Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 122(8): 1052-1068, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29535165

RESUMEN

RATIONALE: Cardiac fibrosis plays a critical role in the pathogenesis of heart failure. Excessive accumulation of extracellular matrix (ECM) resulting from cardiac fibrosis impairs cardiac contractile function and increases arrhythmogenicity. Current treatment options for cardiac fibrosis, however, are limited, and there is a clear need to identify novel mediators of cardiac fibrosis to facilitate the development of better therapeutics. Exploiting coexpression gene network analysis on RNA sequencing data from failing human heart, we identified TXNDC5 (thioredoxin domain containing 5), a cardiac fibroblast (CF)-enriched endoplasmic reticulum protein, as a potential novel mediator of cardiac fibrosis, and we completed experiments to test this hypothesis directly. OBJECTIVE: The objective of this study was to determine the functional role of TXNDC5 in the pathogenesis of cardiac fibrosis. METHODS AND RESULTS: RNA sequencing and Western blot analyses revealed that TXNDC5 mRNA and protein were highly upregulated in failing human left ventricles and in hypertrophied/failing mouse left ventricle. In addition, cardiac TXNDC5 mRNA expression levels were positively correlated with those of transcripts encoding transforming growth factor ß1 and ECM proteins in vivo. TXNDC5 mRNA and protein were increased in human CF (hCF) under transforming growth factor ß1 stimulation in vitro. Knockdown of TXNDC5 attenuated transforming growth factor ß1-induced hCF activation and ECM protein upregulation independent of SMAD3 (SMAD family member 3), whereas increasing expression of TXNDC5 triggered hCF activation and proliferation and increased ECM protein production. Further experiments showed that TXNDC5, a protein disulfide isomerase, facilitated ECM protein folding and that depletion of TXNDC5 led to ECM protein misfolding and degradation in CF. In addition, TXNDC5 promotes hCF activation and proliferation by enhancing c-Jun N-terminal kinase activity via increased reactive oxygen species, derived from NAD(P)H oxidase 4. Transforming growth factor ß1-induced TXNDC5 upregulation in hCF was dependent on endoplasmic reticulum stress and activating transcription factor 6-mediated transcriptional control. Targeted disruption of Txndc5 in mice (Txndc5-/-) revealed protective effects against isoproterenol-induced cardiac hypertrophy, reduced fibrosis (by ≈70%), and markedly improved left ventricle function; post-isoproterenol left ventricular ejection fraction was 59.1±1.5 versus 40.1±2.5 (P<0.001) in Txndc5-/- versus wild-type mice, respectively. CONCLUSIONS: The endoplasmic reticulum protein TXNDC5 promotes cardiac fibrosis by facilitating ECM protein folding and CF activation via redox-sensitive c-Jun N-terminal kinase signaling. Loss of TXNDC5 protects against ß agonist-induced cardiac fibrosis and contractile dysfunction. Targeting TXNDC5, therefore, could be a powerful new therapeutic approach to mitigate excessive cardiac fibrosis, thereby improving cardiac function and outcomes in patients with heart failure.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/patología , Proteína Disulfuro Isomerasas/fisiología , Pliegue de Proteína , Tiorredoxinas/fisiología , Factor de Transcripción Activador 6/biosíntesis , Factor de Transcripción Activador 6/genética , Animales , Cardiomiopatía Hipertrófica/patología , Células Cultivadas , Fibroblastos/patología , Fibrosis/metabolismo , Regulación de la Expresión Génica , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/patología , Humanos , Isoproterenol/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , NADPH Oxidasa 4/biosíntesis , NADPH Oxidasa 4/genética , Células 3T3 NIH , Oxidación-Reducción , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/genética
2.
Circulation ; 129(9): 1009-21, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24429688

RESUMEN

BACKGROUND: Microarrays have been used extensively to profile transcriptome remodeling in failing human heart, although the genomic coverage provided is limited and fails to provide a detailed picture of the myocardial transcriptome landscape. Here, we describe sequencing-based transcriptome profiling, providing comprehensive analysis of myocardial mRNA, microRNA (miRNA), and long noncoding RNA (lncRNA) expression in failing human heart before and after mechanical support with a left ventricular (LV) assist device (LVAD). METHODS AND RESULTS: Deep sequencing of RNA isolated from paired nonischemic (NICM; n=8) and ischemic (ICM; n=8) human failing LV samples collected before and after LVAD and from nonfailing human LV (n=8) was conducted. These analyses revealed high abundance of mRNA (37%) and lncRNA (71%) of mitochondrial origin. miRNASeq revealed 160 and 147 differentially expressed miRNAs in ICM and NICM, respectively, compared with nonfailing LV. Among these, only 2 (ICM) and 5 (NICM) miRNAs are normalized with LVAD. RNASeq detected 18 480, including 113 novel, lncRNAs in human LV. Among the 679 (ICM) and 570 (NICM) lncRNAs differentially expressed with heart failure, ≈10% are improved or normalized with LVAD. In addition, the expression signature of lncRNAs, but not miRNAs or mRNAs, distinguishes ICM from NICM. Further analysis suggests that cis-gene regulation represents a major mechanism of action of human cardiac lncRNAs. CONCLUSIONS: The myocardial transcriptome is dynamically regulated in advanced heart failure and after LVAD support. The expression profiles of lncRNAs, but not mRNAs or miRNAs, can discriminate failing hearts of different pathologies and are markedly altered in response to LVAD support. These results suggest an important role for lncRNAs in the pathogenesis of heart failure and in reverse remodeling observed with mechanical support.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Insuficiencia Cardíaca/metabolismo , Corazón Auxiliar , Corazón/fisiopatología , ARN no Traducido/metabolismo , Análisis de Secuencia de ARN/métodos , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Insuficiencia Cardíaca/terapia , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Miocardio/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , ARN Mitocondrial
3.
J Biol Chem ; 287(46): 38901-12, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23019328

RESUMEN

Sarcolemmal CD36 facilitates myocardial fatty acid (FA) uptake, which is markedly reduced in CD36-deficient rodents and humans. CD36 also mediates signal transduction events involving a number of cellular pathways. In taste cells and macrophages, CD36 signaling was recently shown to regulate store-responsive Ca(2+) flux and activation of Ca(2+)-dependent phospholipases A(2) that cycle polyunsaturated FA into phospholipids. It is unknown whether CD36 deficiency influences myocardial Ca(2+) handling and phospholipid metabolism, which could compromise the heart, typically during stresses. Myocardial function was examined in fed or fasted (18-22 h) CD36(-/-) and WT mice. Echocardiography and telemetry identified conduction anomalies that were associated with the incidence of sudden death in fasted CD36(-/-) mice. No anomalies or death occurred in WT mice during fasting. Optical imaging of perfused hearts from fasted CD36(-/-) mice documented prolongation of Ca(2+) transients. Consistent with this, knockdown of CD36 in cardiomyocytes delayed clearance of cytosolic Ca(2+). Hearts of CD36(-/-) mice (fed or fasted) had 3-fold higher SERCA2a and 40% lower phospholamban levels. Phospholamban phosphorylation by protein kinase A (PKA) was enhanced after fasting reflecting increased PKA activity and cAMP levels in CD36(-/-) hearts. Abnormal Ca(2+) homeostasis in the CD36(-/-) myocardium associated with increased lysophospholipid content and a higher proportion of 22:6 FA in phospholipids suggests altered phospholipase A(2) activity and changes in membrane dynamics. The data support the role of CD36 in coordinating Ca(2+) homeostasis and lipid metabolism and the importance of this role during myocardial adaptation to fasting. Potential relevance of the findings to CD36-deficient humans would need to be determined.


Asunto(s)
Antígenos CD36/biosíntesis , Calcio/metabolismo , Miocardio/metabolismo , Fosfolípidos/metabolismo , Alimentación Animal , Animales , AMP Cíclico/metabolismo , Electrocardiografía/métodos , Ácidos Grasos Insaturados/metabolismo , Homeostasis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfolípidos/química , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores de Tiempo
4.
J Surg Res ; 178(2): 554-62, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22632938

RESUMEN

BACKGROUND: The purpose of this investigation was to characterize differential right atrial (RA) and ventricular (RV) molecular changes in Ca(2+)-handling proteins consequent to RV pressure overload and hypertrophy in two common, yet distinct models of pulmonary hypertension: dehydromonocrotaline (DMCT) toxicity and pulmonary artery (PA) banding. METHODS: A total of 18 dogs underwent sternotomy in four groups: (1) DMCT toxicity (n = 5), (2) mild PA banding over 10 wk to match the RV pressure rise with DMCT (n = 5); (3) progressive PA banding to generate severe RV overload (n = 4); and (4) sternotomy only (n = 4). RESULTS: In the right ventricle, with DMCT, there was no change in sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) or phospholamban (PLB), but we saw a trend toward down-regulation of phosphorylated PLB at serine-16 (p[Ser-16]PLB) (P = 0.07). Similarly, with mild PA banding, there was no change in SERCA or PLB, but p(Ser-16)PLB was down-regulated by 74% (P < 0.001). With severe PA banding, there was no change in PLB, but SERCA fell by 57% and p(Ser-16)PLB fell by 67% (P < 0.001). In the right atrium, with DMCT, there were no significant changes. With both mild and severe PA banding, p(Ser-16)PLB fell (P < 0.001), but SERCA and PLB did not change. CONCLUSIONS: Perturbations in Ca(2+)-handling proteins depend on the degree of RV pressure overload and the model used to mimic the RV effects of pulmonary hypertension. They are similar, but blunted, in the atrium compared with the ventricle.


Asunto(s)
Calcio/metabolismo , Ventrículos Cardíacos/fisiopatología , Disfunción Ventricular Derecha/fisiopatología , Presión Ventricular , Animales , Proteínas de Unión al Calcio/análisis , Modelos Animales de Enfermedad , Perros , Monocrotalina/análogos & derivados , Monocrotalina/toxicidad , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/análisis
5.
J Proteome Res ; 10(3): 1098-109, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21158428

RESUMEN

Connexin43 (Cx43) is a major cardiac gap junction channel protein required for normal electrical and contractile activity. Gap junction channel assembly, function, and turnover are regulated by phosphorylation under both normal and disease conditions. The carboxyl terminus (CT) of Cx43 contains numerous amino acid residues that are phosphorylated by protein kinases. However, our knowledge of the specific residues and kinases involved is incomplete. The objective of this study was to identify amino acid residues in the Cx43-CT that are targets of the multifunctional protein kinase, Ca(2+)/calmodulin protein kinase II (CaMKII), an enzyme known to play critical roles in Ca(2+) homeostasis, transcription, apoptosis, and ischemic heart disease. We subjected fusion protein containing the Cx43-CT to phosphorylation by CaMKII in vitro, digestion with Lys-C and trypsin followed by enrichment for phosphorylated peptides using TiO(2), and analysis in an LTQ XL Orbitrap with collision-induced dissociation and electron transfer dissociation. We deduced the sites of modification by interpreting tandem spectra from these "orthogonal" methods of gas phase peptide fragmentation. We have identified 15 serine residues, including one novel site, in the Cx43-CT that are phosphorylated by CaMKII, the activity of which may be important in regulating Cx43 in normal and diseased hearts.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/química , Conexina 43/química , Espectrometría de Masas/métodos , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Fosfopéptidos/química , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 298(2): H477-87, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19966054

RESUMEN

In addition to mediating cell-to-cell electrical coupling, gap junctions are important in tissue repair, wound healing, and scar formation. The expression and distribution of connexin43 (Cx43), the major gap junction protein expressed in the heart, are altered substantially after myocardial infarction (MI); however, the effects of Cx43 remodeling on wound healing and the attendant ventricular dysfunction are incompletely understood. Cx43-deficient and wild-type mice were subjected to proximal ligation of the anterior descending coronary artery and followed for 6 days or 4 wk to test the hypothesis that reduced expression of Cx43 influences wound healing, fibrosis, and ventricular remodeling after MI. We quantified the progression of infarct healing by measuring neutrophil expression, collagen content, and myofibroblast expression. We found significantly reduced transformation of fibroblasts to myofibroblasts at 6 days and significantly reduced collagen deposition both in the infarct at 6 days and at 4 wk in the noninfarcted region of Cx43-deficient mice. As expected, transforming growth factor (TGF)-beta, a profibrotic cytokine, was dramatically upregulated in MI hearts, but its phosphorylated comediator (pSmad) was significantly downregulated in the nuclei of Cx43-deficient hearts post-MI, suggesting that downstream signaling of TGF-beta is diminished substantially in Cx43-deficient hearts. This diminution in profibrotic TGF-beta signaling resulted in the attenuation of adverse structural remodeling as assessed by echocardiography. These findings suggest that efforts to enhance the expression of Cx43 to maintain intercellular coupling or reduce susceptibility to arrhythmias should be met with caution until the role of Cx43 in infarct healing is fully understood.


Asunto(s)
Conexina 43/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Ventricular/fisiología , Animales , Proliferación Celular , Cicatriz/etiología , Cicatriz/metabolismo , Cicatriz/patología , Conexina 43/genética , Modelos Animales de Enfermedad , Fibroblastos/patología , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Ratones , Ratones Noqueados , Infarto del Miocardio/patología , Factores de Tiempo , Cicatrización de Heridas/fisiología
7.
J Mol Cell Cardiol ; 45(3): 420-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18639555

RESUMEN

Ca(2+)/calmodulin-dependent protein kinase II is a multifunctional serine/threonine kinase with diverse cardiac roles including regulation of excitation contraction, transcription, and apoptosis. Dynamic regulation of CaMKII activity occurs in cardiac disease and is linked to specific disease phenotypes through its effects on ion channels, transporters, transcription and cell death pathways. Recent mathematical models of the cardiomyocyte have incorporated limited elements of CaMKII signaling to advance our understanding of how CaMKII regulates cardiac contractility and excitability. Given the importance of CaMKII in cardiac disease, it is imperative that computer models evolve to capture the dynamic range of CaMKII activity. In this study, using mathematical modeling combined with biochemical and imaging techniques, we test the hypothesis that CaMKII signaling in the canine infarct border zone (BZ) contributes to impaired calcium homeostasis and electrical remodeling. We report that the level of CaMKII autophosphorylation is significantly increased in the BZ region. Computer simulations using an updated mathematical model of CaMKII signaling reproduce abnormal Ca(2+) transients and action potentials characteristic of the BZ. Our simulations show that CaMKII hyperactivity contributes to abnormal Ca(2+) homeostasis and reduced action potential upstroke velocity due to effects on I(Na) gating kinetics. In conclusion, we present a new mathematical tool for studying effects of CaMKII signaling on cardiac excitability and contractility over a dynamic range of kinase activities. Our experimental and theoretical findings establish abnormal CaMKII signaling as an important component of remodeling in the canine BZ.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Calcio/metabolismo , Modelos Animales de Enfermedad , Homeostasis/fisiología , Modelos Cardiovasculares , Infarto del Miocardio/metabolismo , Pericardio/enzimología , Sodio/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Perros , Activación Enzimática/fisiología , Líquido Intracelular/enzimología , Líquido Intracelular/metabolismo , Infarto del Miocardio/patología , Pericardio/citología , Pericardio/patología , Fosforilación , Conejos
8.
Circ Res ; 99(11): 1216-24, 2006 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-17053190

RESUMEN

Atrial tissue expresses both connexin 40 (Cx40) and 43 (Cx43) proteins. To assess the relative roles of Cx40 and Cx43 in atrial electrical propagation, we synthesized cultured strands of atrial myocytes derived from mice with genetic deficiency in Cx40 or Cx43 expression and measured propagation velocity (PV) by high-resolution optical mapping of voltage-sensitive dye fluorescence. The amount of Cx40 and/or Cx43 in gap junctions was measured by immunohistochemistry and total or sarcolemmal Cx43 or Cx40 protein by immunoblotting. Progressive genetic reduction in Cx43 expression decreased PV from 34+/-6 cm/sec in Cx43(+/+) to 30+/-8 cm/sec in Cx43(+/-) and 19+/-11 cm/sec in Cx43(-/-) cultures. Concomitantly, the cell area occupied by Cx40 immunosignal in gap junctions decreased from 2.0+/-1.6% in Cx43(+/+) to 1.7+/-0.5% in Cx43(+/-) and 1.0+/-0.2% in Cx43(-/-) strands. In contrast, progressive genetic reduction in Cx40 expression increased PV from 30+/-2 cm/sec in Cx40(+/+) to 40+/-7 cm/sec in Cx40(+/-) and 45+/-10 cm/sec in Cx40(-/-) cultures. Concomitantly, the cell area occupied by Cx43 immunosignal in gap junctions increased from 1.2+/-0.9% in Cx40(+/+) to 2.8+/-1.4% in Cx40(+/-) and 3.1+/-0.6% in Cx40(-/-) cultures. In accordance with the immunostaining results, immunoblots of the Triton X-100-insoluble fraction revealed an increase of Cx43 in gap junctions in extracts from Cx40-ablated atria, whereas total cellular Cx43 remained unchanged. Our results suggest that the relative abundance of Cx43 and Cx40 is an important determinant of atrial impulse propagation in neonatal hearts, whereby dominance of Cx40 decreases and dominance of Cx43 increases local propagation velocity.


Asunto(s)
Conexina 43/fisiología , Conexinas/fisiología , Sistema de Conducción Cardíaco/fisiología , Miocitos Cardíacos/fisiología , Animales , Animales Recién Nacidos , Función Atrial , Conexina 43/deficiencia , Electrofisiología , Feto , Immunoblotting , Inmunohistoquímica/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Coloración y Etiquetado , Factores de Tiempo , Proteína alfa-5 de Unión Comunicante
9.
Circ Arrhythm Electrophysiol ; 11(1): e005914, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29311162

RESUMEN

BACKGROUND: Myocardial, transient, outward currents, Ito, have been shown to play pivotal roles in action potential (AP) repolarization and remodeling in animal models. The properties and contribution of Ito to left ventricular (LV) repolarization in the human heart, however, are poorly defined. METHODS AND RESULTS: Whole-cell, voltage-clamp recordings, acquired at physiological (35°C to 37°C) temperatures, from myocytes isolated from the LV of nonfailing human hearts identified 2 distinct transient currents, Ito,fast (Ito,f) and Ito,slow (Ito,s), with significantly (P<0.0001) different rates of recovery from inactivation and pharmacological sensitives: Ito,f recovers in ≈10 ms, 100× faster than Ito,s, and is selectively blocked by the Kv4 channel toxin, SNX-482. Current-clamp experiments revealed regional differences in AP waveforms, notably a phase 1 notch in LV subepicardial myocytes. Dynamic clamp-mediated addition/removal of modeled human ventricular Ito,f, resulted in hyperpolarization or depolarization, respectively, of the notch potential, whereas slowing the rate of Ito,f inactivation resulted in AP collapse. AP-clamp experiments demonstrated that changes in notch potentials modified the time course and amplitudes of voltage-gated Ca2+ currents, ICa. In failing LV subepicardial myocytes, Ito,f was reduced and Ito,s was increased, notch and plateau potentials were depolarized (P<0.0001) and AP durations were prolonged (P<0.001). CONCLUSIONS: Ito,f and Ito,s are differentially expressed in nonfailing human LV, contributing to regional heterogeneities in AP waveforms. Ito,f regulates notch and plateau potentials and modulates the time course and amplitude of ICa. Slowing Ito,f inactivation results in dramatic AP shortening. Remodeling of Ito,f in failing human LV subepicardial myocytes attenuates transmural differences in AP waveforms.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Miocardio/metabolismo , Potasio/metabolismo , Femenino , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/patología , Humanos , Masculino , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , Miocardio/patología , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo
10.
Heart Rhythm ; 4(9): 1183-93, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17765619

RESUMEN

BACKGROUND: Ischemic preconditioning delays the onset of electrical uncoupling and prevents loss of the primary ventricular gap junction protein connexin 43 (Cx43) from gap junctions during subsequent ischemia. OBJECTIVE: To test the hypothesis that these effects are mediated by protein kinase C epsilon (PKCepsilon), we studied isolated Langendorff-perfused hearts from mice with homozygous germline deletion of PKCepsilon (PKCepsilon-KO). METHODS: Cx43 phosphorylation and distribution were measured by quantitative immunoblotting and confocal microscopy. Changes in electrical coupling were monitored using the 4-electrode technique to measure whole-tissue resistivity. RESULTS: The amount of Cx43 located in gap junctions, measured by confocal microscopy under basal conditions, was significantly greater in PKCepsilon-KO hearts compared with wild-type, but total Cx43 content measured by immunoblotting was not different. These unanticipated results indicate that PKCepsilon regulates subcellular distribution of Cx43 under normal conditions. Preconditioning prevented loss of Cx43 from gap junctions during ischemia in wild-type but not PKCepsilon-KO hearts. Specific activation of PKCepsilon, but not PKCdelta, also prevented ischemia-induced loss of Cx43 from gap junctions. Preconditioning delayed the onset of uncoupling in wild-type but hastened uncoupling in PKCepsilon-KO hearts. Cx43 phosphorylation at the PKC site Ser368 increased 5-fold after ischemia in wild-type hearts, and surprisingly, by nearly 10-fold in PKCepsilon-KO hearts. Preconditioning prevented phosphorylation of Cx43 in gap junction plaques at Ser368 in wild-type but not PKCepsilon-KO hearts. CONCLUSION: Taken together, these results indicate that PKCepsilon plays a critical role in preconditioning to preserve Cx43 signal in gap junctions and delay electrical uncoupling during ischemia.


Asunto(s)
Conexina 43/metabolismo , Precondicionamiento Isquémico Miocárdico , Isquemia Miocárdica/metabolismo , Proteína Quinasa C-epsilon/fisiología , Animales , Impedancia Eléctrica , Uniones Comunicantes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Reperfusión Miocárdica , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Conejos
11.
Cardiovasc Pathol ; 16(3): 159-64, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17502245

RESUMEN

BACKGROUND: Reduced expression of the major gap junction protein connexin 43 (Cx43) in the failing human heart may lead to arrhythmias and sudden cardiac death. Cx43 interacts with the actin binding protein, zonula occludens-1 (ZO-1), and it has recently been demonstrated that ZO-1 regulates the formation and function of Cx43 gap junctions. We hypothesize that normal expression of ZO-1 and its interaction with Cx43 are required for appropriate assembly and function of Cx43 gap junctions in the heart. Here, we determined whether expression of ZO-1 is altered in patients with heart failure. METHODS: We examined ventricular myocardium from hearts of patients in end-stage heart failure, obtained at transplant, for ZO-1 expression by immunohistochemistry. We also subjected lysates made from this tissue to immunoblotting to determine the level of ZO-1 expression. RESULTS AND CONCLUSIONS: ZO-1 was found at 96% of the intercalated discs in nonfailing control human hearts, where it colocalized with Cx43. In contrast, there was ZO-1 immunostaining at 5% of intercalated discs in failing hearts, coincident with a reduction in Cx43 staining in intercalated discs. Immunoblotting analysis showed that there was a 95% reduction in ZO-1 expression in human heart failure. Loss of ZO-1 at intercalated discs in heart failure may play a critical role in remodeling of Cx43 gap junctions, which may contribute to abnormal impulse propagation and arrhythmogenesis, thereby predisposing patients in heart failure to sudden cardiac death.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Proteínas de la Membrana/metabolismo , Miocardio/metabolismo , Fosfoproteínas/metabolismo , Adulto , Conexina 43/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/cirugía , Trasplante de Corazón , Ventrículos Cardíacos/patología , Humanos , Masculino , Persona de Mediana Edad , Miocardio/patología , Proteína de la Zonula Occludens-1
12.
Circ Res ; 96(5): 558-66, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15705967

RESUMEN

To elucidate mechanisms responsible for mechanotransduction in the heart and define the effects of remodeling of the extracellular matrix, we cultured neonatal rat ventricular myocytes on native type I collagen, fibronectin, or denatured collagen and subjected them to uniaxial, pulsatile stretch. Changes in expression of the cardiac gap junction protein, Cx43, were measured by confocal microscopy and immunoblotting. Cells grown on fibronectin or denatured collagen exhibited significantly greater Cx43 expression than cells grown on native collagen. Stretch induced a approximately 2-fold increase in Cx43 expression in cells grown on native collagen but no increase in cells grown on fibronectin or denatured collagen. Incubation of cells on native collagen with a peptide containing the arginine-glycine-aspartate (RGD) motif upregulated Cx43 expression equivalent to that induced by stretch. Nonselective activation of integrin signaling with MnCl2 also upregulated Cx43 expression in cells grown on native collagen. This effect was blocked completely by pretreatment with anti-beta1 integrin antibody but not by anti-beta3 integrin antibody. Stretch led to a marked increase in beta1 integrin immunofluorescent signal in cells grown on native collagen but not in cells grown on fibronectin or denatured collagen. Stretch-induced upregulation of Cx43 was also blocked by anti-beta1 integrin antibody. Thus, matrix protein-myocyte interactions regulate Cx43 expression via beta1 integrin signaling initiated by mechanical stimulation in cells grown on native type I collagen, or by RGD-integrin signaling independent of mechanical stress in cells grown on fibronectin or denatured collagen. Changes in the composition of the extracellular matrix may affect electrical coupling in cardiac myocytes.


Asunto(s)
Colágeno Tipo I/fisiología , Conexina 43/biosíntesis , Fibronectinas/fisiología , Uniones Comunicantes/fisiología , Regulación de la Expresión Génica/fisiología , Integrina beta1/fisiología , Miocitos Cardíacos/metabolismo , Oligopéptidos/farmacología , Estrés Mecánico , Animales , Anticuerpos Monoclonales/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Conexina 43/genética , Medios de Cultivo , Uniones Comunicantes/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Integrina beta1/inmunología , Integrina beta3/inmunología , Cloruro de Magnesio/farmacología , Microscopía Confocal , Microscopía Fluorescente , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/farmacología
13.
Circ Res ; 97(12): 1342-50, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16293790

RESUMEN

Previous studies have demonstrated a role for voltage-gated K+ (Kv) channel alpha subunits of the Kv4 subfamily in the generation of rapidly inactivating/recovering cardiac transient outward K+ current, I(to,f), channels. Biochemical studies suggest that mouse ventricular I(to,f) channels reflect the heteromeric assembly of Kv4.2 and Kv4.3 with the accessory subunits, KChIP2 and Kvbeta1, and that Kv4.2 is the primary determinant of regional differences in (mouse ventricular) I(to,f) densities. Interestingly, the phenotypic consequences of manipulating I(to,f) expression in different mouse models are distinct. In the experiments here, the effects of the targeted deletion of Kv4.2 (Kv4.2(-/-)) were examined. Unexpectedly, voltage-clamp recordings from Kv4.2(-/-) ventricular myocytes revealed that I(to,f) is eliminated. In addition, the slow transient outward K+ current, I(to,s), and the Kv1.4 protein (which encodes I(to,s)) are upregulated in Kv4.2(-/-) ventricles. Although Kv4.3 mRNA/protein expression is not measurably affected, KChIP2 expression is markedly reduced in Kv4.2(-/-) ventricles. Similar to Kv4.3, expression of Kvbeta1, as well as Kv1.5 and Kv2.1, is similar in wild-type and Kv4.2(-/-) ventricles. In addition, and in marked contrast to previous findings in mice expressing a truncated Kv4.2 transgene, the elimination I(to,f) in Kv4.2(-/-) mice does not result in ventricular hypertrophy. Taken together, these findings demonstrate not only an essential role for Kv4.2 in the generation of mouse ventricular I(to,f) channels but also that the loss of I(to,f) per se does not have overt pathophysiological consequences.


Asunto(s)
Cardiomegalia/etiología , Ventrículos Cardíacos/patología , Miocitos Cardíacos/fisiología , Canales de Potasio Shal/fisiología , Remodelación Ventricular , Animales , Electrocardiografía , Proteínas de Interacción con los Canales Kv/fisiología , Ratones , Ratones Noqueados , Canales de Potasio Shal/genética
14.
Circ Res ; 96(2): 225-33, 2005 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15618539

RESUMEN

Evidence is emerging that systemic metabolic disturbances contribute to cardiac myocyte dysfunction and clinically apparent heart failure, independent of associated coronary artery disease. To test the hypothesis that perturbation of lipid homeostasis in cardiomyocytes contributes to cardiac dysfunction, we engineered transgenic mice with cardiac-specific overexpression of fatty acid transport protein 1 (FATP1) using the alpha-myosin heavy chain gene promoter. Two independent transgenic lines demonstrate 4-fold increased myocardial free fatty acid (FFA) uptake that is consistent with the known function of FATP1. Increased FFA uptake in this model likely contributes to early cardiomyocyte FFA accumulation (2-fold increased) and subsequent increased cardiac FFA metabolism (2-fold). By 3 months of age, transgenic mice have echocardiographic evidence of impaired left ventricular filling and biatrial enlargement, but preserved systolic function. Doppler tissue imaging and hemodynamic studies confirm that these mice have predominantly diastolic dysfunction. Furthermore, ambulatory ECG monitoring reveals prolonged QT(c) intervals, reflecting reductions in the densities of repolarizing, voltage-gated K+ currents in ventricular myocytes. Our results show that in the absence of systemic metabolic disturbances, such as diabetes or hyperlipidemia, perturbation of cardiomyocyte lipid homeostasis leads to cardiac dysfunction with pathophysiological findings similar to those in diabetic cardiomyopathy. Moreover, the MHC-FATP model supports a role for FATPs in FFA import into the heart in vivo.


Asunto(s)
Cardiomiopatías/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Proteínas de Transporte de Membrana/fisiología , Miocitos Cardíacos/metabolismo , Animales , Transporte Biológico , Cardiomiopatías/genética , Cardiomiopatías/patología , Tamaño de la Célula , Diástole , Electrocardiografía , Proteínas de Transporte de Ácidos Grasos , Expresión Génica , Glucosa/metabolismo , Hipertrofia , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/patología , Técnicas de Placa-Clamp , Tomografía de Emisión de Positrones , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteínas Recombinantes de Fusión/fisiología
15.
Circ Res ; 90(6): 671-7, 2002 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-11934834

RESUMEN

Cardiac myocytes can rapidly adjust their expression of gap junction channel proteins in response to changes in load. Previously, we showed that after only 1 hour of linear pulsatile stretch (110% of resting cell length; 3 Hz), expression of connexin43 (Cx43) by cultured neonatal rat ventricular myocytes is increased by approximately 2-fold and impulse propagation is significantly more rapid. In the present study, we tested the hypothesis that vascular endothelial growth factor (VEGF), acting downstream of transforming growth factor-beta (TGF-beta), mediates stretch-induced upregulation of Cx43 expression by cardiac myocytes. Incubation of nonstretched cells with exogenous VEGF (100 ng/mL) or TGF-beta (10 ng/mL) for 1 hour increased Cx43 expression by approximately 1.8-fold, comparable to that observed in cells subjected to pulsatile stretch for 1 hour. Stretch-induced upregulation of Cx43 expression was blocked by either anti-VEGF antibody or anti-TGF-beta antibody. Stretch-induced enhancement of conduction was also blocked by anti-VEGF antibody. ELISA assay showed that VEGF was secreted into the culture medium during stretch. Furthermore, stretch-conditioned medium stimulated Cx43 expression in nonstretched cells. This effect was also blocked by anti-VEGF antibody. Upregulation of Cx43 expression stimulated by exogenous TGF-beta was blocked by anti-VEGF antibody, but VEGF-stimulation of Cx43 expression was not blocked by anti-TGF-beta antibody. Thus, stretch-induced upregulation of Cx43 expression is mediated, at least in part, by VEGF, which acts downstream of TGF-beta. Because the cultures contained only approximately 5% nonmyocytic cells, these results indicate that myocyte-derived VEGF, secreted in response to stretch, acts in an autocrine fashion to enhance intercellular coupling.


Asunto(s)
Conexina 43/fisiología , Factores de Crecimiento Endotelial/fisiología , Corazón/fisiología , Linfocinas/fisiología , Contracción Miocárdica/fisiología , Regulación hacia Arriba/fisiología , Animales , Comunicación Autocrina/fisiología , Células Cultivadas , Conexina 43/genética , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Linfocinas/metabolismo , Linfocinas/farmacología , Ratas , Ratas Endogámicas WKY , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
16.
Circ Res ; 95(2): 170-8, 2004 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-15192022

RESUMEN

To characterize the role of connexin43 (Cx43) as a determinant of cardiac propagation, we synthesized strands and pairs of ventricular myocytes from germline Cx43-/- mice. The amount of Cx43, Cx45, and Cx40 in gap junctions was analyzed by immunohistochemistry and confocal microscopy. Intercellular electrical conductance, gj, was measured by the dual-voltage clamp technique (DVC), and electrical propagation was assessed by multisite optical mapping of transmembrane potential using a voltage-sensitive dye. Compared with wild-type (Cx43+/+) strands, immunoreactive signal for Cx43 was reduced by 46% in Cx43+/- strands and was absent in Cx43-/- strands. Cx45 signal was reduced by 46% in Cx43+/- strands and to the limit of detection in Cx43-/- strands, but total Cx45 protein levels measured in immunoblots of whole cell homogenates were equivalent in all genotypes. Cx40 was detected in 2% of myocytes. Intercellular conductance, gj, was reduced by 32% in Cx43+/- cell pairs and by 96% in Cx43-/- cell pairs. The symmetrical dependence of gj on transjunctional voltage and properties of single-channel recordings indicated that Cx45 was the only remaining connexin in Cx43-/- cells. Propagation in Cx43-/- strands was very slow (2.1 cm/s versus 52 cm/s in Cx43+/+) and highly discontinuous, with simultaneous excitation within and long conduction delays (2 to 3 ms) between individual cells. Propagation was abolished by 1 mmol/L heptanol, indicating residual junctional coupling. In summary, knockout of Cx43 in ventricular myocytes leads to very slow conduction dependent on the presence of Cx45. Electrical field effect transmission does not contribute to propagation in synthetic strands.


Asunto(s)
Conexina 43/fisiología , Miocitos Cardíacos/fisiología , Potenciales de Acción , Animales , Células Cultivadas/fisiología , Conexina 43/genética , Conexinas/análisis , Conexinas/fisiología , Conductividad Eléctrica , Uniones Comunicantes/química , Uniones Comunicantes/fisiología , Mutación de Línea Germinal , Ventrículos Cardíacos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Complejos Multiproteicos , Técnicas de Placa-Clamp , Proteína alfa-5 de Unión Comunicante
17.
Cardiovasc Res ; 67(3): 539-47, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15913582

RESUMEN

OBJECTIVE: We studied a transgenic mouse model of human desmin-related cardiomyopathy with cardiac-specific expression of a 7-amino acid deletion mutation in desmin (D7-des) to test the hypothesis that impaired linkage between desmin and desmosomes alters expression and function of the electrical coupling protein, connexin43 (Cx43). METHODS: Expression of Cx43 and selected mechanical junctions proteins was characterized in left ventrices of D7-des and control mice by quantitative confocal microscopy and immunoblotting. Remodeling of gap junctions was also analyzed by electron microscopic morphometry. The electrophysiological phentoype of D7-des mice was characterized by electrocardiography and optical mapping of transmembrane voltage. RESULTS: Cx43 signal at intercalated disks was decreased by approximately 3-fold in D7-des ventricular tissue due to reductions in both gap junction number and size. Immunoreactive signal at cell-cell junctions was also reduced significantly for adhesion molecules and linker proteins of desmosomes and fascia adherens junctions. Electron microscopy showed decreased gap junction remodeling. However, immunoblotting showed that the total tissue content of Cx43 and mechanical junction proteins was not reduced, suggesting that diminished signal at cell-cell junctions was not due to insufficient protein expression, but to failure of these proteins to assemble properly within electrical and mechanical junctions. Remodeling of gap junctions in D7-des mice led to slowing of ventricular conduction as demonstrated by optical electrophysiological mapping. CONCLUSIONS: These results illustrate how a defect in a protein conventionally thought to fulfill a mechanical function in the heart can also lead to electrophysiological alterations that may contribute to arrhythmogenesis.


Asunto(s)
Cardiomiopatías/metabolismo , Desmina/genética , Uniones Comunicantes/química , Mutación , Contracción Miocárdica , Miocitos Cardíacos/química , Animales , Cardiomiopatías/fisiopatología , Desmina/análisis , Desmina/metabolismo , Electrofisiología , Uniones Comunicantes/metabolismo , Inmunohistoquímica/métodos , Ratones , Ratones Transgénicos , Microscopía Confocal , Microscopía Fluorescente , Modelos Animales , Miocitos Cardíacos/metabolismo
18.
J Am Coll Cardiol ; 41(4): 681-6, 2003 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-12598083

RESUMEN

OBJECTIVES: The purpose of this study was to define the role of cell-cell coupling as an independent determinant of infarct size following coronary occlusion. BACKGROUND: Electrical uncoupling induced by acute ischemia enhances arrhythmogenesis, but it may also protect the heart by limiting intercellular spread of chemical mediators of injury. METHODS: The left anterior descending coronary artery was ligated in wild-type (Cx43(+/+)) mice and Cx43-deficient (Cx43(+/-)) mice that are heterozygous for a null allele in the gene encoding the major gap junction channel protein, connexin43 (Cx43). Ventricular remodeling and infarct size were compared in both groups. RESULTS: Echocardiography at 1 and 10 weeks after infarction showed that left ventricular end-diastolic volume and mass increased and ejection fraction decreased in proportion to infarct size in both Cx43(+/-) and Cx43(+/+) hearts. However, infarct size measured histologically in healing infarcts (eight days after infarction) was 29% smaller in Cx43(+/-) hearts (17 +/- 14% of total left ventricular area, n = 30) than in Cx43(+/+) hearts (24 +/- 15%, n = 23; p = 0.037). Fully healed infarcts were smaller than healing infarcts, owing to resorption of necrotic tissue and maturation of scar, but infarct size at 10 weeks after coronary occlusion was still smaller (by 50%) in Cx43(+/-) hearts (6 +/- 5%, n = 9) compared with Cx43(+/+) hearts (12 +/- 7%, n = 17; p = 0.037). CONCLUSIONS: Cx43-deficient mice develop smaller infarcts than wild-type mice following coronary ligation. New therapies designed to decrease the risk of arrhythmias by enhancing intercellular communication could lead to larger infarcts caused by persistent coronary occlusion.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Conexina 43/farmacología , Conexina 43/fisiología , Enfermedad Coronaria/complicaciones , Enfermedad Coronaria/fisiopatología , Infarto del Miocardio/etiología , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/fisiología , Animales , Enfermedad Coronaria/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/patología , Índice de Severidad de la Enfermedad , Remodelación Ventricular/efectos de los fármacos
19.
Heart Rhythm ; 2(6): 650-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15922277

RESUMEN

Cardiac arrhythmias continue to pose a major medical challenge and significant public health burden. Atrial fibrillation, the most prevalent arrhythmia, affects more than two million Americans annually and is associated with a twofold increase in mortality. In addition, more than 250,000 Americans each year suffer ventricular arrhythmias, often resulting in sudden cardiac death. Despite the high incidence and societal impact of cardiac arrhythmias, presently there are insufficient insights into the molecular mechanisms involved in arrhythmia generation, propagation, and/or maintenance or into the molecular determinants of disease risk, prognosis, and progression. In addition, present therapeutic strategies for arrhythmia abatement often are ineffective or require palliative device therapy after persistent changes in the electrical and conduction characteristics of the heart have occurred, changes that appear to increase the risk for arrhythmia progression. This article reviews our present understanding of the complexity of mechanisms that regulate cardiac membrane excitability and cardiac function and explores the role of derangements in these mechanisms that interact to induce arrhythmogenic substrates. Approaches are recommended for future investigations focused on providing new mechanistic insights and therapeutic interventions.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Sistema de Conducción Cardíaco/fisiología , Arritmias Cardíacas/genética , Progresión de la Enfermedad , Matriz Extracelular/fisiología , Humanos , Canales Iónicos/fisiología , Pronóstico , Remodelación Ventricular/fisiología
20.
Cardiovasc Res ; 53(4): 921-35, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11922902

RESUMEN

OBJECTIVE: Adult ventricular myocytes express two gap junction channel proteins: connexin43 (Cx43) and connexin45 (Cx45). Cx43-deficient mice exhibit slow ventricular epicardial conduction, suggesting that Cx43 plays an important role in intercellular coupling in the ventricle. Cx45 is much less abundant than Cx43 in working ventricular myocytes. Its role in ventricular conduction has not been defined, nor is it known whether expression or distribution of Cx45 is altered in Cx43-deficient mice. The present study was undertaken to determine (1) whether expression of Cx45 is upregulated and (2) whether gap junction structure and distribution are altered in Cx43-deficient mice. METHODS: Ventricular tissue from neonatal Cx43(+/+), Cx43(+/-) and Cx43(-/-) and adult Cx43(+/+) and Cx43(+/-) mice was analyzed by immunoblotting and confocal immunofluorescence microscopy. RESULTS: Total Cx45 protein abundance measured by immunoblotting was not different in Cx43-deficient or null hearts compared to wild-type control hearts. However, the amount and distribution of Cx45 immunoreactive signal measured by quantitative confocal analysis were markedly reduced in both Cx43(+/-) and Cx43(-/-) hearts. CONCLUSION: Although the total content of Cx45 is not upregulated in Cx43-deficient hearts, the localization of Cx45 to cardiac gap junctions depends on the expression level of Cx43 and is dramatically altered in mice that express no Cx43.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Miocardio/metabolismo , Animales , Animales Recién Nacidos , Conexina 43/metabolismo , Conexinas/deficiencia , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Proteína alfa-5 de Unión Comunicante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA