Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
2.
Stem Cell Reports ; 8(2): 226-234, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28111276

RESUMEN

To predict drug-induced serious adverse events (SAE) in clinical trials, a model using a panel of cells derived from human induced pluripotent stem cells (hiPSCs) of individuals with different susceptibilities could facilitate major advancements in translational research in terms of safety and pharmaco-economics. However, it is unclear whether hiPSC-derived cells can recapitulate interindividual differences in drug-induced SAE susceptibility in populations not having genetic disorders such as healthy subjects. Here, we evaluated individual differences in SAE susceptibility based on an in vitro model using hiPSC-derived cardiomyocytes (hiPSC-CMs) as a pilot study. hiPSCs were generated from blood samples of ten healthy volunteers with different susceptibilities to moxifloxacin (Mox)-induced QT prolongation. Different Mox-induced field potential duration (FPD) prolongation values were observed in the hiPSC-CMs from each individual. Interestingly, the QT interval was significantly positively correlated with FPD at clinically relevant concentrations (r > 0.66) in multiple analyses including concentration-QT analysis. Genomic analysis showed no interindividual significant differences in known target-binding sites for Mox and other drugs such as the hERG channel subunit, and baseline QT ranges were normal. The results suggest that hiPSC-CMs from healthy subjects recapitulate susceptibility to Mox-induced QT prolongation and provide proof of concept for in vitro preclinical trials.


Asunto(s)
Fenómenos Electrofisiológicos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Potenciales de Acción/efectos de los fármacos , Alelos , Diferenciación Celular , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Electrocardiografía , Perfilación de la Expresión Génica , Frecuencia de los Genes , Voluntarios Sanos , Sistema de Conducción Cardíaco/efectos de los fármacos , Humanos , Masculino , Mutación , Miocitos Cardíacos/citología , Polimorfismo de Nucleótido Simple
3.
J Biol Chem ; 287(13): 9827-9834, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22228770

RESUMEN

Insulin-like growth factor 1 (IGF1) promotes a physiological type of cardiac hypertrophy and has therapeutic effects in heart disease. Here, we report the relationship of IGF1 to GATA4, an essential transcription factor in cardiac hypertrophy and cell survival. In cultured neonatal rat ventricular myocytes, we compared the responses to IGF1 (10 nmol/liter) and phenylephrine (PE, 20 µmol/liter), a known GATA4 activator, in concentrations promoting a similar extent of hypertrophy. IGF1 and PE both increased nuclear accumulation of GATA4 and phosphorylation at Ser(105) (PE, 2.4-fold; IGF1, 1.8-fold; both, p < 0.05) and increased GATA4 DNA binding activity as indicated by ELISA and by chromatin IP of selected promoters. Although IGF1 and PE each activated GATA4 to the same degree, GATA4 knockdown by RNA interference only blocked hypertrophy by PE but not by IGF1. PE induction of a panel of GATA4 target genes (Nppa, Nppb, Tnni3, Myl1, and Acta1) was inhibited by GATA4 knockdown. In contrast, IGF1 regulated only Acta1 in a GATA4-dependent fashion. Consistent with the in vitro findings, Gata4 haploinsufficiency in mice did not alter cardiac structure, hyperdynamic function, or antifibrotic effects induced by myocardial overexpression of the IGF1 receptor. Our data indicate that GATA4 is activated by the IGF1 pathway, but although it is required for responses to pathological stimuli, it is not necessary for the effects of IGF1 on cardiac structure and function.


Asunto(s)
Cardiomegalia/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor I del Crecimiento Similar a la Insulina/efectos adversos , Miocitos Cardíacos/metabolismo , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/patología , Células Cultivadas , Factor de Transcripción GATA4/genética , Técnicas de Silenciamiento del Gen , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología , Regiones Promotoras Genéticas/genética , Ratas , Ratas Wistar
4.
Circ Res ; 107(10): 1275-89, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20847309

RESUMEN

RATIONALE: Mechanotransduction and the response to biomechanical stress is a fundamental response in heart disease. Loss of phosphoinositide 3-kinase (PI3K)γ, the isoform linked to G protein-coupled receptor signaling, results in increased myocardial contractility, but the response to pressure overload is controversial. OBJECTIVE: To characterize molecular and cellular responses of the PI3Kγ knockout (KO) mice to biomechanical stress. METHODS AND RESULTS: In response to pressure overload, PI3KγKO mice deteriorated at an accelerated rate compared with wild-type mice despite increased basal myocardial contractility. These functional responses were associated with compromised phosphorylation of Akt and GSK-3α. In contrast, isolated single cardiomyocytes from banded PI3KγKO mice maintained their hypercontractility, suggesting compromised interaction with the extracellular matrix as the primary defect in the banded PI3KγKO mice. ß-Adrenergic stimulation increased cAMP levels with increased phosphorylation of CREB, leading to increased expression of cAMP-responsive matrix metalloproteinases (MMPs), MMP2, MT1-MMP, and MMP13 in cardiomyocytes and cardiofibroblasts. Loss of PI3Kγ resulted in increased cAMP levels with increased expression of MMP2, MT1-MMP, and MMP13 and increased MMP2 activation and collagenase activity in response to biomechanical stress. Selective loss of N-cadherin from the adhesion complexes in the PI3KγKO mice resulted in reduced cell adhesion. The ß-blocker propranolol prevented the upregulation of MMPs, whereas MMP inhibition prevented the adverse remodeling with both therapies, preventing the functional deterioration in banded PI3KγKO mice. In banded wild-type mice, long-term propranolol prevented the adverse remodeling and systolic dysfunction with preservation of the N-cadherin levels. CONCLUSIONS: The enhanced propensity to develop heart failure in the PI3KγKO mice is attributable to a cAMP-dependent upregulation of MMP expression and activity and disorganization of the N-cadherin/ß-catenin cell adhesion complex. ß-Blocker therapy prevents these changes thereby providing a novel mechanism of action for these drugs.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Cardiomegalia/enzimología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , AMP Cíclico/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Mecanotransducción Celular , Miocardio/enzimología , Remodelación Ventricular , Antagonistas Adrenérgicos beta/administración & dosificación , Animales , Fenómenos Biomecánicos , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/fisiopatología , Adhesión Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica , Miocardio/patología , Miocitos Cardíacos/enzimología , Fosforilación , Propranolol/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estrés Mecánico , Factores de Tiempo , Remodelación Ventricular/efectos de los fármacos , beta Catenina/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 30(4): 724-32, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20237330

RESUMEN

OBJECTIVE: Myocardial infarction (MI) is a serious complication of atherosclerosis associated with increasing mortality attributable to heart failure. Activation of phosphoinositide 3-kinase [PI3K(p110 alpha)] is considered a new strategy for the treatment of heart failure. However, whether PI3K(p110 alpha) provides protection in a setting of MI is unknown, and PI3K(p110 alpha) is difficult to target because it has multiple actions in numerous cell types. The goal of this study was to assess whether PI3K(p110 alpha) is beneficial in a setting of MI and, if so, to identify cardiac-selective microRNA and mRNA that mediate the protective properties of PI3K(p110 alpha). METHODS AND RESULTS: Cardiomyocyte-specific transgenic mice with increased or decreased PI3K(p110 alpha) activity (caPI3K-Tg and dnPI3K-Tg, respectively) were subjected to MI for 8 weeks. The caPI3K-Tg subjected to MI had better cardiac function than nontransgenic mice, whereas dnPI3K-Tg had worse function. Using microarray analysis, we identified PI3K-regulated miRNA and mRNA that were correlated with cardiac function, including growth factor receptor-bound 14. Growth factor receptor-bound 14 is highly expressed in the heart and positively correlated with PI3K(p110 alpha) activity and cardiac function. Mice deficient in growth factor receptor-bound 14 have cardiac dysfunction. CONCLUSIONS: Activation of PI3K(p110 alpha) protects the heart against MI-induced heart failure. Cardiac-selective targets that mediate the protective effects of PI3K(p110 alpha) represent new drug targets for heart failure.


Asunto(s)
Insuficiencia Cardíaca/prevención & control , MicroARNs/metabolismo , Infarto del Miocardio/enzimología , Miocardio/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas/genética , ARN Mensajero/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Miocardio/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/genética , Proteínas/metabolismo , Factores de Tiempo , Ultrasonografía , Función Ventricular Izquierda , Presión Ventricular
6.
Am J Pathol ; 175(3): 998-1009, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19679877

RESUMEN

Atrial fibrillation (AF) is the most common sustained arrhythmia presenting at cardiology departments. A limited understanding of the molecular mechanisms responsible for the development of AF has hindered treatment strategies. The purpose of this study was to assess whether reduced activation of phosphoinositide 3-kinase (PI3K, p110alpha) makes the compromised heart susceptible to AF. Risk factors for AF, including aging, obesity, and diabetes, have been associated with insulin resistance that leads to depressed/defective PI3K signaling. However, to date, there has been no link between PI3K(p110alpha) and AF. To address this question, we crossed a cardiac-specific transgenic mouse model of dilated cardiomyopathy (DCM) with a cardiac-specific transgenic mouse expressing a dominant negative mutant of PI3K (dnPI3K; reduces PI3K activity). Adult ( approximately 4.5 months) double-transgenic (dnPI3K-DCM), single-transgenic (DCM-Tg, dnPI3K-Tg), and nontransgenic mice were subjected to morphological, functional/ECG, microarray, and biochemical analyses. dnPI3K-DCM mice developed AF and had depressed cardiac function as well as greater atrial enlargement and fibrosis than DCM-Tg mice. AF was not detected in other groups. Aged DCM-Tg mice ( approximately 15 months) with a similar phenotype to dnPI3K-DCM mice (4.5 months) did not develop AF, suggesting loss of PI3K activity directly contributed to the AF phenotype. Furthermore, increasing PI3K activity reduced atrial fibrosis and improved cardiac conduction in DCM-Tg mice. Finally, in atrial appendages from patients with AF, PI3K activation was lower compared with tissue from patients in sinus rhythm. These results suggest a link between PI3K(p110alpha) and AF.


Asunto(s)
Fibrilación Atrial/enzimología , Cardiomiopatía Dilatada/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Fibrilación Atrial/etiología , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/fisiopatología , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Activación Enzimática , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
Stem Cells ; 27(8): 1869-78, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544447

RESUMEN

In vitro differentiation of human embryonic stem cells (hESCs) into pure human cardiomyocytes (hESCMs) would present a powerful tool to further the creation of cell models designed to advance preclinical drug development. Here, we report a novel differentiation method to substantially increase hESCM yield. Upon early and transient treatment of hESCs with Wnt3a, embryoid body and mesendoderm formation is enhanced, leading to greater differentiation toward cardiomyocytes. Moreover, the generated beating clusters are highly enriched with cardiomyocytes (50%) and express genes characteristic of cardiac cells, providing evidence that these hESCMs are competent to develop in vitro into functional and physiologically relevant cardiomyocytes. In summary, this protocol not only has the potential to guarantee a renewable supply of enriched cardiomyocyte populations for developing novel and more predictive cell models, but it also should provide valuable insights into pathways critical for cardiac regeneration.


Asunto(s)
Células Madre Embrionarias/citología , Miocitos Cardíacos/citología , Ingeniería de Tejidos/métodos , Proteínas Wnt/farmacología , Animales , Comunicación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Humanos , Insulina/farmacología , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteínas Recombinantes/farmacología , Proteína Wnt3 , Proteína Wnt3A
8.
Am J Physiol Heart Circ Physiol ; 296(3): H566-72, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19122165

RESUMEN

The phosphoinositide 3-kinase (PI3-kinase)-protein kinase B (Akt) signaling pathway is essential in the induction of physiological cardiac hypertrophy. In contrast, protein kinase C beta2 (PKCbeta2) is implicated in the development of pathological cardiac hypertrophy and heart failure. Thus far, no clear association has been demonstrated between these two pathways. In this study, we examined the potential interaction between the PI3-kinase and PKCbeta2 pathways by crossing transgenic mice with cardiac specific expression of PKCbeta2, constitutively active (ca) PI3-kinase, and dominant-negative (dn) PI3-kinase. In caPI3-kinase/PKCbeta2 and dnPI3-kinase/PKCbeta2 double-transgenic mice, the heart weight-to-body weight ratios and cardiomyocyte sizes were similar to those observed in caPI3-kinase and dnPI3-kinase transgenic mice, respectively, suggesting that the regulation of physiological developmental hypertrophy via modulation of cardiomyocyte size proceeds through the PI3-kinase pathway. In addition, we observed that caPI3-kinase/PKCbeta2 mice showed improved cardiac function while the function of dnPI3-kinase/PKCbeta2 mice was similar to that of the PKCbeta2 group. PKCbeta2 protein levels in both dnPI3-kinase/PKCbeta2 and PKCbeta2 mice were significantly upregulated. Interestingly, however, PKCbeta2 protein expression was significantly attenuated in caPI3-kinase/PKCbeta2 mice. PI3-kinase activity measured by Akt phosphorylation was not affected by PKCbeta2 overexpression. These data suggest a potential interaction between these two pathways in the heart, where PI3-kinase is predominantly responsible for the regulation of physiological developmental hypertrophy and may act as an upstream modulator of PKCbeta2 with the potential for rescuing the pathological cardiac dysfunction induced by overexpression of PKCbeta.


Asunto(s)
Cardiomegalia/enzimología , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Cardiomegalia/fisiopatología , Bovinos , Tamaño de la Célula , Células Cultivadas , Femenino , Frecuencia Cardíaca , Masculino , Ratones , Ratones Transgénicos , Mutación , Contracción Miocárdica , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteína Quinasa C/genética , Proteína Quinasa C beta , Subunidades de Proteína , Ratas , Ratas Sprague-Dawley , Función Ventricular Izquierda
9.
J Am Coll Cardiol ; 54(25): 2435-46, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20082935

RESUMEN

OBJECTIVES: The extent of adverse myocardial remodeling contributes essentially to the prognosis after myocardial infarction (MI). In this study we investigated whether inhibition of "mammalian target of rapamycin" (mTOR) attenuates left ventricular (LV) remodeling after MI. BACKGROUND: Therapeutic strategies to inhibit remodeling are currently limited to inhibition of neurohumoral activation. The mTOR-dependent signaling mechanisms are centrally involved in remodeling processes and provide new therapeutic opportunities. METHODS: Everolimus (RAD) treatment was initiated on the day after or 3 days after induction of myocardial infarction (MI) in rats. RESULTS: After 28 days, RAD-treated animals had reduced post-MI remodeling, with improved LV function and smaller LV end-diastolic diameters (8.9 + or - 0.3 mm vs. 11.4 + or - 0.2 mm, p < 0.05), end-diastolic volumes (304 + or - 30 microl vs. 414 + or - 16 microl, p < 0.05), and cardiac myocyte size (-40% vs. vehicle, p < 0.05). Infarct size was significantly reduced compared with vehicle-treated animals. The mTOR inhibition increased autophagy and concomitantly decreased proteasome activity in the border zone of the infarcted myocardium. Measurement of autophagic flux demonstrated that RAD did not decrease autophagosome clearance. When RAD treatment was initiated 3 days after MI, adverse remodeling was still attenuated and increased autophagy was still present. Sustained improvement of LV function was observed 3 months after MI, even when RAD treatment was discontinued after 1 month. CONCLUSIONS: Inhibition of mTOR is a potential therapeutic strategy to limit infarct size and to attenuate adverse LV remodeling after MI.


Asunto(s)
Inmunosupresores/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Infarto del Miocardio/fisiopatología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Sirolimus/análogos & derivados , Remodelación Ventricular/efectos de los fármacos , Animales , Factor Natriurético Atrial/metabolismo , Autofagia/efectos de los fármacos , Diástole/fisiología , Ecocardiografía , Everolimus , Ventrículos Cardíacos/diagnóstico por imagen , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Proteínas Asociadas a Microtúbulos/fisiología , Miocitos Cardíacos/patología , FN-kappa B/efectos de los fármacos , Fosforilación , Proteínas Serina-Treonina Quinasas/fisiología , Ratas , Ratas Wistar , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Regulación hacia Arriba , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/fisiología
10.
Circ Res ; 103(6): 643-53, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18688045

RESUMEN

Ischemic preconditioning (IPC) is a potent cellular protective mechanism whereby brief periods of sublethal ischemia protect the myocardium from prolonged ischemia-induced injury. We demonstrate the selective role of phosphatidylinositol 3-kinase (PI3K) isoforms in IPC. Hearts from PI3Kgamma knockout mice (PI3Kgamma(-/-)) displayed poorer functional recovery and greater tissue injury following IPC compared to wild-type and PI3Kgamma(+/-) hearts. Examination of the cell-signaling pathways revealed restored phosphorylation levels of Akt and glycogen synthase kinase (GSK)3beta in wild-type hearts, which were abolished in PI3Kgamma(-/-) hearts subjected to IPC. Inhibition of GSK3beta by LiCl reversed the loss in protection in PI3Kgamma(-/-) hearts. In contrast, mice expressing a cardiac-specific kinase-deleted PI3Kalpha (PI3KalphaDN) were resistant to injury induced by 30 minutes of ischemia followed by 40 minutes of reperfusion. Furthermore, the resistance of PI3KalphaDN hearts to ischemia/reperfusion correlated with the persistent expression of p110gamma and was blocked by the PI3K inhibitor wortmannin, suggesting the possible enhanced cell signaling through the PI3Kgamma pathway. These results demonstrate the importance of the PI3Kgamma-Akt-GSK3beta signaling pathway in IPC. Selective activation of myocardial PI3Kgamma may be an attractive target for the treatment of ischemic heart disease.


Asunto(s)
Adenosina/farmacología , Precondicionamiento Isquémico Miocárdico/métodos , Isquemia Miocárdica/enzimología , Isquemia Miocárdica/prevención & control , Fosfatidilinositol 3-Quinasas/fisiología , Adenosina/uso terapéutico , Animales , Fosfatidilinositol 3-Quinasa Clase Ib , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Isquemia Miocárdica/genética , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/prevención & control , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
Cardiovasc Res ; 78(3): 546-53, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18267957

RESUMEN

AIMS: The ability of lipopolysaccharide (LPS) pre-treatment to induce cardioprotection following ischaemia/reperfusion (I/R) has been well documented; however, the mechanisms have not been fully elucidated. LPS is a Toll-like receptor 4 (TLR4) ligand. Recent evidence indicates that there is cross-talk between the TLR and phosphoinositide 3-kinase/Akt (PI3K/Akt) signalling pathways. We hypothesized that activation of PI3K/Akt signalling plays a critical role in LPS-induced cardioprotection. METHODS AND RESULTS: To evaluate this hypothesis, we pre-treated mice with LPS 24 h before the hearts were subjected to ischaemia (45 min) and reperfusion (4 h). We examined activation of the PI3K/Akt/GSK-3beta signalling pathway. The effect of PI3K/Akt inhibition on LPS-induced cardioprotection was also evaluated. LPS pre-treatment significantly reduced infarct size (71.25%) compared with the untreated group (9.3+/-1.58 vs. 32.3+/-2.92%, P<0.01). Cardiac myocyte apoptosis and caspase-3 activity in LPS-pre-treated mice were significantly reduced following I/R. LPS pre-treatment significantly increased the levels of phospho-Akt, phospho-GSK-3beta, and heat shock protein 27 in the myocardium. Pharmacological inhibition of PI3K by LY294002 or genetic modulation employing kinase-defective Akt transgenic mice abolished the cardioprotection induced by LPS. CONCLUSION: These results indicate that LPS-induced cardioprotection in I/R injury is mediated through a PI3K/Akt-dependent mechanism.


Asunto(s)
Lipopolisacáridos/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Cromonas/farmacología , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Masculino , Ratones , Ratones Transgénicos , Morfolinas/farmacología , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética
12.
Cell Metab ; 6(4): 294-306, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17908558

RESUMEN

Physiological cardiac hypertrophy is associated with mitochondrial adaptations that are characterized by activation of PGC-1alpha and increased fatty acid oxidative (FAO) capacity. It is widely accepted that phosphatidylinositol 3-kinase (PI3K) signaling to Akt1 is required for physiological cardiac growth. However, the signaling pathways that coordinate physiological hypertrophy and metabolic remodeling are incompletely understood. We show here that activation of PI3K is sufficient to increase myocardial FAO capacity and that inhibition of PI3K signaling prevents mitochondrial adaptations in response to physiological hypertrophic stimuli despite increased expression of PGC-1alpha. We also show that activation of the downstream kinase Akt is not required for the mitochondrial adaptations that are secondary to PI3K activation. Thus, in physiological cardiac growth, PI3K is an integrator of cellular growth and metabolic remodeling. Although PI3K signaling to Akt1 is required for cellular growth, Akt-independent pathways mediate the accompanying mitochondrial adaptations.


Asunto(s)
Cardiomegalia/enzimología , Mitocondrias/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Condicionamiento Físico Animal , Proteínas Proto-Oncogénicas c-akt/fisiología , Adaptación Fisiológica , Animales , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Isoenzimas/antagonistas & inhibidores , Ratones , Ratones Mutantes , Miocitos Cardíacos/enzimología , Oxidación-Reducción , Fosfatidilinositol 3-Quinasas/genética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal
13.
J Mol Cell Cardiol ; 43(5): 601-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17904155

RESUMEN

Resistin, an adipocyte-derived hormone, is thought to represent a link between obesity and insulin-resistant diabetes. The potential role of resistin as a cardioprotective agent has not been explored. Our hypothesis is that resistin has a cardioprotective effect that is mediated by the resistin receptor-coupled activation of PI3K/Akt/PKC/K(ATP) dependent pathways. Our studies demonstrated that pretreatment of mouse hearts with 10 nM resistin for 5 min protected the heart against I/R injury in a mouse heart perfusion model. When mouse hearts were subjected to 60 min of LAD ligation followed by 4 h of reperfusion, resistin pretreatment (33 microg/kg) for 30 min or 24 h before ligation was able to significantly reduce the infarct size/risk area. The protective effect of resistin was abolished by wortmannin, as well as by an Akt inhibitor, triciribine. Resistin's protective effect was absent in Akt kinase-deficient mutant mice. The protective effect was also blocked by chelerythrine, a PKC inhibitor, and epsilonV1-2, a PKCepsilon inhibitor. Finally, the protective effect was blocked by 5-hydroxydecanoate, which blocks the opening of mitoK(ATP) channels. Resistin-induced Akt phosphorylation in HL-1 cells was inhibited by wortmannin and triciribine. Resistin also induced PKCepsilon phosphorylation, which was blocked by triciribine. These studies demonstrate that resistin's cardioprotective effect is mediated by PI3K/Akt/PKC dependent pathways. In addition to cardiomyocytes, resistin also induced Akt phosphorylation in endothelial cells and smooth muscle cells, suggesting that resistin receptors are present in these cells. The effect of resistin on apoptosis was assessed in hearts subjected to 30 min of ischemia and 3 h of reperfusion. There were significantly fewer in situ oligo ligation-positive myocyte nuclei in mice treated with resistin. Our results show that resistin can dramatically reduce apoptosis and infarct size, thus protecting the heart against I/R injury.


Asunto(s)
Infarto del Miocardio/prevención & control , Resistina/uso terapéutico , Adipoquinas/uso terapéutico , Androstadienos/uso terapéutico , Animales , Aorta/fisiología , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Endotelio Vascular/fisiología , Ventrículos Cardíacos/fisiopatología , Masculino , Ratones , Ratones Endogámicos ICR , Músculo Liso Vascular/fisiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/prevención & control , Resistina/fisiología , Ribonucleósidos/uso terapéutico , Wortmanina
14.
Nat Med ; 13(8): 952-61, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17660828

RESUMEN

Cardiac fibrosis, associated with a decreased extent of microvasculature and with disruption of normal myocardial structures, results from excessive deposition of extracellular matrix, which is mediated by the recruitment of fibroblasts. The source of these fibroblasts is unclear and specific anti-fibrotic therapies are not currently available. Here we show that cardiac fibrosis is associated with the emergence of fibroblasts originating from endothelial cells, suggesting an endothelial-mesenchymal transition (EndMT) similar to events that occur during formation of the atrioventricular cushion in the embryonic heart. Transforming growth factor-beta1 (TGF-beta1) induced endothelial cells to undergo EndMT, whereas bone morphogenic protein 7 (BMP-7) preserved the endothelial phenotype. The systemic administration of recombinant human BMP-7 (rhBMP-7) significantly inhibited EndMT and the progression of cardiac fibrosis in mouse models of pressure overload and chronic allograft rejection. Our findings show that EndMT contributes to the progression of cardiac fibrosis and that rhBMP-7 can be used to inhibit EndMT and to intervene in the progression of chronic heart disease associated with fibrosis.


Asunto(s)
Diferenciación Celular , Fibrosis Endomiocárdica/patología , Células Endoteliales/patología , Mesodermo/patología , Animales , Células de la Médula Ósea/patología , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/farmacología , Proteínas Morfogenéticas Óseas/uso terapéutico , Línea Celular , Células Cultivadas , Enfermedad Crónica , Fibrosis Endomiocárdica/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/patología , Mesodermo/efectos de los fármacos , Ratones , Ratones Transgénicos , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/uso terapéutico , Factor de Crecimiento Transformador beta1/farmacología
15.
Cell ; 129(7): 1365-76, 2007 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-17604724

RESUMEN

The cardiac conduction system is an anatomically discrete segment of specialized myocardium that initiates and propagates electrical impulses to coordinate myocardial contraction. To define the molecular composition of the mouse ventricular conduction system we used microdissection and transcriptional profiling by serial analysis of gene expression (SAGE). Conduction-system-specific expression for Id2, a member of the Id gene family of transcriptional repressors, was identified. Analyses of Id2-deficient mice demonstrated structural and functional conduction system abnormalities, including left bundle branch block. A 1.2 kb fragment of the Id2 promoter proved sufficient for cooperative regulation by Nkx2-5 and Tbx5 in vitro and for conduction-system-specific gene expression in vivo. Furthermore, compound haploinsufficiency of Tbx5 and Nkx2-5 or Tbx5 and Id2 prevented embryonic specification of the ventricular conduction system. We conclude that a molecular pathway including Tbx5, Nkx2-5, and Id2 coordinates specification of ventricular myocytes into the ventricular conduction system lineage.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Sistema de Conducción Cardíaco/anomalías , Cardiopatías Congénitas/genética , Ventrículos Cardíacos/anomalías , Proteínas de Homeodominio/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular/fisiología , Línea Celular , Linaje de la Célula/fisiología , Chlorocebus aethiops , Perfilación de la Expresión Génica , Sistema de Conducción Cardíaco/metabolismo , Cardiopatías Congénitas/metabolismo , Ventrículos Cardíacos/metabolismo , Proteína Homeótica Nkx-2.5 , Proteína 2 Inhibidora de la Diferenciación/deficiencia , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas/genética , Elementos Reguladores de la Transcripción/genética , Transducción de Señal/genética , Proteínas de Dominio T Box/deficiencia , Factores de Transcripción/deficiencia
16.
Dev Biol ; 303(2): 740-53, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17250822

RESUMEN

The ventricular conduction system is responsible for rapid propagation of electrical activity to coordinate ventricular contraction. To investigate the role of the transcription factor Nkx2.5 in the morphogenesis of the ventricular conduction system, we crossed Nkx2.5(+/-) mice with Cx40(eGFP/+) mice in which eGFP expression permits visualization of the His-Purkinje conduction system. Major anatomical and functional disturbances were detected in the His-Purkinje system of adult Nkx2.5(+/-)/Cx40(eGFP/+) mice, including hypoplasia of eGFP-positive Purkinje fibers and the disorganization of the Purkinje fiber network in the ventricular apex. Although the action potential properties of the individual eGFP-positive cells were normal, the deficiency of Purkinje fibers in Nkx2.5 haploinsufficient mice was associated with abnormalities of ventricular electrical activation, including slowed and decremented conduction along the left bundle branch. During embryonic development, eGFP expression in the ventricular trabeculae of Nkx2.5(+/-) hearts was qualitatively normal, with a measurable deficiency in eGFP-positive cells being observed only after birth. Chimeric analyses showed that maximal Nkx2.5 levels are required cell-autonomously. Reduced Nkx2.5 levels are associated with a delay in cell cycle withdrawal in surrounding GFP-negative myocytes. Our results suggest that the formation of the peripheral conduction system is time- and dose-dependent on the transcription factor Nkx2.5 that is cell-autonomously required for the postnatal differentiation of Purkinje fibers.


Asunto(s)
Sistema de Conducción Cardíaco/crecimiento & desarrollo , Sistema de Conducción Cardíaco/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Fascículo Atrioventricular/anomalías , Fascículo Atrioventricular/crecimiento & desarrollo , Fascículo Atrioventricular/fisiología , Diferenciación Celular , Electrofisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Sistema de Conducción Cardíaco/anomalías , Proteína Homeótica Nkx-2.5 , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Cardiovasculares , Ramos Subendocárdicos/anomalías , Ramos Subendocárdicos/crecimiento & desarrollo , Ramos Subendocárdicos/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Transcripción/deficiencia , Función Ventricular
17.
Proc Natl Acad Sci U S A ; 104(2): 612-7, 2007 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-17202264

RESUMEN

Physical activity protects against cardiovascular disease, and physiological cardiac hypertrophy associated with regular exercise is usually beneficial, in marked contrast to pathological hypertrophy associated with disease. The p110alpha isoform of phosphoinositide 3-kinase (PI3K) plays a critical role in the induction of exercise-induced hypertrophy. Whether it or other genes activated in the athlete's heart might have an impact on cardiac function and survival in a setting of heart failure is unknown. To examine whether progressive exercise training and PI3K(p110alpha) activity affect survival and/or cardiac function in two models of heart disease, we subjected a transgenic mouse model of dilated cardiomyopathy (DCM) to swim training, genetically crossed cardiac-specific transgenic mice with increased or decreased PI3K(p110alpha) activity to the DCM model, and subjected PI3K(p110alpha) transgenics to acute pressure overload (ascending aortic constriction). Life-span, cardiac function, and molecular markers of pathological hypertrophy were examined. Exercise training and increased cardiac PI3K(p110alpha) activity prolonged survival in the DCM model by 15-20%. In contrast, reduced PI3K(p110alpha) activity drastically shortened lifespan by approximately 50%. Increased PI3K(p110alpha) activity had a favorable effect on cardiac function and fibrosis in the pressure-overload model and attenuated pathological growth. PI3K(p110alpha) signaling negatively regulated G protein-coupled receptor stimulated extracellular responsive kinase and Akt (via PI3K, p110gamma) activation in isolated cardiomyocytes. These findings suggest that exercise and enhanced PI3K(p110alpha) activity delay or prevent progression of heart disease, and that supraphysiologic activity can be beneficial. Identification of genes important for hypertrophy in the athlete's heart could offer new strategies for treating heart failure.


Asunto(s)
Cardiomiopatía Dilatada/prevención & control , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica/prevención & control , Cardiomiopatía Hipertrófica/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Esfuerzo Físico/fisiología , Animales , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica/patología , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Condicionamiento Físico Animal , Transducción de Señal
18.
Novartis Found Symp ; 274: 90-111; discussion 111-7, 152-5, 272-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17019808

RESUMEN

Growth of the heart can be induced by physiological stimuli (e.g. postnatal development or chronic exercise training: 'the athlete's heart') or pathological stimuli (e.g. pressure or volume overload). Physiological hypertrophy is characterized by the normal organization of sarcomeres and fibres, normal or enhanced cardiac function and a relatively normal pattern of cardiac gene expression; whereas pathological hypertrophy is associated with an altered pattern of cardiac gene expression, fibrosis, cardiac dysfunction and increased mortality. Previously, an unresolved question in cardiac biology was whether distinct signalling pathways are responsible for the development of pathological and physiological cardiac hypertrophy. Recent studies have identified several signalling pathways that play unique roles in the regulation of pathological and physiological cardiac hypertrophy. This review focuses largely on the role of the insulin-like growth factor 1 (IGF1)/phosphoinositide-3-kinase (PI3K) pathway in mediating physiological cardiac growth.


Asunto(s)
Cardiomegalia/patología , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Cardiomegalia/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal
19.
Proc Natl Acad Sci U S A ; 103(39): 14471-6, 2006 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-16983087

RESUMEN

An important event in the pathogenesis of heart failure is the development of pathological cardiac hypertrophy. In cultured cardiomyocytes, the transcription factor Gata4 is required for agonist-induced hypertrophy. We hypothesized that, in the intact organism, Gata4 is an important regulator of postnatal heart function and of the hypertrophic response of the heart to pathological stress. To test this hypothesis, we studied mice heterozygous for deletion of the second exon of Gata4 (G4D). At baseline, G4D mice had mild systolic and diastolic dysfunction associated with reduced heart weight and decreased cardiomyocyte number. After transverse aortic constriction (TAC), G4D mice developed overt heart failure and eccentric cardiac hypertrophy, associated with significantly increased fibrosis and cardiomyocyte apoptosis. Inhibition of apoptosis by overexpression of the insulin-like growth factor 1 receptor prevented TAC-induced heart failure in G4D mice. Unlike WT-TAC controls, G4D-TAC cardiomyocytes hypertrophied by increasing in length more than width. Gene expression profiling revealed up-regulation of genes associated with apoptosis and fibrosis, including members of the TGF-beta pathway. Our data demonstrate that Gata4 is essential for cardiac function in the postnatal heart. After pressure overload, Gata4 regulates the pattern of cardiomyocyte hypertrophy and protects the heart from load-induced failure.


Asunto(s)
Gasto Cardíaco Bajo/inducido químicamente , Gasto Cardíaco Bajo/prevención & control , Factor de Transcripción GATA4/metabolismo , Corazón/fisiología , Presión Ventricular/fisiología , Animales , Aorta/fisiología , Apoptosis , Cardiomegalia/patología , Células Cultivadas , Diástole/fisiología , Fibrosis , Factor de Transcripción GATA4/genética , Expresión Génica , Regulación de la Expresión Génica , Corazón/fisiopatología , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor IGF Tipo 1/metabolismo , Sístole/fisiología
20.
Development ; 133(18): 3607-18, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914500

RESUMEN

Cardiac malformations due to aberrant development of the atrioventricular (AV) valves are among the most common forms of congenital heart disease. At localized swellings of extracellular matrix known as the endocardial cushions, the endothelial lining of the heart undergoes an epithelial to mesenchymal transition (EMT) to form the mesenchymal progenitors of the AV valves. Further growth and differentiation of these mesenchymal precursors results in the formation of portions of the atrial and ventricular septae, and the generation of thin, pliable valves. Gata4, which encodes a zinc finger transcription factor, is expressed in the endothelium and mesenchyme of the AV valves. Using a Tie2-Cre transgene, we selectively inactivated Gata4 within endothelial-derived cells. Mutant endothelium failed to undergo EMT, resulting in hypocellular cushions. Mutant cushions had decreased levels of Erbb3, an EGF-family receptor essential for EMT in the atrioventricular cushions. In Gata4 mutant embryos, Erbb3 downregulation was associated with impaired activation of Erk, which is also required for EMT. Expression of a Gata4 mutant protein defective in interaction with Friend of Gata (FOG) cofactors rescued the EMT defect, but resulted in a decreased proliferation of mesenchyme and hypoplastic cushions that failed to septate the ventricular inlet. We demonstrate two novel functions of Gata4 in development of the AV valves. First, Gata4 functions as an upstream regulator of an Erbb3-Erk pathway necessary for EMT, and second, Gata4 acts to promote cushion mesenchyme growth and remodeling.


Asunto(s)
Endotelio/metabolismo , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica/genética , Válvulas Cardíacas/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Endotelio/citología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Factor de Transcripción GATA4/fisiología , Válvulas Cardíacas/embriología , Hibridación in Situ , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Morfogénesis/genética , Morfogénesis/fisiología , Mutación/genética , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA