Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Circ Res ; 109(2): 161-71, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21617130

RESUMEN

RATIONALE: Overexpression of muscle atrophy F-box (MAFbx/atrogin-1), an E3 ubiquitin ligase, induces proteasomal degradation in cardiomyocytes. The role of endogenous MAFbx in regulating cardiac hypertrophy and failure remains unclear. OBJECTIVE: We investigated the role of MAFbx in regulating cardiac hypertrophy and function in response to pressure overload. Transverse aortic constriction (TAC) was applied to MAFbx knockout (KO) and wild-type (WT) mice. METHODS AND RESULTS: Expression of MAFbx in WT mice was significantly increased by TAC. TAC-induced increases in cardiac hypertrophy were significantly smaller in MAFbx KO than in WT mice. There was significantly less lung congestion and interstitial fibrosis in MAFbx KO than in WT mice. MAFbx KO also inhibited ß-adrenergic cardiac hypertrophy. DNA microarray analysis revealed that activation of genes associated with the transcription factor binding site for the nuclear factor-κB family were inhibited in MAFbx KO mice compared with WT mice after TAC. Although the levels of IκB-α were significantly decreased after TAC in WT mice, they were increased in MAFbx KO mice. MAFbx regulates ubiquitination and proteasomal degradation of IκB-α in cardiomyocytes. In primary cultured rat cardiomyocytes, phenylephrine-induced activation of nuclear factor-κB and hypertrophy were significantly suppressed by MAFbx knockdown but were partially rescued by overexpression of nuclear factor-κB p65. CONCLUSIONS: MAFbx plays an essential role in mediating cardiac hypertrophy in response to pressure overload. Downregulation of MAFbx inhibits cardiac hypertrophy in part through stabilization of IκB-α and inactivation of nuclear factor-κB. Taken together, inhibition of MAFbx attenuates pathological hypertrophy, thereby protecting the heart from progression into heart failure.


Asunto(s)
Cardiomegalia/metabolismo , Proteínas Musculares/fisiología , FN-kappa B/metabolismo , Proteínas Ligasas SKP Cullina F-box/fisiología , Animales , Cardiomegalia/etiología , Células Cultivadas , Constricción Patológica , Expresión Génica , Regulación de la Expresión Génica/fisiología , Proteínas I-kappa B/metabolismo , Ratones , Ratones Noqueados , Proteínas Musculares/deficiencia , Proteínas Musculares/metabolismo , Inhibidor NF-kappaB alfa , Sustancias Protectoras , Ratas , Proteínas Ligasas SKP Cullina F-box/deficiencia , Proteínas Ligasas SKP Cullina F-box/metabolismo
2.
Circulation ; 124(4): 406-15, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21747053

RESUMEN

BACKGROUND: Cardiac overload, a major cause of heart failure, induces the expression of the heat shock protein H11 kinase/Hsp22 (Hsp22). METHODS AND RESULTS: To determine the specific function of Hsp22 in that context, a knockout mouse model of Hsp22 deletion was generated. Although comparable to wild-type mice in basal conditions, knockout mice exposed to pressure overload developed less hypertrophy and showed ventricular dilation, impaired contractile function, increased myocyte length and accumulation of interstitial collagen, faster transition into heart failure, and increased mortality. Microarrays revealed that hearts from knockout mice failed to transactivate genes regulated by the transcription factor STAT3. Accordingly, nuclear STAT3 tyrosine phosphorylation was decreased in knockout mice. Silencing and overexpression experiments in isolated neonatal rat cardiomyocytes showed that Hsp22 activates STAT3 via production of interleukin-6 by the transcription factor nuclear factor-κB. In addition to its transcriptional function, STAT3 translocates to the mitochondria where it increases oxidative phosphorylation. Both mitochondrial STAT3 translocation and respiration were also significantly decreased in knockout mice. CONCLUSIONS: This study found that Hsp22 represents a previously undescribed activator of both nuclear and mitochondrial functions of STAT3, and its deletion in the context of pressure overload in vivo accelerates the transition into heart failure and increases mortality.


Asunto(s)
Eliminación de Gen , Proteínas del Choque Térmico HSP20/genética , Insuficiencia Cardíaca/genética , Mitocondrias Cardíacas/genética , Proteínas Musculares/genética , Factor de Transcripción STAT3/genética , Animales , Cardiomegalia/enzimología , Cardiomegalia/genética , Núcleo Celular/enzimología , Núcleo Celular/genética , Células Cultivadas , Colágeno/metabolismo , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/mortalidad , Proteínas de Choque Térmico , Interleucina-6/biosíntesis , Masculino , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/enzimología , Chaperonas Moleculares , Miocitos Cardíacos/enzimología , FN-kappa B/metabolismo , Fosforilación Oxidativa , Ratas
3.
J Biol Chem ; 285(26): 20281-90, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20404348

RESUMEN

MicroRNA-21 (miR-21) is highly up-regulated during hypertrophic and cancerous cell growth. In contrast, we found that it declines in cardiac myocytes upon exposure to hypoxia. Thus, the objective was to explore its role during hypoxia. We show that miR-21 not only regulates phosphatase and tensin homologue deleted on chromosome 10 (PTEN), but also targets Fas ligand (FasL). During prolonged hypoxia, down-regulation of miR-21 proved necessary and sufficient for enhancing expression of both proteins. We demonstrate here for the first time that miR-21 is positively regulated via an AKT-dependent pathway, which is depressed during prolonged hypoxia. Accordingly, hypoxia-induced down-regulation of miR-21 and up-regulation of FasL and PTEN were reversed by activated AKT and reproduced by a dominant negative mutant, wortmannin, or PTEN. Moreover, the antiapoptotic function of AKT partly required miR-21, which was sufficient for inhibition of caspase-8 activity and mitochondrial damage. In consensus, overexpression of miR-21 in a transgenic mouse heart resulted in suppression of ischemia-induced up-regulation of PTEN and FasL expression, an increase in phospho-AKT, a smaller infarct size, and ameliorated heart failure. Thus, we have identified a unique aspect of the function of AKT by which it inhibits apoptosis through miR-21-dependent suppression of FasL.


Asunto(s)
Apoptosis , Proteína Ligando Fas/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regiones no Traducidas 3'/genética , Animales , Animales Recién Nacidos , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Células Cultivadas , Proteína Ligando Fas/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , MicroARNs/genética , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN , Ratas , Ratas Sprague-Dawley
4.
Proc Natl Acad Sci U S A ; 105(52): 20900-5, 2008 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-19106302

RESUMEN

Glycogen synthase kinase-3 (GSK-3) is a master regulator of growth and death in cardiac myocytes. GSK-3 is inactivated by hypertrophic stimuli through phosphorylation-dependent and -independent mechanisms. Inactivation of GSK-3 removes the negative constraint of GSK-3 on hypertrophy, thereby stimulating cardiac hypertrophy. N-terminal phosphorylation of the GSK-3 isoforms GSK-3alpha and GSK-3beta by upstream kinases (e.g., Akt) is a major mechanism of GSK-3 inhibition. Nonetheless, its role in mediating cardiac hypertrophy and failure remains to be established. Here we evaluated the role of Serine(S)21 and S9 phosphorylation of GSK-3alpha and GSK-3beta in the regulation of cardiac hypertrophy and function during pressure overload (PO), using GSK-3alpha S21A knock-in (alphaKI) and GSK-3beta S9A knock-in (betaKI) mice. Although inhibition of S9 phosphorylation during PO in the betaKI mice attenuated hypertrophy and heart failure (HF), inhibition of S21 phosphorylation in the alphaKI mice unexpectedly promoted hypertrophy and HF. Inhibition of S21 phosphorylation in GSK-3alpha, but not of S9 phosphorylation in GSK-3beta, caused phosphorylation and down-regulation of G1-cyclins, due to preferential localization of GSK-3alpha in the nucleus, and suppressed E2F and markers of cell proliferation, including phosphorylated histone H3, under PO, thereby contributing to decreases in the total number of myocytes in the heart. Restoration of the E2F activity by injection of adenovirus harboring cyclin D1 with a nuclear localization signal attenuated HF under PO in the alphaKI mice. Collectively, our results reveal that whereas S9 phosphorylation of GSK-3beta mediates pathological hypertrophy, S21 phosphorylation of GSK-3alpha plays a compensatory role during PO, in part by alleviating the negative constraint on the cell cycle machinery in cardiac myocytes.


Asunto(s)
Presión Sanguínea , Glucógeno Sintasa Quinasa 3/metabolismo , Miocardio/enzimología , Miocitos Cardíacos/enzimología , Animales , Presión Sanguínea/genética , Cardiomegalia/enzimología , Cardiomegalia/genética , Cardiomegalia/patología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina G , Ciclina G1 , Ciclinas/genética , Ciclinas/metabolismo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Técnicas de Sustitución del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos/patología , Fosforilación/genética
5.
Am J Physiol Heart Circ Physiol ; 299(3): H707-12, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20562336

RESUMEN

Adenylyl cyclase (AC) type 5 (AC5) and AC type 6 (AC6) are the two major AC isoforms in the heart. Cardiac overexpression of AC6 has been shown to be protective in response to several interventions. In this investigation, we examined the effects of chronic pressure overload in AC6 transgenic (TG) mice. In the absence of any stress, AC6 TG mice exhibited enhanced contractile function compared with their wild-type (WT) littermates, i.e., increased (P < 0.05) left ventricular (LV) ejection fraction (EF) (75 +/- 0.9 vs. 71 +/- 0.5%) and LV dP/dt (7,850 +/- 526 vs. 6,374 +/- 315 mmHg/s). Forskolin (25 microg x kg(-1) x min(-1) for 5 min) increased LVEF more (P < 0.05) in AC6 TG mice (14.8 +/- 1.0%) than in WT mice (7.7 +/- 1.0%). Also, isoproterenol (0.04 microg x kg(-1) x min(-1) for 5 min) increased LVEF more (P < 0.05) in AC6 TG mice (18.0 +/- 1.2%) than in WT mice (11.6 +/- 2.1%). Pressure overload, induced by 4 wk of transverse aortic constriction (TAC), increased the LV weight-to-body weight ratio and myocyte cross-sectional area similarly in both groups, but reduced LVEF more in AC6 TG mice (22%) compared with WT mice (9%), despite the higher starting level of LVEF in AC6 TG mice. LV systolic wall stress increased more in AC6 TG mice than in WT mice, which could be responsible for the reduced LVEF in AC6 TG mice with chronic pressure overload. In addition, LV dP/dt was no longer elevated in AC6 TG mice after TAC compared with WT mice. LV end-diastolic diameter was also greater (P < 0.05) in AC6 TG mice (3.8 +/- 0.07 mm) than in WT mice (3.6 +/- 0.05 mm) after TAC. Thus, in contrast to other interventions previously reported to be salutary with cardiac AC6 overpression, the response to chronic pressure overload was not; actually, AC6 TG mice fared worse than WT mice. The mechanism may be due to the increased LV systolic wall stress in AC6 TG mice with chronic pressure overload.


Asunto(s)
Adenilil Ciclasas/metabolismo , Ventrículos Cardíacos/metabolismo , Corazón/fisiopatología , Hipertrofia Ventricular Izquierda/metabolismo , Miocardio/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Adenilil Ciclasas/genética , Análisis de Varianza , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Colforsina/farmacología , Ecocardiografía , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Hemodinámica , Hipertrofia Ventricular Izquierda/fisiopatología , Isoproterenol , Ratones , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Estrés Fisiológico/fisiología , Disfunción Ventricular Izquierda/fisiopatología
6.
Circ Res ; 103(11): 1309-18, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18927464

RESUMEN

Mammalian sterile 20-like kinase (Mst)1 plays an important role in mediating apoptosis and inhibiting hypertrophy in the heart. Because Hippo, a Drosophila homolog of Mst1, forms a signaling complex with Warts, a serine/threonine kinase, which in turn stimulates cell death and inhibits cell proliferation, mammalian homologs of Warts, termed Lats1 and Lats2, may mediate the function of Mst1. We here show that Lats2, but not Lats1, dose-dependently increased apoptosis in cultured cardiac myocytes. Lats2 also dose-dependently reduced [(3)H]phenylalanine incorporation and cardiac myocyte size, whereas dominant negative Lats2 (DN-Lats2) increased them, suggesting that endogenous Lats2 negatively regulates myocyte growth. DN-Lats2 significantly attenuated induction of apoptosis and inhibition of hypertrophy by Mst1, indicating that Lats2 mediates the function of Mst1 in cardiac myocytes. Cardiac specific overexpression of Lats2 in transgenic mice significantly reduced the size of left and right ventricles, whereas that of DN-Lats2 caused hypertrophy in both ventricles. Overexpression of Lats2 reduced left ventricular systolic and diastolic function without affecting baseline levels of myocardial apoptosis. Expression of endogenous Lats2 was significantly upregulated in response to transverse aortic constriction. Overexpression of DN-Lats2 significantly enhanced cardiac hypertrophy and inhibited cardiac myocyte apoptosis induced by transverse aortic constriction. These results suggest that Lats2 is necessary and sufficient for negatively regulating ventricular mass in the heart. Although Lats2 is required for cardiac myocyte apoptosis in response to pressure overload, it was not sufficient to induce apoptosis at baseline. In conclusion, Lats2 affects both growth and death of cardiac myocytes, but it primarily regulates the size of the heart and acts as an endogenous negative regulator of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/prevención & control , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Adenoviridae/genética , Análisis de Varianza , Animales , Apoptosis , Tamaño de la Célula , Células Cultivadas , Fragmentación del ADN , Genes Dominantes , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/deficiencia , ARN Mensajero/genética , Proteínas Supresoras de Tumor/deficiencia
7.
Am J Physiol Heart Circ Physiol ; 297(5): H1814-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19717730

RESUMEN

Calcineurin is a Ca(2+)/calmodulin-dependent protein phosphatase that induces myocardial growth in response to several physiological and pathological stimuli. Calcineurin inhibition, induced either via cyclosporine or genetically, can decrease myocardial hypertrophy secondary to pressure overload without affecting left ventricular (LV) systolic function. Since hypertrophy can also affect LV diastolic function, the goal of this study was to examine the effects of chronic pressure overload (2 wk aortic banding) in transgenic (Tg) mice overexpressing Zaki-4beta (TgZ), a specific endogenous inhibitor of calcineurin, on LV diastolic function. As expected, in the TgZ mice with calcineurin inhibitor overexpression, aortic banding reduced the degree of LV hypertrophy, as assessed by LV weight-to-body weight ratio (3.5 + or - 0.1) compared with that in non-Tg mice (4.6 + or - 0.2). LV systolic function remained compensated in both groups with pressure overload. However, the LV end-diastolic stress-to-LV end-diastolic dimension ratio, an index of diastolic stiffness and LV pressure half-time and isovolumic relaxation time, two indexes of isovolumic relaxation, increased significantly more in TgZ mice with aortic banding. Protein levels of phosphorylated phospholamban (PS16), sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, phosphorylated ryanodine receptor, and the Na(+)/Ca(2+) exchanger were also reduced significantly (P < 0.05) in the banded TgZ mice. As expected, genetic calcineurin inhibition inhibited the development of LV hypertrophy with chronic pressure overload but also induced LV diastolic dysfunction, as reflected by both impaired isovolumic relaxation and increased myocardial stiffness. Thus genetic calcineurin inhibition reveals a new mechanism regulating LV diastolic function.


Asunto(s)
Inhibidores de la Calcineurina , Diástole , Hipertrofia Ventricular Izquierda/prevención & control , Miocardio/enzimología , Proteínas/metabolismo , Disfunción Ventricular Izquierda/enzimología , Animales , Aorta/cirugía , Calcineurina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diástole/genética , Modelos Animales de Enfermedad , Elasticidad , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Péptidos y Proteínas de Señalización Intracelular , Ligadura , Ratones , Ratones Transgénicos , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Fosforilación , Proteínas/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Volumen Sistólico , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Presión Ventricular
8.
Circ Res ; 100(3): 416-24, 2007 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-17234972

RESUMEN

MicroRNAs are naturally existing, small, noncoding RNA molecules that downregulate posttranscriptional gene expression. Their expression pattern and function in the heart remain unknown. Here we report an array of microRNAs that are differentially and temporally regulated during cardiac hypertrophy. Significantly, the muscle-specific microRNA-1 (miR-1) was singularly downregulated as early as day 1 (0.56+/-0.036), persisting through day 7 (0.29+/-0.14), after aortic constriction-induced hypertrophy in a mouse model. Overexpression experiments showed that miR-1 inhibited its in silico-predicted, growth-related targets, including Ras GTPase-activating protein (RasGAP), cyclin-dependent kinase 9 (Cdk9), fibronectin, and Ras homolog enriched in brain (Rheb), in addition to protein synthesis and cell size. Thus, we propose that microRNAs play an essential regulatory role in the development of cardiac hypertrophy, wherein downregulation of miR-1 is necessary for the relief of growth-related target genes from its repressive influence and induction of hypertrophy.


Asunto(s)
Cardiomiopatía Hipertrófica/etiología , Regulación de la Expresión Génica , MicroARNs/fisiología , Animales , Estenosis de la Válvula Aórtica/complicaciones , Northern Blotting , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , División Celular/efectos de los fármacos , Tamaño de la Célula , Células Cultivadas/metabolismo , Constricción , Medios de Cultivo/farmacología , Medio de Cultivo Libre de Suero/farmacología , Citomegalovirus/genética , Progresión de la Enfermedad , Regulación hacia Abajo , Perfilación de la Expresión Génica , Vectores Genéticos/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , MicroARNs/clasificación , MicroARNs/genética , MicroARNs/aislamiento & purificación , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Polimerasa III/fisiología , ARN Nuclear Pequeño/fisiología , Ratas , Ratas Sprague-Dawley
9.
Circ Res ; 100(9): 1344-52, 2007 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-17395874

RESUMEN

Mammalian sterile 20-like kinase-1 (Mst1) plays an important role in mediating cardiac myocyte apoptosis in response to ischemia/reperfusion. Whether or not Mst1 is also involved in the long-term development of heart failure after myocardial infarction (MI) is unknown. We addressed this issue using transgenic mice with cardiac specific overexpression of dominant negative Mst1 (Tg-DN-Mst1). The left coronary artery was permanently ligated, and the size of MI was similar between Tg-DN-Mst1 and nontransgenic controls (NTg). After 4 weeks, Mst1 was significantly activated in the remodeling area in NTg, but not in Tg-DN-Mst1. Although left ventricular (LV) enlargement was significantly attenuated in Tg-DN-Mst1 compared with NTg, neither LV weight/body weight nor myocyte cross sectional area was statistically different between Tg-DN-Mst1 and NTg. LV ejection fraction was significantly greater in Tg-DN-Mst1 than in NTg (53 versus 38%, P<0.01), whereas LV end-diastolic pressure (6 versus 12 mm Hg, P<0.05) and lung weight/body weight (9.8 versus 12.2 P<0.05) were significantly smaller in Tg-DN-Mst1 than in NTg. The number of TUNEL-positive myocytes (0.17 versus 0.28%, P<0.05) and amount of interstitial fibrosis (5.0 versus 7.1%, P<0.05) in the remodeling area were significantly less in Tg-DN-Mst1 than in NTg. Upregulation of matrix metalloproteinase 2 and proinflammatory cytokines was significantly attenuated in Tg-DN-Mst1. These results indicate that endogenous Mst1 plays an important role in mediating cardiac dilation, apoptosis, fibrosis, and cardiac dysfunction, but not cardiac hypertrophy, after MI. Inhibition of Mst1 improves cardiac function without attenuating cardiac hypertrophy. Thus, Mst1 may be an important target of heart failure treatment.


Asunto(s)
Apoptosis , Cardiomegalia/etiología , Factor de Crecimiento de Hepatocito/fisiología , Infarto del Miocardio/complicaciones , Miocardio/patología , Proteínas Proto-Oncogénicas/fisiología , Disfunción Ventricular Izquierda/prevención & control , Animales , Citocinas/fisiología , Fibrosis , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/patología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Remodelación Ventricular
10.
Circ Res ; 101(11): 1164-74, 2007 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-17901358

RESUMEN

Glycogen synthase kinase (GSK)-3, a negative regulator of cardiac hypertrophy, is inactivated in failing hearts. To examine the histopathological and functional consequence of the persistent inhibition of GSK-3beta in the heart in vivo, we generated transgenic mice with cardiac-specific overexpression of dominant negative GSK-3beta (Tg-GSK-3beta-DN) and tetracycline-regulatable wild-type GSK-3beta. GSK-3beta-DN significantly reduced the kinase activity of endogenous GSK-3beta, inhibited phosphorylation of eukaryotic translation initiation factor 2B epsilon, and induced accumulation of beta-catenin and myeloid cell leukemia-1, confirming that GSK-3beta-DN acts as a dominant negative in vivo. Tg-GSK-3beta-DN exhibited concentric hypertrophy at baseline, accompanied by upregulation of the alpha-myosin heavy chain gene and increases in cardiac function, as evidenced by a significantly greater Emax after dobutamine infusion and percentage of contraction in isolated cardiac myocytes, indicating that inhibition of GSK-3beta induces well-compensated hypertrophy. Although transverse aortic constriction induced a similar increase in hypertrophy in both Tg-GSK-3beta-DN and nontransgenic mice, Tg-GSK-3beta-DN exhibited better left ventricular function and less fibrosis and apoptosis than nontransgenic mice. Induction of the GSK-3beta transgene in tetracycline-regulatable wild-type GSK-3beta mice induced left ventricular dysfunction and premature death, accompanied by increases in apoptosis and fibrosis. Overexpression of GSK-3beta-DN in cardiac myocytes inhibited tumor necrosis factor-alpha-induced apoptosis, and the antiapoptotic effect of GSK-3beta-DN was abrogated in the absence of myeloid cell leukemia-1. These results suggest that persistent inhibition of GSK-3beta induces compensatory hypertrophy, inhibits apoptosis and fibrosis, and increases cardiac contractility and that the antiapoptotic effect of GSK-3beta inhibition is mediated by myeloid cell leukemia-1. Thus, downregulation of GSK-3beta during heart failure could be compensatory.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Animales , Apoptosis , Cardiomegalia/etiología , Cardiotónicos , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Fibrosis/etiología , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Transgénicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Contracción Miocárdica , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología
11.
Circ Res ; 98(2): 280-8, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16373598

RESUMEN

Ischemic preconditioning confers powerful protection against myocardial infarction through pre-emptive activation of survival signaling pathways, but it remains difficult to apply to patients with ischemic heart disease, and its effects are transient. Promoting a sustained activation of preconditioning mechanisms in vivo would represent a novel approach of cardioprotection. We tested the role of the protein H11 kinase (H11K), which accumulates by 4- to 6-fold in myocardium of patients with chronic ischemic heart disease and in experimental models of ischemia. This increased expression was quantitatively reproduced in cardiac myocytes using a transgenic (TG) mouse model. After 45 minutes of coronary artery occlusion and reperfusion, hearts from TG mice showed an 82+/-5% reduction in infarct size compared with wild-type (WT), which was similar to the 84+/-4% reduction of infarct size observed in WT after a protocol of ischemic preconditioning. Hearts from TG mice showed significant activation of survival kinases participating in preconditioning, including Akt and the 5'AMP-activated protein kinase (AMPK). H11K directly binds to both Akt and AMPK and promotes their nuclear translocation and their association in a multiprotein complex, which results in a stimulation of survival mechanisms in cytosol and nucleus, including inhibition of proapoptotic effectors (glycogen synthase kinase-3beta, Bad, and Foxo), activation of antiapoptotic effectors (protein kinase Cepsilon, endothelial and inducible NO synthase isoforms, and heat shock protein 70), increased expression of the hypoxia-inducible factor-1alpha, and genomic switch to glucose utilization. Therefore, activation of survival pathways by H11K preemptively triggers the antiapoptotic and metabolic response to ischemia and is sufficient to confer cardioprotection in vivo equally potent to preconditioning.


Asunto(s)
Proteínas del Choque Térmico HSP20/fisiología , Precondicionamiento Isquémico Miocárdico , Proteínas Musculares/fisiología , Infarto del Miocardio/prevención & control , Proteínas Quinasas Activadas por AMP , Animales , Apoptosis , Supervivencia Celular , Células Cultivadas , Citoprotección , Proteínas de Choque Térmico , Ratones , Ratones Transgénicos , Chaperonas Moleculares , Complejos Multienzimáticos/metabolismo , Miocitos Cardíacos/citología , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
12.
Circulation ; 114(17): 1821-8, 2006 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-17043166

RESUMEN

BACKGROUND: The adaptation of cardiac mass to hemodynamic overload requires an adaptation of protein turnover, ie, the balance between protein synthesis and degradation. We tested 2 hypotheses: (1) chronic left ventricular hypertrophy (LVH) activates the proteasome system of protein degradation, especially in the myocardium submitted to the highest wall stress, ie, the subendocardium, and (2) the proteasome system is required for the development of LVH. METHODS AND RESULTS: Gene and protein expression of proteasome subunits and proteasome activity were measured separately from left ventricular subendocardium and subepicardium, right ventricle, and peripheral tissues in a canine model of severe, chronic (2 years) LVH induced by aortic banding and then were compared with controls. Both gene and protein expressions of proteasome subunits were increased in LVH versus control (P<0.05), which was accompanied by a significant (P<0.05) increase in proteasome activity. Posttranslational modification of the proteasome was also detected by 2-dimensional gel electrophoresis. These changes were found specifically in left ventricular subendocardium but not in left ventricular subepicardium, right ventricle, or noncardiac tissues from the same animals. In a mouse model of chronic pressure overload, a 50% increase in heart mass and a 2-fold increase in proteasome activity (both P<0.05 versus sham) were induced. In that model, the proteasome inhibitor epoxomicin completely prevented LVH while blocking proteasome activation. CONCLUSIONS: The increase in proteasome expression and activity found during chronic pressure overload in myocardium submitted to higher stress is also required for the establishment of LVH.


Asunto(s)
Hipertrofia Ventricular Izquierda/metabolismo , Proteínas Musculares/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Remodelación Ventricular/fisiología , Adaptación Fisiológica , Animales , Aorta Torácica , Modelos Animales de Enfermedad , Perros , Electroforesis en Gel Bidimensional , Femenino , Perfilación de la Expresión Génica , Ligadura , Masculino , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Oligopéptidos/farmacología , Poliubiquitina/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Subunidades de Proteína , Estrés Fisiológico/metabolismo
13.
J Clin Invest ; 112(9): 1395-406, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14597765

RESUMEN

Thioredoxin 1 (Trx1) has redox-sensitive cysteine residues and acts as an antioxidant in cells. However, the extent of Trx1 contribution to overall antioxidant mechanisms is unknown in any organs. We generated transgenic mice with cardiac-specific overexpression of a dominant negative (DN) mutant (C32S/C35S) of Trx1 (Tg-DN-Trx1 mice), in which the activity of endogenous Trx was diminished. Markers of oxidative stress were significantly increased in hearts from Tg-DN-Trx1 mice compared with those from nontransgenic (NTg) mice. Tg-DN-Trx1 mice exhibited cardiac hypertrophy with maintained cardiac function at baseline. Intraperitoneal injection of N-2-mercaptopropionyl glycine, an antioxidant, normalized cardiac hypertrophy in Tg-DN-Trx1 mice. Thoracic aortic banding caused greater increases in myocardial oxidative stress and enhanced hypertrophy in Tg-DN-Trx1 compared with NTg mice. In contrast, transgenic mice with cardiac-specific overexpression of wild-type Trx1 did not show cardiac hypertrophy at baseline but exhibited reduced levels of hypertrophy and oxidative stress in response to pressure overload. These results demonstrate that endogenous Trx1 is an essential component of the cellular antioxidant mechanisms and plays a critical role in regulating oxidative stress in the heart in vivo. Furthermore, inhibition of endogenous Trx1 in the heart primarily stimulates hypertrophy, both under basal conditions and in response to pressure overload through redox-sensitive mechanisms.


Asunto(s)
Cardiomegalia/etiología , Estrés Oxidativo , Tiorredoxinas/antagonistas & inhibidores , Animales , Cardiomegalia/metabolismo , Catalasa/metabolismo , Células Cultivadas , Ratones , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Superóxido Dismutasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
14.
J Clin Invest ; 110(2): 271-9, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12122119

RESUMEN

Mitogen-activated protein kinase kinase kinase (MEKK1) mediates activation of c-Jun NH(2)-terminal kinase (JNK). Although previous studies using cultured cardiac myocytes have suggested that the MEKK1-JNK pathway plays a key role in hypertrophy and apoptosis, its effects in cardiac hypertrophy and apoptosis are not fully understood in adult animals in vivo. We examined the role of the MEKK1-JNK pathway in pressure-overloaded hearts by using mice deficient in MEKK1. We found that transverse aortic banding significantly increased JNK activity in Mekk1(+/+) but not Mekk1(-/-) mice, indicating that MEKK1 mediates JNK activation by pressure overload. Nevertheless, pressure overload caused significant levels of cardiac hypertrophy and expression of atrial natriuretic factor in Mekk1(-/-) animals, which showed higher mortality and lung/body weight ratio than were seen in controls. Fourteen days after banding, Mekk1(-/-) hearts were dilated, and their left ventricular ejection fraction was low. Pressure overload caused elevated levels of apoptosis and inflammatory lesions in these mice and produced a smaller increase in TGF-beta and TNF-alpha expression than occurred in wild-type controls. Thus, MEKK1 appears to be required for pressure overload-induced JNK activation and cytokine upregulation but to be dispensable for pressure overload-induced cardiac hypertrophy. MEKK1 also prevents apoptosis and inflammation, thereby protecting against heart failure and sudden death following cardiac pressure overload.


Asunto(s)
Presión Sanguínea/fisiología , Hipertrofia Ventricular Izquierda/etiología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Apoptosis/fisiología , Factor Natriurético Atrial/genética , Activación Enzimática , Expresión Génica , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Factor de Crecimiento Transformador beta/genética , Factor de Necrosis Tumoral alfa/genética
15.
J Clin Invest ; 111(10): 1463-74, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12750396

RESUMEN

Activation of mammalian sterile 20-like kinase 1 (Mst1) by genotoxic compounds is known to stimulate apoptosis in some cell types. The importance of Mst1 in cell death caused by clinically relevant pathologic stimuli is unknown, however. In this study, we show that Mst1 is a prominent myelin basic protein kinase activated by proapoptotic stimuli in cardiac myocytes and that Mst1 causes cardiac myocyte apoptosis in vitro in a kinase activity-dependent manner. In vivo, cardiac-specific overexpression of Mst1 in transgenic mice results in activation of caspases, increased apoptosis, and dilated cardiomyopathy. Surprisingly, however, Mst1 prevents compensatory cardiac myocyte elongation or hypertrophy despite increased wall stress, thereby obscuring the use of the Frank-Starling mechanism, a fundamental mechanism by which the heart maintains cardiac output in response to increased mechanical load at the single myocyte level. Furthermore, Mst1 is activated by ischemia/reperfusion in the mouse heart in vivo. Suppression of endogenous Mst1 by cardiac-specific overexpression of dominant-negative Mst1 in transgenic mice prevents myocyte death by pathologic insults. These results show that Mst1 works as both an essential initiator of apoptosis and an inhibitor of hypertrophy in cardiac myocytes, resulting in a previously unrecognized form of cardiomyopathy.


Asunto(s)
Apoptosis , Cardiomegalia/etiología , Cardiomiopatía Dilatada/etiología , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Alcaloides , Animales , Apoptosis/efectos de los fármacos , Benzofenantridinas , Cardiomegalia/patología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genes Dominantes , Ventrículos Cardíacos/patología , Toxinas Marinas , Ratones , Ratones Transgénicos , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Especificidad de Órganos , Oxazoles/farmacología , Fenantridinas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Wistar , Transducción Genética
16.
Circ Res ; 97(3): 219-26, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-16037571

RESUMEN

Endocardial cushions are precursors of mature atrioventricular (AV) valves. Their formation is induced by signaling molecules originating from the AV myocardium, including bone morphogenetic proteins (BMPs). Here, we hypothesized that BMP signaling plays an important role in the AV myocardium during the maturation of AV valves from the cushions. To test our hypothesis, we used a unique Cre/lox system to target the deletion of a floxed Alk3 allele, the type IA receptor for BMPs, to cardiac myocytes of the AV canal (AVC). Lineage analysis indicated that cardiac myocytes of the AVC contributed to the tricuspid mural and posterior leaflets, the mitral septal leaflet, and the atrial border of the annulus fibrosus. When Alk3 was deleted in these cells, defects were seen in the same leaflets, ie, the tricuspid mural leaflet and mitral septal leaflet were longer, the tricuspid posterior leaflet was displaced and adherent to the ventricular wall, and the annulus fibrosus was disrupted resulting in ventricular preexcitation. The defects seen in mice with AVC-targeted deletion of Alk3 provide strong support for a role of Alk3 in human congenital heart diseases, such as Ebstein's anomaly. In conclusion, our mouse model demonstrated critical roles for Alk3 signaling in the AV myocardium during the development of AV valves and the annulus fibrosus.


Asunto(s)
Válvulas Cardíacas/embriología , Corazón/embriología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Proteínas de Unión al ADN/genética , Factor de Transcripción GATA6 , Válvulas Cardíacas/anomalías , Integrasas/genética , Ratones , Ratones Transgénicos , Miocitos Cardíacos/fisiología , Transducción de Señal , Factores de Transcripción/genética
17.
Circ Res ; 93(4): 364-71, 2003 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-12869393

RESUMEN

In a genetically engineered mouse line with disruption of type 5 adenylyl cyclase (AC5-/-), a major cardiac isoform, there was no compensatory increase in other isoforms of AC in the heart. Both basal and isoproterenol (ISO)-stimulated AC activities were decreased by 30% to 40% in cardiac membranes. The reduced AC activity did not affect cardiac function (left ventricular ejection fraction [LVEF]) at baseline. However, increases in LVEF after ISO were significantly attenuated in AC5-/- (P<0.05, n=11). Paradoxically, conscious AC5-/- mice had a higher heart rate compared with wild-type (WT) mice (613+/-8 versus 523+/-11 bpm, P<0.01, n=14 to 15). Muscarinic agonists decreased AC activity, LVEF, and heart rate more in WT than in AC5-/-. In addition, baroreflex-mediated, ie, neuronally regulated, bradycardia after phenylephrine was also attenuated in AC5-/-. The carbachol-activated outward potassium current (at -40 mV) normalized to cell capacitance in AC5-/- (2.6+/-0.4 pA/pF, n=16) was similar to WT (2.9+/-0.3 pA/pF, n=27), but calcium (Ca2+)-mediated inhibition of AC activity and Ca2+ channel function were diminished in AC5-/-. Thus, AC5-/- attenuates sympathetic responsiveness and also impairs parasympathetic and Ca2+-mediated regulation of the heart, indicating that those actions are not only regulated at the level of the receptor and G-protein but also at the level of type 5 AC.


Asunto(s)
Adenilil Ciclasas/genética , Calcio/farmacología , Corazón/efectos de los fármacos , Miocardio/enzimología , Sistema Nervioso Parasimpático/fisiología , Sistema Nervioso Simpático/fisiología , Acetilcolina/farmacología , Adenilil Ciclasas/metabolismo , Animales , Atropina/farmacología , Unión Competitiva , Western Blotting , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/fisiología , Cardiotónicos/farmacología , Electrocardiografía , Femenino , Genotipo , Corazón/inervación , Corazón/fisiología , Frecuencia Cardíaca/fisiología , Isoenzimas/genética , Isoenzimas/metabolismo , Isoproterenol/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Antagonistas Muscarínicos/farmacología , Miocardio/metabolismo , Canales de Potasio/fisiología , Receptores Adrenérgicos beta/metabolismo , Vasodilatadores/farmacología , Verapamilo/farmacología
18.
Circulation ; 105(5): 614-20, 2002 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-11827928

RESUMEN

BACKGROUND: To understand further the pathogenesis of familial hypertrophic cardiomyopathy, we determined how the cardiomyopathy induced by an Arg403-->Gln missense mutation in the alpha-myosin heavy chain (403) is affected by chronically enhancing sympathetic drive by mating the mice with those overexpressing G(s)alpha (G(s)alpha x403). METHODS AND RESULTS: Heart rate in 3-month-old conscious mice was elevated similarly (P<0.05) in mice overexpressing G(s)alpha (G(s)alpha mice; 746 +/- 14 bpm) and G(s)alpha x403 mice (718+/- 19 bpm) compared with littermate wild-type mice (WT; 623+/- 18 bpm) and 403 mice (594+/- 16 bpm). Left ventricular ejection fraction (LVEF), as determined by echocardiography, was enhanced in G(s)alpha x403 mice (88+/- 1%, P<0.001) compared with WT (69+/- 1%), 403 (75+/- 1%), and G(s)alpha (69 +/- 2%) mice. Isolated cardiomyocytes from G(s)alpha x403 mice also exhibited higher (P<0.001) baseline percent contraction (11.9+/- 0.5%) than WT (7.0+/- 0.5%), 403 (5.5+/- 0.5%), and G(s)alpha (7.8+/- 0.3%) cardiomyocytes. Relaxation of myocytes was impaired in 403 mice compared with WT but enhanced in G(s)alpha and normalized in G(s)alpha x403 mice. This was also observed in vivo. In vivo isoproterenol (0.1 microgram . kg(-1) . min(-1)) increased LVEF to maximal levels in G(s)alpha x403 and G(s)alpha, whereas in 403, the response was attenuated compared with WT. At 10 months of age, G(s)alpha x403 had significantly depressed LVEF (57 +/- 4%). Histopathological examination demonstrated that myocyte hypertrophy and fibrosis were already present in young G(s)alpha x403 mice and that old animals had severe cardiomyopathy. By 15 months of age, the survival of G(s)alpha x403 was 0% compared with 100% for WT, 71% for G(s)alpha, and 100% for 403 mice (P<0.05). CONCLUSIONS: These results show that the cardiomyopathy developed by G(s)alpha x403 mice is synergistic rather than additive, most likely owing to the elevated baseline function combined with enhanced responsiveness to sympathetic stimulation.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Mutación Missense , Cadenas Pesadas de Miosina/genética , Animales , Peso Corporal , Cardiomiopatía Hipertrófica Familiar/patología , Separación Celular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ecocardiografía , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Frecuencia Cardíaca/genética , Técnicas In Vitro , Ratones , Ratones Transgénicos , Contracción Miocárdica/genética , Miocardio/metabolismo , Miocardio/patología , Cadenas Pesadas de Miosina/metabolismo , Tamaño de los Órganos , Volumen Sistólico/genética , Tasa de Supervivencia , Función Ventricular Izquierda/genética
19.
Cardiovasc Res ; 59(3): 649-57, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14499866

RESUMEN

OBJECTIVE: The calcineurin pathway has been involved in the development of cardiac hypertrophy, yet it remains unknown whether calcineurin activity can be regulated in myocardium independently from hypertrophy and cardiac load. METHODS: To test that hypothesis, we measured calcineurin activity in a rat model of infrarenal aortic constriction (IR), which affects neurohormonal pathways without increasing cardiac afterload. RESULTS: In this model, there was no change in arterial pressure over the 4-week experimental period, and the left ventricle/body weight ratio did not increase. At 2 weeks after IR, calcineurin activity was increased 1.8-fold (P<0.05) and remained elevated at 4 weeks (1.7-fold, P<0.05). Similarly, the cardiac activity of calcium calmodulin kinase II (CaMKII) was increased significantly after IR, which confirms a regulation of Ca(2+)-dependent enzymes in this model. In cardiac myocytes, the increased activity of calcineurin was accompanied by a significant decrease in L-type Ca(2+) channel activity (I(Ca)) and contraction velocity (-dL/dt). Cardiac denervation prevented the activation of calcineurin after IR, which demonstrates that a neurohormonal mechanism is responsible for the changes in enzymatic activity. In addition, cardiac denervation suppressed the effects of IR on I(Ca) and -dL/dt, which shows that calcineurin activation is related to altered contractility. However, action potential duration, the densities of inward rectifier K(+) currents (I(K1)), and outward K(+) currents (I(to) and I(K)) were not altered in IR myocytes. CONCLUSIONS: Calcineurin can be activated in the heart through a neural stimulus, which induces alterations in Ca(2+) currents and contractility. These effects occur in the absence of myocyte hypertrophy, electrophysiological changes in action potential, and K(+) channel currents.


Asunto(s)
Estenosis Aórtica Subvalvular/metabolismo , Sistema Nervioso Autónomo/fisiopatología , Calcineurina/metabolismo , Contracción Miocárdica , Miocardio/metabolismo , Animales , Aorta Abdominal , Estenosis Aórtica Subvalvular/fisiopatología , Factor Natriurético Atrial/genética , Desnervación Autonómica , Calcineurina/análisis , Señalización del Calcio , Electrofisiología , Immunoblotting , Masculino , Modelos Animales , Canales de Potasio/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley
20.
PLoS One ; 8(1): e51632, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23308102

RESUMEN

Autophagy is a bulk degradation mechanism for cytosolic proteins and organelles. The heart undergoes hypertrophy in response to mechanical load but hypertrophy can regress upon unloading. We hypothesize that autophagy plays an important role in mediating regression of cardiac hypertrophy during unloading. Mice were subjected to transverse aortic constriction (TAC) for 1 week, after which the constriction was removed (DeTAC). Regression of cardiac hypertrophy was observed after DeTAC, as indicated by reduction of LVW/BW and cardiomyocyte cross-sectional area. Indicators of autophagy, including LC3-II expression, p62 degradation and GFP-LC3 dots/cell, were significantly increased after DeTAC, suggesting that autophagy is induced. Stimulation of autophagy during DeTAC was accompanied by upregulation of FoxO1. Upregulation of FoxO1 and autophagy was also observed in vitro when cultured cardiomyocytes were subjected to mechanical stretch followed by incubation without stretch (de-stretch). Transgenic mice with cardiac-specific overexpression of FoxO1 exhibited smaller hearts and upregulation of autophagy. Overexpression of FoxO1 in cultured cardiomyocytes significantly reduced cell size, an effect which was attenuated when autophagy was inhibited. To further examine the role of autophagy and FoxO1 in mediating the regression of cardiac hypertrophy, beclin1+/- mice and cultured cardiomyocytes transduced with adenoviruses harboring shRNA-beclin1 or shRNA-FoxO1 were subjected to TAC/stretch followed by DeTAC/de-stretch. Regression of cardiac hypertrophy achieved after DeTAC/de-stretch was significantly attenuated when autophagy was suppressed through downregulation of beclin1 or FoxO1. These results suggest that autophagy and FoxO1 play an essential role in mediating regression of cardiac hypertrophy during mechanical unloading.


Asunto(s)
Autofagia , Cardiomegalia/fisiopatología , Corazón/fisiopatología , Animales , Autofagia/efectos de los fármacos , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Tamaño de la Célula , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Regulación hacia Arriba , Soporte de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA