Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochemistry ; 56(35): 4722-4731, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28782937

RESUMEN

Focal adhesion kinase (FAK) plays a key role in integrin and growth factor signaling pathways. FAK-related non-kinase (FRNK) is an endogenous inhibitor of FAK that shares its primary structure with the C-terminal third of FAK. FAK S910 phosphorylation is known to regulate FAK protein-protein interactions, but the role of the equivalent site on FRNK (S217) is unknown. Here we determined that S217 is highly phosphorylated by ERK in cultured rat aortic smooth muscle cells. Blocking phosphorylation by mutation (S217A) greatly increased FRNK inhibitory potency, resulting in strong inhibition of FAK autophosphorylation at Y397 and induction of smooth muscle cell apoptosis. FRNK has been proposed to compete for FAK anchoring sites in focal adhesions, but we did not detect displacement of FAK by WT-FRNK or superinhibitory S217A-FRNK. Instead, we found FRNK interacted directly with FAK, and this interaction is markedly strengthened for the superinhibitory S217A-FRNK. The results suggest that FRNK limits growth and survival signaling pathways by binding directly to FAK in an inhibitory complex, and this inhibition is relieved by phosphorylation of FRNK at S217.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Miocitos del Músculo Liso/enzimología , Inhibidores de Proteínas Quinasas/metabolismo , Animales , Quinasa 1 de Adhesión Focal , Músculo Liso Vascular/citología , Fosforilación , Ratas , Transducción de Señal/fisiología
2.
Psychopharmacology (Berl) ; 233(13): 2581-92, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27150078

RESUMEN

RATIONALE: Anxiety is a common comorbidity that develops after myocardial infarction and is now an established independent risk factor for cardiovascular mortality. OBJECTIVE: Here, we assessed anxiety and mapped neural activity of forebrain regions that regulate anxiety in a rat model of myocardial infarction in order to identify sites of dysregulation. METHODS: Anxiety responses to novel (open field) or aversive stimuli (discriminative auditory fear conditioning) were assessed in rats subjected to coronary artery ligation (CAL) or sham ligation. Forebrain metabolic activity was measured by cytochrome oxidase (CO) histochemistry. Changes in CO activity and the incidence of ventricular arrhythmias were also assessed during modulation of fear circuitry induced by electrical stimulation of the locus coeruleus. RESULTS: Coronary artery ligation had negligible effects on open-field behavior, but increased expression of learned fear and impaired fear cue discrimination. Cytochrome oxidase activity was increased in the medial prefrontal cortex and in the lateral amygdala after CAL. Locus coeruleus stimulation reduced CO activity in the infralimbic medial prefrontal cortex only in rats subjected to CAL. Stimulation of the LC also elicited new ventricular arrhythmias in rats subjected to CAL. CONCLUSION: Coronary artery ligation sensitizes the infralimbic medial prefrontal cortex to the inhibitory effects of locus coeruleus stimulation. Suppression of infralimbic medial prefrontal cortical activity may impair the ability of rats subjected to CAL to discriminate between cues that signal aversive and neutral events which, in turn, may promote excessive sympathetic activation of the cardiovascular system in response to innocuous stimuli.


Asunto(s)
Inhibición Psicológica , Locus Coeruleus/fisiología , Infarto del Miocardio/fisiopatología , Corteza Prefrontal/fisiología , Amígdala del Cerebelo/fisiología , Análisis de Varianza , Animales , Ansiedad/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Complejo IV de Transporte de Electrones/metabolismo , Conducta Exploratoria/fisiología , Miedo/fisiología , Masculino , Infarto del Miocardio/complicaciones , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
3.
J Mol Cell Cardiol ; 89(Pt B): 214-22, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26549358

RESUMEN

Cardiac structural changes associated with dilated cardiomyopathy (DCM) include cardiomyocyte hypertrophy and myocardial fibrosis. Connective tissue growth factor (CTGF) has been associated with tissue remodeling and is highly expressed in failing hearts. Our aim was to test if inhibition of CTGF would alter the course of cardiac remodeling and preserve cardiac function in the protein kinase Cε (PKCε) mouse model of DCM. Transgenic mice expressing constitutively active PKCε in cardiomyocytes develop cardiac dysfunction that was evident by 3 months of age, and that progressed to cardiac fibrosis, heart failure, and increased mortality. Beginning at 3 months of age, PKCε mice were treated with a neutralizing monoclonal antibody to CTGF (FG-3149) for an additional 3 months. CTGF inhibition significantly improved left ventricular (LV) systolic and diastolic functions in PKCε mice, and slowed the progression of LV dilatation. Using gene arrays and quantitative PCR, the expression of many genes associated with tissue remodeling was elevated in PKCε mice, but significantly decreased by CTGF inhibition. However total collagen deposition was not attenuated. The observation of significantly improved LV function by CTGF inhibition in PKCε mice suggests that CTGF inhibition may benefit patients with DCM. Additional studies to explore this potential are warranted.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Envejecimiento/patología , Animales , Anticuerpos Neutralizantes/farmacología , Cardiomiopatía Dilatada/patología , Colágeno/metabolismo , Diástole/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Perfilación de la Expresión Génica , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Proteína Quinasa C-epsilon/metabolismo , Transducción de Señal/efectos de los fármacos , Sístole/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
4.
Cardiovasc Res ; 106(3): 509-19, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25883219

RESUMEN

AIMS: Vascular injury leading to endothelial dysfunction is a characteristic feature of chronic renal disease, diabetes mellitus, and systemic inflammatory conditions, and predisposes to apoptosis and atherogenesis. Thus, endothelial dysfunction represents a potential therapeutic target for atherosclerosis prevention. The observation that activity of either protein kinase C epsilon (PKCε) or haem oxygenase-1 (HO-1) enhances endothelial cell (EC) resistance to inflammation and apoptosis led us to test the hypothesis that HO-1 is a downstream target of PKCε. METHODS AND RESULTS: Expression of constitutively active PKCε in human EC significantly increased HO-1 mRNA and protein, whereas conversely aortas or cardiac EC from PKCε-deficient mice exhibited reduced HO-1 when compared with wild-type littermates. Angiotensin II activated PKCε and induced HO-1 via a PKCε-dependent pathway. PKCε activation significantly attenuated TNFα-induced intercellular adhesion molecule-1, and increased resistance to serum starvation-induced apoptosis. These responses were reversed by the HO antagonist zinc protoporphyrin IX. Phosphokinase antibody array analysis identified CREB1((Ser133)) phosphorylation as a PKCε signalling intermediary, and cAMP response element-binding protein 1 (CREB1) siRNA abrogated PKCε-induced HO-1 up-regulation. Likewise, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) was identified as a PKCε target using nuclear translocation and DNA-binding assays, and Nrf2 siRNA prevented PKCε-mediated HO-1 induction. Moreover, depletion of CREB1 inhibited PKCε-induced Nrf2 DNA binding, suggestive of transcriptional co-operation between CREB1 and Nrf2. CONCLUSIONS: PKCε activity in the vascular endothelium regulates HO-1 via a pathway requiring CREB1 and Nrf2. Given the potent protective actions of HO-1, we propose that this mechanism is an important contributor to the emerging role of PKCε in the maintenance of endothelial homeostasis and resistance to injury.


Asunto(s)
Apoptosis , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células Endoteliales/enzimología , Hemo-Oxigenasa 1/biosíntesis , Inflamación/prevención & control , Proteínas de la Membrana/biosíntesis , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Transducción de Señal , Angiotensina II/farmacología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Activación Enzimática , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/enzimología , Inflamación/genética , Inflamación/patología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Fosforilación , Proteína Quinasa C-epsilon/genética , Protoporfirinas/farmacología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transcripción Genética , Transfección
5.
J Mol Cell Cardiol ; 72: 281-91, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24713463

RESUMEN

Up-regulation and activation of PYK2, a member of the FAK family of protein tyrosine kinases, is involved in the pathogenesis of left ventricular (LV) remodeling and heart failure (HF). PYK2 activation can be prevented by CRNK, the C-terminal domain of PYK2. We previously demonstrated that adenoviral-mediated CRNK gene transfer improved survival and LV function, and slowed LV remodeling in a rat model of coronary artery ligation-induced HF. We now interrogate whether cardiomyocyte-specific, transgenic CRNK expression prevents LV remodeling and HF in a mouse model of dilated cardiomyopathy (DCM) caused by constitutively active Protein Kinase Cε (caPKCε). Transgenic (TG; FVB/N background) mice were engineered to express rat CRNK under control of the α-myosin heavy chain promoter, and crossed with FVB/N mice with cardiomyocyte-specific expression of caPKCε to create double TG mice. LV structure, function, and gene expression were evaluated in all 4 groups (nonTG FVB/N; caPKCε(+/-); CRNK(+/-); and caPKCε×CRNK (PXC) double TG mice) at 1, 3, 6, 9 and 12mo of age. CRNK expression followed a Mendelian distribution, and CRNK mice developed and survived normally through 12mo. Cardiac structure, function and selected gene expression of CRNK mice were similar to nonTG littermates. CRNK had no effect on caPKCε expression and vice versa. PYK2 was up-regulated ~6-fold in caPKCε mice, who developed a non-hypertrophic, progressive DCM with reduced systolic (Contractility Index=151±5 vs. 90±4s(-1)) and diastolic (Tau=7.5±0.5 vs. 14.7±1.3ms) function, and LV dilatation (LV Remodeling Index (LVRI)=4.2±0.1 vs. 6.0±0.3 for FVB/N vs. caPKCε mice, respectively; P<0.05 for each at 12mo). In double TG PXC mice, CRNK expression significantly prolonged survival, improved contractile function (Contractile Index=115±8s(-1); Tau=9.5±1.0ms), and reduced LV remodeling (LVRI=4.9±0.1). Cardiomyocyte-specific expression of CRNK improves contractile function and slows LV remodeling in a mouse model of DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Quinasa 2 de Adhesión Focal/genética , Miocitos Cardíacos/metabolismo , Transgenes , Función Ventricular/fisiología , Remodelación Ventricular , Animales , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Quinasa 2 de Adhesión Focal/deficiencia , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Longevidad , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Regiones Promotoras Genéticas , Proteína Quinasa C-epsilon/deficiencia , Proteína Quinasa C-epsilon/genética , Estructura Terciaria de Proteína
6.
Pflugers Arch ; 466(6): 1101-11, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24515292

RESUMEN

In this brief review, recent evidence is presented to indicate a role for specific components of the cardiomyocyte costamere (and its related structure the focal adhesion complex of cultured cardiomyocytes) in initiating and sustaining the aberrant signal transduction that contributes to myocardial remodeling and the progression to heart failure (HF). Special attention is devoted to the focal adhesion kinase family of nonreceptor protein tyrosine kinases in bidirectional signal transduction during cardiac remodeling and HF progression. Finally, some speculations and directions for future study are provided for this rapidly developing field of research.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Insuficiencia Cardíaca/metabolismo , Mecanotransducción Celular , Animales , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Sarcómeros/metabolismo , Sarcómeros/fisiología
7.
Physiol Genomics ; 45(14): 597-605, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695888

RESUMEN

Human dilated cardiomyopathy (DCM) is characterized by congestive heart failure and altered myocardial gene expression. Epigenetic changes, including DNA methylation, are implicated in the development of DCM but have not been studied extensively. Clinical human DCM and nonfailing control left ventricle samples were individually analyzed for DNA methylation and expressional changes. Expression microarrays were used to identify 393 overexpressed and 349 underexpressed genes in DCM (GEO accession number: GSE43435). Gene promoter microarrays were utilized for DNA methylation analysis, and the resulting data were analyzed by two different computational methods. In the first method, we utilized subtractive analysis of DNA methylation peak data to identify 158 gene promoters exhibiting DNA methylation changes that correlated with expression changes. In the second method, a two-stage approach combined a particle swarm optimization feature selection algorithm and a discriminant analysis via mixed integer programming classifier to identify differentially methylated gene promoters. This analysis identified 51 hypermethylated promoters and six hypomethylated promoters in DCM with 100% cross-validation accuracy in the group assignment. Generation of a composite list of genes identified by subtractive analysis and two-stage computation analysis revealed four genes that exhibited differential DNA methylation by both methods in addition to altered gene expression. Computationally identified genes (AURKB, BTNL9, CLDN5, and TK1) define a central set of differentially methylated gene promoters that are important in classifying DCM. These genes have no previously reported role in DCM. This study documents that rigorous computational analysis applied to microarray analysis of healthy and diseased human heart samples helps to define clinically relevant DNA methylation and expressional changes in DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Metilación de ADN/genética , Perfilación de la Expresión Génica/métodos , Miocardio/metabolismo , Aurora Quinasa B/genética , Butirofilinas , Claudina-5/genética , Biología Computacional , Ventrículos Cardíacos/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Timidina Quinasa/genética
8.
Physiol Genomics ; 45(14): 590-6, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695887

RESUMEN

This study addresses how depletion of human cardiac left ventricle (LV) mitochondrial DNA (mtDNA) and epigenetic nuclear DNA methylation promote cardiac dysfunction in human dilated cardiomyopathy (DCM) through regulation of pyrimidine nucleotide kinases. Samples of DCM LV and right ventricle (n = 18) were obtained fresh at heart transplant surgery. Parallel samples from nonfailing (NF) controls (n = 12) were from donor hearts found unsuitable for clinical use. We analyzed abundance of mtDNA and nuclear DNA (nDNA) using qPCR. LV mtDNA was depleted in DCM (50%, P < 0.05 each) compared with NF. No detectable change in RV mtDNA abundance occurred. DNA methylation and gene expression were determined using microarray analysis (GEO accession number: GSE43435). Fifty-seven gene promoters exhibited DNA hypermethylation or hypomethylation in DCM LVs. Among those, cytosolic thymidine kinase 1 (TK1) was hypermethylated. Expression arrays revealed decreased abundance of the TK1 mRNA transcript with no change in transcripts for other relevant thymidine metabolism enzymes. Quantitative immunoblots confirmed decreased TK1 polypeptide steady state abundance. TK1 activity remained unchanged in DCM samples while mitochondrial thymidine kinase (TK2) activity was significantly reduced. Compensatory TK activity was found in cardiac myocytes in the DCM LV. Diminished TK2 activity is mechanistically important to reduced mtDNA abundance and identified in DCM LV samples here. Epigenetic and genetic changes result in changes in mtDNA and in nucleotide substrates for mtDNA replication and underpin energy starvation in DCM.


Asunto(s)
Cardiomiopatías/genética , ADN Mitocondrial/genética , Epigénesis Genética/genética , Timidina Quinasa/genética , Western Blotting , Metilación de ADN/genética , Humanos , Técnicas In Vitro , Persona de Mediana Edad
9.
J Card Fail ; 19(4): 283-94, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23582094

RESUMEN

BACKGROUND: Heart failure (HF) is associated with excessive extracellular matrix (ECM) deposition and abnormal ECM degradation leading to cardiac fibrosis. Connective tissue growth factor (CTGF) modulates ECM production during inflammatory tissue injury, but available data on CTGF gene expression in failing human heart and its response to mechanical unloading are limited. METHODS AND RESULTS: Left ventricle (LV) tissue from patients undergoing cardiac transplantation for ischemic (ICM; n = 20) and dilated (DCM; n = 20) cardiomyopathies and from nonfailing (NF; n = 20) donor hearts were examined. Paired samples (n = 15) from patients undergoing LV assist device (LVAD) implantation as "bridge to transplant" (34-1,145 days) also were analyzed. There was more interstitial fibrosis in both ICM and DCM compared with NF hearts. Hydroxyproline concentration was also significantly increased in DCM compared with NF samples. The expression of CTGF, transforming growth factor (TGF) ß1, collagen (COL) 1-α1, COL3-α1, matrix metalloproteinase (MMP) 2, and MMP9 mRNA in ICM and DCM were also significantly elevated compared with NF samples. Although TGF-ß1, CTGF, COL1-α1, and COL3-α1 mRNA levels were reduced by unloading, there was only a modest reduction in tissue fibrosis and no difference in protein-bound hydroxyproline concentration between pre- and post-LVAD tissue samples. The persistent fibrosis may be related to a concomitant reduction in MMP9 mRNA and protein levels following unloading. CONCLUSIONS: CTGF may be a key regulator of fibrosis during maladaptive remodeling and progression to HF. Although mechanical unloading normalizes most genotypic and functional abnormalities, its effect on ECM remodeling during HF is incomplete.


Asunto(s)
Cardiomiopatía Dilatada/patología , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Trasplante de Corazón , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Dilatada/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibrosis , Insuficiencia Cardíaca/diagnóstico , Trasplante de Corazón/tendencias , Humanos , Remodelación Ventricular/genética
11.
J Biol Chem ; 288(6): 4252-64, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23266827

RESUMEN

Integrins are adhesive, signaling, and mechanotransduction proteins. Talin (Tln) activates integrins and links it to the actin cytoskeleton. Vertebrates contain two talin genes, tln1 and tln2. How Tln1 and Tln2 function in cardiac myocytes (CMs) is unknown. Tln1 and Tln2 expression were evaluated in the normal embryonic and adult mouse heart as well as in control and failing human adult myocardium. Tln1 function was then tested in the basal and mechanically stressed myocardium after cardiomyocyte-specific excision of the Tln1 gene. During embryogenesis, both Tln forms are highly expressed in CMs, but in the mature heart Tln2 becomes the main Tln isoform, localizing to the costameres. Tln1 expression is minimal in the adult CM. With pharmacological and mechanical stress causing hypertrophy, Tln1 is up-regulated in CMs and is specifically detected at costameres, suggesting its importance in the compensatory response to CM stress. In human failing heart, CM Tln1 also increases compared with control samples from normal functioning myocardium. To directly test Tln1 function in CMs, we generated CM-specific Tln1 knock-out mice (Tln1cKO). Tln1cKO mice showed normal basal cardiac structure and function but when subjected to pressure overload showed blunted hypertrophy, less fibrosis, and improved cardiac function versus controls. Acute responses of ERK1/2, p38, Akt, and glycogen synthase kinase 3 after mechanical stress were strongly blunted in Tln1cKO mice. Given these results, we conclude that Tln1 and Tln2 have distinct functions in the myocardium. Our data show that reduction of CM Tln1 expression can lead to improved cardiac remodeling following pressure overload.


Asunto(s)
Cardiomegalia/metabolismo , Miocardio/metabolismo , Talina/biosíntesis , Adulto , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Femenino , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estrés Fisiológico/genética , Talina/genética , Regulación hacia Arriba/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Biomech Model Mechanobiol ; 12(1): 95-109, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22407215

RESUMEN

Cardiac muscle cells are known to adapt to their physical surroundings, optimizing intracellular organization and contractile function for a given culture environment. A previously developed in vitro model system has shown that the inclusion of discrete microscale domains (or microrods) in three dimensions (3D) can alter long-term growth responses of neonatal ventricular myocytes. The aim of this work was to understand how cellular contact with such a domain affects various mechanical changes involved in cardiac muscle cell remodeling. Myocytes were maintained in 3D gels over 5 days in the presence or absence of 100-µm-long microrods, and the effect of this local heterogeneity on cell behavior was analyzed via several imaging techniques. Microrod abutment resulted in approximately twofold increases in the maximum displacement of spontaneously beating myocytes, as based on confocal microscopy scans of the gel xy-plane or the myocyte long axis. In addition, microrods caused significant increases in the proportion of aligned myofibrils (≤20° deviation from long axis) in fixed myocytes. Microrod-related differences in axial contraction could be abrogated by long-term interruption of certain signals of the RhoA-/Rho-associated kinase (ROCK) or protein kinase C (PKC) pathway. Furthermore, microrod-induced increases in myocyte size and protein content were prevented by ROCK inhibition. In all, the data suggest that microdomain heterogeneity in 3D appears to promote the development of axially aligned contractile machinery in muscle cells, an observation that may have relevance to a number of cardiac tissue engineering interventions.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Mecanotransducción Celular/fisiología , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Ingeniería de Tejidos/métodos , Proteína de Unión al GTP rhoA/metabolismo , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula/instrumentación , Células Cultivadas , Ratas , Ratas Sprague-Dawley , Ingeniería de Tejidos/instrumentación
13.
Biochem J ; 447(2): 193-204, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22849349

RESUMEN

Vascular endothelial injury predisposes to endothelial dysfunction and atherogenesis. We have investigated the hypothesis that PKCε (protein kinase Cε) is an important upstream regulator of cytoprotective pathways in vascular ECs (endothelial cells). Depletion of PKCε in human ECs reduced expression of the cytoprotective genes A1, A20 and Bcl-2. Conversely, constitutively active PKCε expressed in human ECs increased mRNA and protein levels of these cytoprotective genes, with up-regulation dependent upon ERK1/2 (extracellular-signal-regulated kinase 1/2) activation. Furthermore, inhibition of NF-κB (nuclear factor κB) by the pharmacological antagonist BAY 11-7085 or an IκB (inhibitor of NF-κB) SuperRepressor prevented cytoprotective gene induction. Activation of PKCε enhanced p65 NF-κB DNA binding and elevated NF-κB transcriptional activity. Importantly, although NF-κB activation by PKCε induced cytoprotective genes, it did not up-regulate pro-inflammatory NF-κB targets [E-selectin, VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1)]. Indeed, PKCε exhibited cytoprotective and anti-inflammatory actions, including inhibition of TNFα (tumour necrosis factor α)-induced JNK (c-Jun N-terminal kinase) phosphorylation and ICAM-1 up-regulation, a response attenuated by depletion of A20. Thus we conclude that PKCε plays an essential role in endothelial homoeostasis, acting as an upstream co-ordinator of gene expression through activation of ERK1/2, inhibition of JNK and diversion of the NF-κB pathway to cytoprotective gene induction, and propose that PKCε represents a novel therapeutic target for endothelial dysfunction.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Proteínas de Unión al ADN/biosíntesis , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Antígenos de Histocompatibilidad Menor , Proteínas Nucleares/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Molécula 1 de Adhesión Celular Vascular/biosíntesis
14.
J Signal Transduct ; 2012: 473410, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22900166

RESUMEN

Intracellular nitric oxide (NO(i)) is a physiological regulator of excitation-contraction coupling, but is also involved in the development of cardiac dysfunction during hypertrophy and heart failure. To determine whether contractile activity regulates nitric oxide synthase (NOS) expression, spontaneously contracting, neonatal rat ventricular myocytes (NRVM) were treat with L-type calcium channel blockers (nifedipine and verapamil) or myosin II ATPase inhibitors (butanedione monoxime (BDM) and blebbistatin) to produce contractile arrest. Both types of inhibitors significantly reduced iNOS but not eNOS expression, and also reduced NO(i) production. Inhibiting contractile activity also reduced focal adhesion kinase (FAK) and AKT phosphorylation. Contraction-induced iNOS expression required FAK and phosphatidylinositol 3-kinase (PI(3)K), as both PF573228 and LY294002 (10 µM, 24 h) eliminated contraction-induced iNOS expression. Similarly, shRNAs specific for FAK (shFAK) caused FAK knockdown, reduced AKT phosphorylation at T308 and S473, and reduced iNOS expression. In contrast, shRNA-mediated knockdown of PYK2, the other member of the FAK-family of protein tyrosine kinases, had much less of an effect. Conversely, overexpression of a constitutively active form of FAK (CD2-FAK) or AKT (Myr-AKT) reversed the inhibitory effect of BDM on iNOS expression and NO(i) production. Thus, contraction-induced iNOS expression and NO(i) production in NRVM are mediated via a FAK-PI(3)K-AKT signaling pathway.

15.
Cardiovasc Res ; 92(3): 409-19, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21937583

RESUMEN

AIMS: Tyrosine-phosphorylated focal adhesion kinase (FAK) is required for the hypertrophic response of cardiomyocytes to growth factors and mechanical load, but the role of FAK serine phosphorylation in this process is unknown. The aims of the present study were to characterize FAK serine phosphorylation in cultured neonatal rat ventricular myocytes (NRVM), analyse its functional significance during hypertrophic signalling, and examine its potential role in the pathogenesis of human dilated cardiomyopathy (DCM). METHODS AND RESULTS: Endothelin-1 (ET-1) and other hypertrophic factors induced a time- and dose-dependent increase in FAK-S910 phosphorylation. ET-1-induced FAK-S910 phosphorylation required ET(A)R-dependent activation of PKCδ and Src via parallel Raf-1 → MEK1/2 → ERK1/2 and MEK5 → ERK5 signalling pathways. Replication-deficient adenoviruses expressing wild-type (WT) FAK and a non-phosphorylatable, S910A-FAK mutant were then used to examine the functional significance of FAK-S910 phosphorylation. Unlike WT-FAK, S910A-FAK increased the half-life of GFP-tagged paxillin within costameres (as determined by total internal reflection fluorescence microscopy and fluorescence recovery after photobleaching) and increased the steady-state FAK-paxillin interaction (as determined by co-immunoprecipitation and western blotting). These alterations resulted in reduced NRVM sarcomere reorganization and cell spreading. Finally, we found that FAK was serine-phosphorylated at multiple sites in non-failing, human left ventricular tissue. FAK-S910 phosphorylation and ERK5 expression were dramatically reduced in patients undergoing heart transplantation for end-stage DCM. CONCLUSION: FAK undergoes S910 phosphorylation via PKCδ and Src-dependent pathways that are important for cell spreading and sarcomere reorganization. Reduced FAK-S910 phosphorylation may contribute to sarcomere disorganization in DCM.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Insuficiencia Cardíaca/enzimología , Miocitos Cardíacos/enzimología , Sarcómeros/enzimología , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Cardiomiopatía Dilatada/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelina-1/farmacología , Activación Enzimática , Recuperación de Fluorescencia tras Fotoblanqueo , Quinasa 1 de Adhesión Focal/genética , Insuficiencia Cardíaca/patología , Humanos , Inmunoprecipitación , Factor I del Crecimiento Similar a la Insulina/farmacología , Microscopía Fluorescente , Mutación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Paxillin/genética , Paxillin/metabolismo , Fenilefrina/farmacología , Fosforilación , Proteína Quinasa C-delta/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Sarcómeros/efectos de los fármacos , Sarcómeros/patología , Serina , Transducción de Señal , Factores de Tiempo , Transfección , Familia-src Quinasas/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 31(11): 2432-40, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21852560

RESUMEN

OBJECTIVE: Focal adhesion kinase-related nonkinase (FRNK), the C-terminal domain of focal adhesion kinase (FAK), is a tyrosine-phosphorylated, vascular smooth muscle cell (VSMC)-specific inhibitor of cell migration. FRNK inhibits both FAK and proline-rich tyrosine kinase 2 (PYK2) in cultured VSMCs, and both kinases may be involved in VSMC invasion during vascular remodeling. METHODS AND RESULTS: Adenovirally mediated gene transfer of green fluorescent protein-tagged, wild-type (wt) FRNK into balloon-injured rat carotid arteries confirmed that FRNK overexpression inhibited both FAK and PYK2 phosphorylation and downstream signaling in vivo. To identify which kinase was involved in regulating VSMC invasion, adenovirally mediated expression of specific short hairpin RNAs was used to knock down FAK versus PYK2 in cultured VSMCs, but only FAK short hairpin RNA was effective in reducing VSMC invasion. The role of FRNK tyrosine phosphorylation was then examined using adenoviruses expressing nonphosphorylatable (Tyr168Phe-, Tyr232Phe-, and Tyr168,232Phe-) green fluorescent protein-FRNK mutants. wtFRNK and all FRNK mutants localized to FAs, but only Tyr168 phosphorylation was required for FRNK to inhibit invasion. Preventing Tyr168 phosphorylation also increased FRNK-paxillin interaction, as determined by coimmunoprecipitation, total internal reflection fluorescence microscopy, and fluorescence recovery after photobleaching. Furthermore, wtFRNK competed with FAK for binding to p130(Cas) (a critically important regulator of cell migration) and prevented its phosphorylation. However, Tyr168Phe-FRNK was unable to bind p130(Cas). CONCLUSION: We propose a 3-stage mechanism for FRNK inhibition: focal adhesion targeting, Tyr168 phosphorylation, and competition with FAK for p130 binding and phosphorylation, which are all required for FRNK to inhibit VSMC invasion.


Asunto(s)
Movimiento Celular/fisiología , Proteína Sustrato Asociada a CrK/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Proteínas Tirosina Quinasas/metabolismo , Adenoviridae/genética , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Cateterismo/efectos adversos , Células Cultivadas , Quinasa 2 de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Modelos Animales , Fosforilación/fisiología , Unión Proteica/fisiología , ARN Interferente Pequeño/farmacología , Ratas
17.
Am J Physiol Renal Physiol ; 301(1): F197-208, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21289057

RESUMEN

PKC-ε activation mediates protection from ischemia-reperfusion injury in the myocardium. Mitochondria are a subcellular target of these protective mechanisms of PKC-ε. Previously, we have shown that PKC-ε activation is involved in mitochondrial dysfunction in oxidant-injured renal proximal tubular cells (RPTC; Nowak G, Bakajsova D, Clifton GL Am J Physiol Renal Physiol 286: F307-F316, 2004). The goal of this study was to examine the role of PKC-ε activation in mitochondrial dysfunction and to identify mitochondrial targets of PKC-ε in RPTC. The constitutively active and inactive mutants of PKC-ε were overexpressed in primary cultures of RPTC using the adenoviral technique. Increases in active PKC-ε levels were accompanied by PKC-ε translocation to mitochondria. Sustained PKC-ε activation resulted in decreases in state 3 respiration, electron transport rate, ATP production, ATP content, and activities of complexes I and IV and F(0)F(1)-ATPase. Furthermore, PKC-ε activation increased mitochondrial membrane potential and oxidant production and induced mitochondrial fragmentation and RPTC death. Accumulation of the dynamin-related protein in mitochondria preceded mitochondrial fragmentation. Antioxidants blocked PKC-ε-induced increases in the oxidant production but did not prevent mitochondrial fragmentation and cell death. The inactive PKC-ε mutant had no effect on mitochondrial functions, morphology, oxidant production, and RPTC viability. We conclude that active PKC-ε targets complexes I and IV and F(0)F(1)-ATPase in RPTC. PKC-ε activation mediates mitochondrial dysfunction, hyperpolarization, and fragmentation. It also induces oxidant generation and cell death, but oxidative stress is not the mechanism of RPTC death. These results show that in contrast to protective effects of PKC-ε activation in cardiomyocytes, sustained PKC-ε activation is detrimental to mitochondrial function and viability in RPTC.


Asunto(s)
Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/fisiopatología , Mitocondrias/fisiología , Proteína Quinasa C-epsilon/metabolismo , Adenosina Trifosfato/biosíntesis , Adenoviridae/genética , Animales , Muerte Celular/efectos de los fármacos , Separación Celular , Supervivencia Celular , Células Cultivadas , Activación Enzimática/fisiología , Femenino , Túbulos Renales Proximales/citología , Cinética , L-Lactato Deshidrogenasa/metabolismo , Potenciales de la Membrana/fisiología , Mitocondrias/ultraestructura , Membranas Mitocondriales/fisiología , Consumo de Oxígeno/fisiología , Proteína Quinasa C-epsilon/biosíntesis , Proteína Quinasa C-epsilon/genética , ATPasas de Translocación de Protón/metabolismo , Conejos , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transfección/métodos
18.
Am J Physiol Heart Circ Physiol ; 300(2): H617-26, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21131481

RESUMEN

Endothelin-1 (ET-1) and activation of protein kinase C (PKC) have been implicated in alterations of myocyte function in cardiac hypertrophy and heart failure. Changes in cellular Ca2+ handling and electrophysiological properties also occur in these states and may contribute to mechanical dysfunction and arrhythmias. While ET-1 or PKC stimulation induces cellular hypertrophy in cultured neonatal rat ventricular myocytes (NRVMs), a system widely used in studies of hypertrophic signaling, there is little data about electrophysiological changes. Here we studied the effects of ET-1 (100 nM) or the PKC activator phorbol 12-myristate 13-acetate (PMA, 1 µM) on ionic currents in NRVMs. The acute effects of PMA or ET-1 (≤30 min) were small or insignificant. However, PMA or ET-1 exposure for 48-72 h increased cell capacitance by 100 or 25%, respectively, indicating cellular hypertrophy. ET-1 also slightly increased Ca2+ current density (T and L type). Na+/Ca2+ exchange current was increased by chronic pretreatment with either PMA or ET-1. In contrast, transient outward and delayed rectifier K+ currents were strongly downregulated by PMA or ET-1 pretreatment. Inward rectifier K+ current tended toward a decrease at larger negative potential, but time-independent outward K+ current was unaltered by either treatment. The enhanced inward and reduced outward currents also result in action potential prolongation after PMA or ET-1 pretreatment. We conclude that chronic PMA or ET-1 exposure in cultured NRVMs causes altered functional expression of cardiac ion currents, which mimic electrophysiological changes seen in whole animal and human hypertrophy and heart failure.


Asunto(s)
Canales de Calcio/biosíntesis , Endotelina-1/farmacología , Miocitos Cardíacos/metabolismo , Canales de Potasio/biosíntesis , Intercambiador de Sodio-Calcio/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Western Blotting , Canales de Calcio/efectos de los fármacos , Tamaño de la Célula , Células Cultivadas , Fenómenos Electrofisiológicos , Técnicas In Vitro , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Fosforilación , Canales de Potasio/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Arterioscler Thromb Vasc Biol ; 30(11): 2226-33, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20705914

RESUMEN

OBJECTIVE: To examine whether interference with FRNK targeting to focal adhesions (FAs) affects its inhibitory activity and tyrosine phosphorylation. METHODS AND RESULTS: Focal adhesion kinase and its autonomously expressed C-terminal inhibitor, focal adhesion kinase-related nonkinase (FRNK), regulate vascular smooth muscle cell (VSMC) signaling and migration. FRNK-paxillin binding was reduced by a point mutation in its FA targeting domain (L341S-FRNK). Green fluorescent protein-tagged wild type and L341S-FRNK were then adenovirally expressed in VSMCs. L341S-FRNK targeted to VSMC FAs, despite previous studies in other cell types. L341S-FRNK affected FA binding kinetics (assessed by total internal reflection fluorescnece [TIRF] microscopy and fluorescence recovery after photobleaching [FRAP]) and reduced its steady-state paxillin interaction (determined by coimmunoprecipitation). Both wt-FRNK and L341S-FRNK lowered basal and angiotensin II-stimulated focal adhesion kinase, paxillin, and extracellular signal-regulated kinase 1/2 phosphorylation. However, the degree of inhibition was significantly reduced by L341S-FRNK. L341S-FRNK also demonstrated significantly greater migratory activity compared with wt-FRNK-expressing VSMCs. Angiotensin II-induced Y168 phosphorylation was Src dependent, as evident by a significant reduction in Y168 phosphorylation by the Src family kinase inhibitor PP2 is 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). Surprisingly, Y168 phosphorylation was unaffected by its targeting. Furthermore, Y232 phosphorylation increased approximately 3-fold in L341S-FRNK, which was less sensitive to PP2. CONCLUSIONS: FRNK inhibition of VSMC migration requires both FA targeting and Y168 phosphorylation by Src family kinases. FRNK-Y232 phosphorylation occurs outside of FAs, probably by a PP2-insensitive kinase.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Miocitos del Músculo Liso/fisiología , Paxillin/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Movimiento Celular/fisiología , Músculo Liso Vascular , Fosforilación , Mutación Puntual , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/fisiología , Ratas , Transducción de Señal
20.
J Mol Cell Cardiol ; 48(5): 817-23, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20188736

RESUMEN

A ventricular myocyte experiences changes in length and load during every beat of the heart and has the ability to remodel cell shape to maintain cardiac performance. Specifically, myocytes elongate in response to increased diastolic strain by adding sarcomeres in series, and they thicken in response to continued systolic stress by adding filaments in parallel. Myocytes do this while still keeping the resting sarcomere length close to its optimal value at the peak of the length-tension curve. This review focuses on the little understood mechanisms by which direction of growth is matched in a physiologically appropriate direction. We propose that the direction of strain is detected by differential phosphorylation of proteins in the costamere, which then transmit signaling to the Z-disc for parallel or series addition of thin filaments regulated via the actin capping processes. In this review, we link mechanotransduction to the molecular mechanisms for regulation of myocyte length and width.


Asunto(s)
Miocardio/metabolismo , Sarcómeros/metabolismo , Sarcómeros/fisiología , Estrés Mecánico , Animales , Humanos , Modelos Biológicos , Desarrollo de Músculos/fisiología , Miocardio/enzimología , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...