus of GRK5 and this interaction occurs after selective activation of hypertrophic Gq-coupled receptors. Stimulation of myocytes with phenylephrine or angiotensinII causes GRK5 to leave the sarcolemmal membrane and accumulate in the nucleus, while the endothelin-1 does not cause nuclear GRK5 localization. A mutation within the amino-terminus of GRK5 negating CaM binding attenuates GRK5 movement from the sarcolemma to the nucleus and, importantly, overexpression of this mutant does not facilitate cardiac hypertrophy and related gene transcription in vitro and in vivo. Our data reveal that CaM binding to GRK5 is a physiologically relevant event that is absolutely required for nuclear GRK5 localization downstream of hypertrophic stimuli, thus facilitating GRK5-dependent regulation of maladaptive hypertrophy.


Assuntos
Calmodulina/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação Enzimológica da Expressão Gênica , Miocárdio/metabolismo , Transporte Ativo do Núcleo Celular , Adenoviridae/metabolismo , Angiotensina II/metabolismo , Animais , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Hipertrofia , Ligantes , Masculino , Camundongos , Camundongos Transgênicos , Fenilefrina/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Coelhos , Ratos
6.
Nat Med ; 14(5): 510-7, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425130

RESUMO

Beta-adrenergic receptor (betaAR) blockade is a standard therapy for cardiac failure and ischemia. G protein-coupled receptor kinases (GRKs) desensitize betaARs, suggesting that genetic GRK variants might modify outcomes in these syndromes. Re-sequencing of GRK2 and GRK5 revealed a nonsynonymous polymorphism of GRK5, common in African Americans, in which leucine is substituted for glutamine at position 41. GRK5-Leu41 uncoupled isoproterenol-stimulated responses more effectively than did GRK5-Gln41 in transfected cells and transgenic mice, and, like pharmacological betaAR blockade, GRK5-Leu41 protected against experimental catecholamine-induced cardiomyopathy. Human association studies showed a pharmacogenomic interaction between GRK5-Leu41 and beta-blocker treatment, in which the presence of the GRK5-Leu41 polymorphism was associated with decreased mortality in African Americans with heart failure or cardiac ischemia. In 375 prospectively followed African-American subjects with heart failure, GRK5-Leu41 protected against death or cardiac transplantation. Enhanced betaAR desensitization of excessive catecholamine signaling by GRK5-Leu41 provides a 'genetic beta-blockade' that improves survival in African Americans with heart failure, suggesting a reason for conflicting results of beta-blocker clinical trials in this population.


Assuntos
Quinase 5 de Receptor Acoplado a Proteína G/genética , Insuficiência Cardíaca/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/genética , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Negro ou Afro-Americano/genética , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Frequência do Gene , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Farmacogenética/métodos , Estudos Prospectivos , Análise de Sequência de DNA , Estados Unidos , População Branca/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa