Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
2.
Int J Biochem Cell Biol ; 120: 105668, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31877385

RESUMEN

ETS domain-containing protein-1 (ELK1) is a transcription factor important in regulating αvß6 integrin expression. αvß6 integrins activate the profibrotic cytokine Transforming Growth Factor ß1 (TGFß1) and are increased in the alveolar epithelium in idiopathic pulmonary fibrosis (IPF). IPF is a disease associated with aging and therefore we hypothesised that aged animals lacking Elk1 globally would develop spontaneous fibrosis in organs where αvß6 mediated TGFß activation has been implicated. Here we identify that Elk1-knockout (Elk1-/0) mice aged to one year developed spontaneous fibrosis in the absence of injury in both the lung and the liver but not in the heart or kidneys. The lungs of Elk1-/0 aged mice demonstrated increased collagen deposition, in particular collagen 3α1, located in small fibrotic foci and thickened alveolar walls. Despite the liver having relatively low global levels of ELK1 expression, Elk1-/0 animals developed hepatosteatosis and fibrosis. The loss of Elk1 also had differential effects on Itgb1, Itgb5 and Itgb6 expression in the four organs potentially explaining the phenotypic differences in these organs. To understand the potential causes of reduced ELK1 in human disease we exposed human lung epithelial cells and murine lung slices to cigarette smoke extract, which lead to reduced ELK1 expression andmay explain the loss of ELK1 in human disease. These data support a fundamental role for ELK1 in protecting against the development of progressive fibrosis via transcriptional regulation of beta integrin subunit genes, and demonstrate that loss of ELK1 can be caused by cigarette smoke.


Asunto(s)
Bronquios/patología , Pulmón/patología , Proteína Elk-1 con Dominio ets/deficiencia , Factores de Edad , Animales , Bronquios/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Elk-1 con Dominio ets/metabolismo
3.
Nucleic Acids Res ; 47(21): e138, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31529053

RESUMEN

To understand the molecular pathogenesis of human disease, precision analyses to define alterations within and between disease-associated cell populations are desperately needed. Single-cell genomics represents an ideal platform to enable the identification and comparison of normal and diseased transcriptional cell populations. We created cellHarmony, an integrated solution for the unsupervised analysis, classification, and comparison of cell types from diverse single-cell RNA-Seq datasets. cellHarmony efficiently and accurately matches single-cell transcriptomes using a community-clustering and alignment strategy to compute differences in cell-type specific gene expression over potentially dozens of cell populations. Such transcriptional differences are used to automatically identify distinct and shared gene programs among cell-types and identify impacted pathways and transcriptional regulatory networks to understand the impact of perturbations at a systems level. cellHarmony is implemented as a python package and as an integrated workflow within the software AltAnalyze. We demonstrate that cellHarmony has improved or equivalent performance to alternative label projection methods, is able to identify the likely cellular origins of malignant states, stratify patients into clinical disease subtypes from identified gene programs, resolve discrete disease networks impacting specific cell-types, and illuminate therapeutic mechanisms. Thus, this approach holds tremendous promise in revealing the molecular and cellular origins of complex disease.


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Bases de Datos Genéticas , Conjuntos de Datos como Asunto , Humanos
4.
JACC Basic Transl Sci ; 4(2): 188-199, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31061921

RESUMEN

Cardiomyocyte-specific increases in phosphorylated Hsp20 (S16D-Hsp20) to levels similar to those observed in human failing hearts are associated with early fibrotic remodeling and depressed left ventricular function, symptoms which progress to heart failure and early death. The underlying mechanisms appear to involve translocation of phosphorylated Hsp20 to the nucleus and upregulation of interleukin (IL)-6, which subsequently activates cardiac fibroblasts in a paracrine fashion through transcription factor STAT3 signaling. Accordingly, treatment of S16D-Hsp20 mice with a rat anti-mouse IL-6 receptor monoclonal antibody (MR16-1) attenuated interstitial fibrosis and preserved cardiac function. These findings suggest that phosphorylated Hsp20 may be a potential therapeutic target in heart failure.

5.
JCI Insight ; 4(4)2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30668549

RESUMEN

RNA binding proteins represent an emerging class of proteins with a role in cardiac dysfunction. We show that activation of the RNA binding protein human antigen R (HuR) is increased in the failing human heart. To determine the functional role of HuR in pathological cardiac hypertrophy, we created an inducible cardiomyocyte-specific HuR-deletion mouse and showed that HuR deletion reduces left ventricular hypertrophy, dilation, and fibrosis while preserving cardiac function in a transverse aortic constriction (TAC) model of pressure overload-induced hypertrophy. Assessment of HuR-dependent changes in global gene expression suggests that the mechanistic basis for this protection occurs through a reduction in fibrotic signaling, specifically through a reduction in TGF-ß (Tgfb) expression. Finally, pharmacological inhibition of HuR at a clinically relevant time point following the initial development of pathological hypertrophy after TAC also yielded a significant reduction in pathological progression, as marked by a reduction in hypertrophy, dilation, and fibrosis and preserved function. In summary, this study demonstrates a functional role for HuR in the progression of pressure overload-induced cardiac hypertrophy and establishes HuR inhibition as a viable therapeutic approach for pathological cardiac hypertrophy and heart failure.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Insuficiencia Cardíaca/patología , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Miocardio/patología , Animales , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Modelos Animales de Enfermedad , Proteína 1 Similar a ELAV/antagonistas & inhibidores , Proteína 1 Similar a ELAV/genética , Fibrosis , Insuficiencia Cardíaca/tratamiento farmacológico , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , RNA-Seq , Remodelación Ventricular/efectos de los fármacos
6.
Pediatr Radiol ; 49(3): 332-341, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30607435

RESUMEN

BACKGROUND: Primary sclerosing cholangitis, autoimmune hepatitis and autoimmune sclerosing cholangitis are forms of chronic, progressive autoimmune liver disease (AILD) that can affect the pediatric population. OBJECTIVE: To determine whether quantitative MRI- and laboratory-based biomarkers are associated with conventional imaging findings of portal hypertension (radiologic portal hypertension) in children and young adults with AILD. MATERIALS AND METHODS: Forty-four patients with AILD enrolled in an institutional registry underwent a research abdominal MRI examination at 1.5 tesla (T). Five quantitative MRI techniques were performed: liver MR elastography, spleen MR elastography, liver iron-corrected T1 mapping, liver T2 mapping, and liver diffusion-weighted imaging (DWI, quantified as apparent diffusion coefficients). Two anatomical sequences were used to document splenomegaly, varices and ascites. We calculated aspartate aminotransferase (AST)-to-platelet ratio index (APRI) and fibrosis-4 (FIB-4) scores - laboratory-based biomarkers of liver fibrosis. We used receiver operating characteristic (ROC) curve analyses to establish the diagnostic performance of quantitative MRI and laboratory biomarkers for indicating the presence of radiologic portal hypertension. RESULTS: Twenty-three (52%) patients were male; mean age was 15.2±4.0 years. Thirteen (30%) patients had radiologic portal hypertension. Liver and spleen stiffness demonstrated the greatest diagnostic performance for indicating the presence of portal hypertension (area-under-the-ROC-curve [AUROC]=0.98 and 0.96, respectively). The APRI and FIB-4 scores also demonstrated good diagnostic performance (AUROC=0.87 and 0.88, respectively). CONCLUSION: MRI-derived measures of liver and spleen stiffness as well as laboratory-based APRI and FIB-4 scores are highly associated with imaging findings of portal hypertension in children and young adults with AILD and thus might be useful for predicting portal hypertension impending onset and directing personalized patient management.


Asunto(s)
Colangitis Esclerosante/diagnóstico por imagen , Hepatitis Autoinmune/diagnóstico por imagen , Hipertensión Portal/diagnóstico por imagen , Hipertensión Portal/inmunología , Imagen por Resonancia Magnética/métodos , Adolescente , Aspartato Aminotransferasas/análisis , Biomarcadores/análisis , Niño , Estudios Transversales , Imagen de Difusión por Resonancia Magnética , Diagnóstico por Imagen de Elasticidad , Femenino , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Valor Predictivo de las Pruebas , Sistema de Registros , Adulto Joven
7.
Circulation ; 138(12): 1236-1252, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-29653926

RESUMEN

BACKGROUND: Fibronectin (FN) polymerization is necessary for collagen matrix deposition and is a key contributor to increased abundance of cardiac myofibroblasts (MFs) after cardiac injury. We hypothesized that interfering with FN polymerization or its genetic ablation in fibroblasts would attenuate MF and fibrosis and improve cardiac function after ischemia/reperfusion (I/R) injury. METHODS: Mouse and human MFs were used to assess the impact of the FN polymerization inhibitor (pUR4) in attenuating pathological cellular features such as proliferation, migration, extracellular matrix deposition, and associated mechanisms. To evaluate the therapeutic potential of inhibiting FN polymerization in vivo, wild-type mice received daily intraperitoneal injections of either pUR4 or control peptide (III-11C) immediately after cardiac surgery for 7 consecutive days. Mice were analyzed 7 days after I/R to assess MF markers and inflammatory cell infiltration or 4 weeks after I/R to evaluate long-term effects of FN inhibition on cardiac function and fibrosis. Furthermore, inducible, fibroblast-restricted, FN gene-ablated (Tcf21MerCreMer; Fnflox) mice were used to evaluate cell specificity of FN expression and polymerization in the heart. RESULTS: pUR4 administration on activated MFs reduced FN and collagen deposition into the extracellular matrix and attenuated cell proliferation, likely mediated through decreased c-myc signaling. pUR4 also ameliorated fibroblast migration accompanied by increased ß1 integrin internalization and reduced levels of phosphorylated focal adhesion kinase protein. In vivo, daily administration of pUR4 for 7 days after I/R significantly reduced MF markers and neutrophil infiltration. This treatment regimen also significantly attenuated myocardial dysfunction, pathological cardiac remodeling, and fibrosis up to 4 weeks after I/R. Last, inducible ablation of FN in fibroblasts after I/R resulted in significant functional cardioprotection with reduced hypertrophy and fibrosis. The addition of pUR4 to the FN-ablated mice did not confer further cardioprotection, suggesting that the salutary effects of inhibiting FN polymerization may be mediated largely through effects on FN secreted from the cardiac fibroblast lineage. CONCLUSIONS: Inhibiting FN polymerization or cardiac fibroblast gene expression attenuates pathological properties of MFs in vitro and ameliorates adverse cardiac remodeling and fibrosis in an in vivo model of heart failure. Interfering with FN polymerization may be a new therapeutic strategy for treating cardiac fibrosis and heart failure.


Asunto(s)
Fibronectinas/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miofibroblastos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Quinasa 1 de Adhesión Focal/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Integrina beta1/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Infiltración Neutrófila/efectos de los fármacos , Fosforilación , Polimerizacion , Transducción de Señal/efectos de los fármacos
8.
Heart Lung Circ ; 25(5): 425-34, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26993094

RESUMEN

Heart failure is a significant global health problem, which is becoming worse as the population ages, and remains one of the biggest burdens on our economy. Despite significant advances in cardiovascular medicine, management and surgery, mortality rates remain high, with almost half of patients with heart failure dying within five years of diagnosis. As a multifactorial clinical syndrome, heart failure still represents an epidemic threat, highlighting the need for deeper insights into disease mechanisms and the development of innovative therapeutic strategies for both treatment and prevention. In this review, we discuss conventional heart failure therapies and highlight new pharmacological agents targeting pathophysiological features of the failing heart, for example, non-coding RNAs, angiotensin receptor-neprilysin inhibitors, cardiac myosin activators, BGP-15 and molecules targeting GRK2 including M119, gallein and paroxetine. Finally, we address the disparity between phase II and phase III clinical trials that prevent the translation of emerging HF therapies into new and approved therapies.


Asunto(s)
Antagonistas de Receptores de Angiotensina/uso terapéutico , Ciclohexanos/uso terapéutico , Insuficiencia Cardíaca/terapia , Oximas/uso terapéutico , Paroxetina/uso terapéutico , Piperidinas/uso terapéutico , Xantenos/uso terapéutico , Miosinas Cardíacas/metabolismo , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/fisiopatología , Humanos , Neprilisina/antagonistas & inhibidores , Neprilisina/metabolismo , ARN no Traducido/metabolismo
9.
J Mol Cell Cardiol ; 91: 228-37, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26778458

RESUMEN

The extracellular matrix (ECM) is a complex and dynamic scaffold that maintains tissue structure and dynamics. However, the view of the ECM as an inert architectural support has been increasingly challenged. The ECM is a vibrant meshwork, a crucial organizer of cellular microenvironments. It plays a direct role in cellular interactions regulating cell growth, survival, spreading, proliferation, differentiation and migration through the intricate relationship among cellular and acellular tissue components. This complex interrelationship preserves cardiac function during homeostasis; however it is also responsible for pathologic remodeling following myocardial injury. Therefore, enhancing our understanding of this cross-talk may provide mechanistic insights into the pathogenesis of heart failure and suggest new approaches to novel, targeted pharmacologic therapies. This review explores the implications of ECM-cell interactions in myocardial cell behavior and cardiac function at baseline and following myocardial injury.


Asunto(s)
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Insuficiencia Cardíaca/genética , Lesiones Cardíacas/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Comunicación Celular , Colágeno/genética , Colágeno/metabolismo , Citoesqueleto/química , Citoesqueleto/metabolismo , Matriz Extracelular/química , Fibroblastos/patología , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Miocardio/patología , Miocitos Cardíacos/patología , Osteonectina/genética , Osteonectina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Transducción de Señal , Tenascina/genética , Tenascina/metabolismo , Trombospondinas/genética , Trombospondinas/metabolismo
10.
J Am Coll Cardiol ; 63(23): 2549-2557, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24703913

RESUMEN

OBJECTIVES: The authors propose simultaneous inhibition of Gßγ signaling in the heart and the adrenal gland as a novel therapeutic approach for heart failure (HF). BACKGROUND: Elevated sympathetic nervous system activity is a salient characteristic of HF progression. It causes pathologic desensitization of ß-adrenergic receptors (ß-AR), facilitated predominantly through Gßγ-mediated signaling. The adrenal glands are key contributors to the chronically elevated plasma catecholamine levels observed in HF, where adrenal α2-AR feedback inhibitory function is impaired also through Gßγ-mediated signaling. METHODS: We investigated the efficacy of a small molecule Gßγ inhibitor, gallein, in a clinically relevant, pressure-overload model of HF. RESULTS: Daily gallein treatment (10 mg/kg/day), initiated 4 weeks after transverse aortic constriction, improved survival and cardiac function and attenuated cardiac remodeling. Mechanistically, gallein restored ß-AR membrane density in cardiomyocytes, attenuated Gßγ-mediated G-protein-coupled receptor kinase 2-phosphoinositide 3-kinase γ membrane recruitment, and reduced Akt (protein kinase B) and glycogen synthase kinase 3ß phosphorylation. Gallein also reduced circulating plasma catecholamine levels and catecholamine production in isolated mouse adrenal glands by restoring adrenal α2-AR feedback inhibition. In human adrenal endocrine tumors (pheochromocytoma), gallein attenuated catecholamine secretion, as well as G-protein-coupled receptor kinase 2 expression and membrane translocation. CONCLUSIONS: These data suggest small molecule Gßγ inhibition as a systemic pharmacologic therapy for HF by simultaneously normalizing pathologic adrenergic/Gßγ signaling in both the heart and the adrenal gland. Our data also suggest important endocrine/cardiovascular interactions and a possible role for small molecule Gßγ inhibition in treating endocrine tumors such as pheochromocytoma, in addition to HF.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Corazón/efectos de los fármacos , Miocardio/metabolismo , Xantenos/uso terapéutico , Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica , Miocardio/patología , Volumen Sistólico , Resultado del Tratamiento , Xantenos/administración & dosificación
11.
J Mol Cell Cardiol ; 64: 11-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23988739

RESUMEN

Phosphodiesterase 3A (PDE3A) is a major regulator of cAMP in cardiomyocytes. PDE3 inhibitors are used for acute treatment of congestive heart failure, but are associated with increased incidence of arrhythmias and sudden death with long-term use. We previously reported that chronic PDE3A downregulation or inhibition induced myocyte apoptosis in vitro. However, the cardiac protective effect of PDE3A has not been demonstrated in vivo in disease models. In this study, we examined the role of PDE3A in regulating myocardial function and survival in vivo using genetically engineered transgenic mice with myocardial overexpression of the PDE3A1 isozyme (TG). TG mice have reduced cardiac function characterized by reduced heart rate and ejection fraction (52.5±7.8% vs. 83.9±4.7%) as well as compensatory expansion of left ventricular diameter (4.19±0.19mm vs. 3.10±0.18mm). However, there was no maladaptive increase of fibrosis and apoptosis in TG hearts compared to wild type (WT) hearts, and the survival rates also remained the same. The diminution of cardiac contractile function is very likely attributed to a decrease in beta-adrenergic receptor (ß-AR) response in TG mice. Importantly, the myocardial infarct size (4.0±1.8% vs. 24.6±3.8%) and apoptotic cell number (1.3±1.0% vs. 5.6±1.5%) induced by ischemia/reperfusion (I/R) injury were significantly attenuated in TG mice. This was associated with decreased expression of inducible cAMP early repressor (ICER) and increased expression of anti-apoptotic protein BCL-2. To further verify the anti-apoptotic effects of PDE3A1, we performed in vitro apoptosis study in isolated adult TG and WT cardiomyocytes. We found that the apoptotic rates stimulated by hypoxia/reoxygenation or H2O2 were indeed significantly reduced in TG myocytes, and the differences between TG and WT myocytes were completely reversed in the presence of the PDE3 inhibitor milrinone. These together indicate that PDE3A1 negatively regulates ß-AR signaling and protects against I/R injury by inhibiting cardiomyocyte apoptosis.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Expresión Génica , Hemodinámica , Ratones , Ratones Transgénicos , Contracción Miocárdica/genética , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Especificidad de Órganos , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal
12.
Am J Physiol Heart Circ Physiol ; 305(6): H875-84, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23832697

RESUMEN

Mammalian enabled (Mena) is a key regulator of cytoskeletal actin dynamics, which has been implicated in heart failure (HF). We have previously demonstrated that cardiac Mena deletion produced cardiac dysfunction with conduction abnormalities and hypertrophy. Moreover, elevated Mena expression correlates with HF in human and animal models, yet the precise role of Mena in cardiac pathophysiology is unclear. In these studies, we evaluated mice with cardiac myocyte-specific Mena overexpression (TTA/TgTetMena) comparable to that observed in cardiac pathology. We found that the hearts of TTA/TgTetMena mice were functionally and morphologically comparable to wild-type littermates, except for mildly increased heart mass in the transgenic mice. Interestingly, TTA/TgTetMena mice were particularly susceptible to cardiac injury, as these animals experienced pronounced decreases in ejection fraction and fractional shortening as well as heart dilatation and hypertrophy after transverse aortic constriction (TAC). By "turning off" Mena overexpression in TTA/TgTetMena mice either immediately prior to or immediately after TAC surgery, we discovered that normalizing Mena levels eliminated cardiac hypertrophy in TTA/TgTetMena animals but did not preclude post-TAC cardiac functional deterioration. These findings indicate that hearts with increased levels of Mena fare worse when subjected to cardiac injury and suggest that Mena contributes to HF pathophysiology.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/fisiopatología , Miocardio/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Animales , Insuficiencia Cardíaca/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos , Tamaño de los Órganos , Regulación hacia Arriba , Disfunción Ventricular Izquierda/etiología
13.
Cell ; 153(1): 216-27, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23540699

RESUMEN

Phospholipase Cε (PLCε) is a multifunctional enzyme implicated in cardiovascular, pancreatic, and inflammatory functions. Here we show that conditional deletion of PLCε in mouse cardiac myocytes protects from stress-induced pathological hypertrophy. PLCε small interfering RNA (siRNA) in ventricular myocytes decreases endothelin-1 (ET-1)-dependent elevation of nuclear calcium and activation of nuclear protein kinase D (PKD). PLCε scaffolded to muscle-specific A kinase-anchoring protein (mAKAP), along with PKCε and PKD, localizes these components at or near the nuclear envelope, and this complex is required for nuclear PKD activation. Phosphatidylinositol 4-phosphate (PI4P) is identified as a perinuclear substrate in the Golgi apparatus for mAKAP-scaffolded PLCε. We conclude that perinuclear PLCε, scaffolded to mAKAP in cardiac myocytes, responds to hypertrophic stimuli to generate diacylglycerol (DAG) from PI4P in the Golgi apparatus, in close proximity to the nuclear envelope, to regulate activation of nuclear PKD and hypertrophic signaling pathways.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/patología , Fosfatos de Fosfatidilinositol/metabolismo , Fosfoinositido Fosfolipasa C/metabolismo , Animales , Aorta/patología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Aparato de Golgi/metabolismo , Corazón , Ventrículos Cardíacos/citología , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Membrana Nuclear/metabolismo , Fosfoinositido Fosfolipasa C/genética , Ratas , Transducción de Señal
14.
Circulation ; 125(24): 2993-3003, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22610965

RESUMEN

BACKGROUND: Chronic ß-adrenergic receptor (ß-AR) overstimulation, a hallmark of heart failure, is associated with increased cardiac expression of matrix metalloproteinases (MMPs). MMP-1 has been shown to cleave and activate the protease-activated receptor 1 (PAR1) in noncardiac cells. In the present study, we hypothesized that ß-AR stimulation would result in MMP-dependent PAR1 transactivation in cardiac cells. METHODS AND RESULTS: ß-AR stimulation of neonatal rat ventricular myocytes (NRVMs) or cardiac fibroblasts with isoproterenol transduced with an alkaline phosphatase-tagged PAR1 elicited a significant increase in alkaline phosphatase-PAR1 cleavage. This isoproterenol-dependent cleavage was significantly reduced by the broad-spectrum MMP inhibitor GM6001. Importantly, specific MMP-13 inhibitors also decreased alkaline phosphatase-PAR1 cleavage in isoproterenol-stimulated NRVMs, as well as in NRVMs stimulated with conditioned medium from isoproterenol-stimulated cardiac fibroblasts. Moreover, we found that recombinant MMP-13 stimulation cleaved alkaline phosphatase-PAR1 in NRVMs at DPRS(42)↓(43)FLLRN. This also led to the activation of the ERK1/2 pathway through Gαq in NRVMs and via the Gαq/ErbB receptor pathways in cardiac fibroblasts. MMP-13 elicited similar levels of ERK1/2 activation but lower levels of generation of inositol phosphates in comparison to thrombin. Finally, we demonstrated that either PAR1 genetic ablation or pharmacological inhibition of MMP-13 prevented isoproterenol-dependent cardiac dysfunction in mice. CONCLUSIONS: In this study, we demonstrate that ß-AR stimulation leads to MMP-13 transactivation of PAR1 in both cardiac fibroblasts and cardiomyocytes and that this likely contributes to pathological activation of Gαq and ErbB receptor-dependent pathways in the heart. We propose that this mechanism may underlie the development of ß-AR overstimulation-dependent cardiac dysfunction.


Asunto(s)
Metaloproteinasa 13 de la Matriz/fisiología , Miocitos Cardíacos/metabolismo , Receptor PAR-1/metabolismo , Receptores Adrenérgicos beta/fisiología , Activación Transcripcional , Animales , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Oncogénicas v-erbB/fisiología , Transducción de Señal
15.
J Mol Cell Cardiol ; 51(5): 769-76, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21756914

RESUMEN

G-protein-coupled receptor (GPCR)-kinase interacting protein-1 (GIT1) is a multi-function scaffold protein. However, little is known about its physiological role in the heart. Here we sought to identify the cardiac function of GIT1. Global GIT1 knockout (KO) mice were generated and exhibited significant cardiac hypertrophy that progressed to heart failure. Electron microscopy revealed that the hearts of GIT1 KO mice demonstrated significant morphological abnormities in mitochondria, including decreased mitochondrial volume density, cristae density and increased vacuoles. Moreover, mitochondrial biogenesis-related gene peroxisome proliferator-activated receptor γ (PPARγ) co-activator-1α (PGC-1α), PGC-1ß, mitochondrial transcription factor A (Tfam) expression, and total mitochondrial DNA were remarkably decreased in hearts of GIT1 KO mice. These animals also had impaired mitochondrial function, as evidenced by reduced ATP production and dissipated mitochondrial membrane potential (Ψ(m)) in adult cardiomyocytes. Concordant with these mitochondrial observations, GIT1 KO mice showed enhanced cardiomyocyte apoptosis and cardiac dysfunction. In conclusion, our findings identify GIT1 as a new regulator of mitochondrial biogenesis and function, which is necessary for postnatal cardiac maturation.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Proteínas de Ciclo Celular , Proteínas Activadoras de GTPasa , Insuficiencia Cardíaca/metabolismo , Potencial de la Membrana Mitocondrial/genética , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/genética , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Insuficiencia Cardíaca/genética , Ratones , Ratones Noqueados , Microscopía Electrónica , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 31(5): 1116-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21330604

RESUMEN

OBJECTIVE: Cyclophilin A (CyPA, encoded by Ppia) is a proinflammatory protein secreted in response to oxidative stress in mice and humans. We recently demonstrated that CyPA increased angiotensin II (Ang II)-induced reactive oxygen species (ROS) production in the aortas of apolipoprotein E (Apoe)-/- mice. In this study, we sought to evaluate the role of CyPA in Ang II-induced cardiac hypertrophy. METHODS AND RESULTS: Cardiac hypertrophy was not significantly different between Ppia+/+ and Ppia-/- mice infused with Ang II (1000 ng/min per kg for 4 weeks). Therefore, we investigated the effect of CyPA under conditions of high ROS and inflammation using the Apoe-/- mice. In contrast to Apoe-/- mice, Apoe-/-Ppia-/- mice exhibited significantly less Ang II-induced cardiac hypertrophy. Bone marrow cell transplantation showed that CyPA in cells intrinsic to the heart plays an important role in the cardiac hypertrophic response. Ang II-induced ROS production, cardiac fibroblast proliferation, and cardiac fibroblast migration were markedly decreased in Apoe-/-Ppia-/- cardiac fibroblasts. Furthermore, CyPA directly induced the hypertrophy of cultured neonatal cardiac myocytes. CONCLUSIONS: CyPA is required for Ang II-mediated cardiac hypertrophy by directly potentiating ROS production, stimulating the proliferation and migration of cardiac fibroblasts, and promoting cardiac myocyte hypertrophy.


Asunto(s)
Apolipoproteínas E/deficiencia , Cardiomegalia/enzimología , Ciclofilina A/metabolismo , Miocardio/enzimología , Angiotensina II , Animales , Animales Recién Nacidos , Apolipoproteínas E/genética , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/inmunología , Cardiomegalia/patología , Cardiomegalia/prevención & control , Comunicación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Ciclofilina A/deficiencia , Ciclofilina A/genética , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo
17.
J Biol Chem ; 284(3): 1514-22, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-18957419

RESUMEN

Recently, we identified a novel signaling pathway involving Epac, Rap, and phospholipase C (PLC)epsilon that plays a critical role in maximal beta-adrenergic receptor (betaAR) stimulation of Ca2+-induced Ca2+ release (CICR) in cardiac myocytes. Here we demonstrate that PLCepsilon phosphatidylinositol 4,5-bisphosphate hydrolytic activity and PLCepsilon-stimulated Rap1 GEF activity are both required for PLCepsilon-mediated enhancement of sarcoplasmic reticulum Ca2+ release and that PLCepsilon significantly enhances Rap activation in response to betaAR stimulation in the heart. Downstream of PLCepsilon hydrolytic activity, pharmacological inhibition of PKC significantly inhibited both betaAR- and Epac-stimulated increases in CICR in PLCepsilon+/+ myocytes but had no effect in PLCepsilon-/- myocytes. betaAR and Epac activation caused membrane translocation of PKCepsilon in PLCepsilon+/+ but not PLCepsilon-/- myocytes and small interfering RNA-mediated PKCepsilon knockdown significantly inhibited both betaAR and Epac-mediated CICR enhancement. Further downstream, the Ca2+/calmodulin-dependent protein kinase II (CamKII) inhibitor, KN93, inhibited betaAR- and Epac-mediated CICR in PLCepsilon+/+ but not PLCepsilon-/- myocytes. Epac activation increased CamKII Thr286 phosphorylation and enhanced phosphorylation at CamKII phosphorylation sites on the ryanodine receptor (RyR2) (Ser2815) and phospholamban (Thr17) in a PKC-dependent manner. Perforated patch clamp experiments revealed that basal and betaAR-stimulated peak L-type current density are similar in PLCepsilon+/+ and PLCepsilon-/- myocytes suggesting that control of sarcoplasmic reticulum Ca2+ release, rather than Ca2+ influx through L-type Ca2+ channels, is the target of regulation of a novel signal transduction pathway involving sequential activation of Epac, PLCepsilon, PKCepsilon, and CamKII downstream of betaAR activation.


Asunto(s)
Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Miocardio/enzimología , Fosfoinositido Fosfolipasa C/metabolismo , Animales , Bencilaminas/farmacología , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Membrana Celular/enzimología , Membrana Celular/genética , Activación Enzimática/fisiología , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Ratones Noqueados , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/enzimología , Fosfatidilinositol 4,5-Difosfato , Fosfoinositido Fosfolipasa C/genética , Fosforilación/fisiología , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/fisiología , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/enzimología , Retículo Sarcoplasmático/genética , Sulfonamidas/farmacología
18.
Circ Res ; 104(1): 113-23, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19023134

RESUMEN

By mimicking sympathetic stimulation in vivo, we previously reported that mice globally lacking serotonin 5-HT(2B) receptors did not develop isoproterenol-induced left ventricular hypertrophy. However, the exact cardiac cell type(s) expressing 5-HT(2B) receptors (cardiomyocytes versus noncardiomyocytes) involved in pathological heart hypertrophy was never addressed in vivo. We report here that mice expressing the 5-HT(2B) receptor solely in cardiomyocytes, like global 5-HT(2B) receptor-null mice, are resistant to isoproterenol-induced cardiac hypertrophy and dysfunction, as well as to isoproterenol-induced increases in cytokine plasma-levels. These data reveal a key role of noncardiomyocytes in isoproterenol-induced hypertrophy in vivo. Interestingly, we show that primary cultures of angiotensinogen null adult cardiac fibroblasts are releasing cytokines on stimulation with either angiotensin II or serotonin, but not in response to isoproterenol stimulation, demonstrating a critical role of angiotensinogen in adrenergic-dependent cytokine production. We then show a functional interdependence between AT(1)Rs and 5-HT(2B) receptors in fibroblasts by revealing a transinhibition mechanism that may involve heterodimeric receptor complexes. Both serotonin- and angiotensin II-dependent cytokine production occur via a Src/heparin-binding epidermal growth factor-dependent transactivation of epidermal growth factor receptors in cardiac fibroblasts, supporting a common signaling pathway. Finally, we demonstrate that 5-HT(2B) receptors are overexpressed in hearts from patients with congestive heart failure, this overexpression being positively correlated with cytokine and norepinephrine plasma levels. Collectively, these results reveal for the first time that interactions between AT(1) and 5-HT(2B) receptors coexpressed by noncardiomyocytes are limiting key events in adrenergic agonist-induced, angiotensin-dependent cardiac hypertrophy. Accordingly, antagonists of 5-HT(2B) receptors might represent novel therapeutics for sympathetic overstimulation-dependent heart failure.


Asunto(s)
Fibroblastos/fisiología , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/fisiopatología , Miocardio/patología , Receptor de Angiotensina Tipo 1/fisiología , Receptor de Serotonina 5-HT2B/fisiología , Adulto , Angiotensina II/deficiencia , Angiotensina II/fisiología , Angiotensina II/toxicidad , Animales , Células Cultivadas/metabolismo , Citocinas/sangre , Citocinas/metabolismo , Receptores ErbB/fisiología , Femenino , Fibroblastos/efectos de los fármacos , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Hipertrofia Ventricular Izquierda/inducido químicamente , Hipertrofia Ventricular Izquierda/prevención & control , Péptidos y Proteínas de Señalización Intercelular/fisiología , Isoproterenol/toxicidad , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Norepinefrina/fisiología , Mapeo de Interacción de Proteínas , Antagonistas del Receptor de Serotonina 5-HT2 , Antagonistas de la Serotonina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/fisiología
19.
Am J Physiol Heart Circ Physiol ; 294(3): H1335-47, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18192223

RESUMEN

In contrast to the other heterotrimeric GTP-binding proteins (G proteins) Gs and Gi, the functional role of G o is still poorly defined. To investigate the role of G alpha o in the heart, we generated transgenic mice with cardiac-specific expression of a constitutively active form of G alpha o1* (G alpha o*), the predominant G alpha o isoform in the heart. G alpha o expression was increased 3- to 15-fold in mice from 5 independent lines, all of which had a normal life span and no gross cardiac morphological abnormalities. We demonstrate enhanced contractile function in G alpha o* transgenic mice in vivo, along with increased L-type Ca2+ channel current density, calcium transients, and cell shortening in ventricular G alpha o*-expressing myocytes compared with wild-type controls. These changes were evident at baseline and maintained after isoproterenol stimulation. Expression levels of all major Ca2+ handling proteins were largely unchanged, except for a modest reduction in Na+/Ca2+ exchanger in transgenic ventricles. In contrast, phosphorylation of the ryanodine receptor and phospholamban at known PKA sites was increased 1.6- and 1.9-fold, respectively, in G alpha o* ventricles. Density and affinity of beta-adrenoceptors, cAMP levels, and PKA activity were comparable in G alpha o* and wild-type myocytes, but protein phosphatase 1 activity was reduced upon G alpha o* expression, particularly in the vicinity of the ryanodine receptor. We conclude that G alpha o* exerts a positive effect on Ca2+ cycling and contractile function. Alterations in protein phosphatase 1 activity rather than PKA-mediated phosphorylation might be involved in hyperphosphorylation of key Ca2+ handling proteins in hearts with constitutive G alpha o activation.


Asunto(s)
Calcio/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/biosíntesis , Contracción Miocárdica/genética , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Citoesqueleto de Actina/fisiología , Infecciones por Adenoviridae/patología , Agonistas Adrenérgicos beta/farmacología , Animales , Animales Modificados Genéticamente , Northern Blotting , Western Blotting , Canales de Calcio Tipo L/fisiología , Señalización del Calcio/fisiología , Separación Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocromos c/biosíntesis , Isoproterenol/farmacología , Ratones , Miocitos Cardíacos/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/fisiología
20.
Physiol Genomics ; 32(2): 182-9, 2008 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-17971502

RESUMEN

Numerous genetically engineered animal models of heart failure (HF) exhibit multiple characteristics of human HF, including aberrant beta-adrenergic signaling. Several of these HF models can be rescued by cardiac-targeted expression of the Gbetagamma inhibitory carboxy-terminus of the beta-adrenergic receptor kinase (betaARKct). We recently reported microarray analysis of gene expression in multiple animal models of HF and their betaARKct rescue, where we identified gene expression patterns distinct and predictive of HF and rescue. We have further investigated the muscle LIM protein knockout model of HF (MLP-/-), which closely parallels human dilated cardiomyopathy disease progression and aberrant beta-adrenergic signaling, and their betaARKct rescue. A group of known and novel genes was identified and validated by quantitative real-time PCR whose expression levels predicted phenotype in both the larger HF group and in the MLP-/- subset. One of these novel genes is herein identified as Nogo, a protein widely studied in the nervous system, where it plays a role in regeneration. Nogo expression is altered in HF and normalized with rescue, in an isoform-specific manner, using left ventricular tissue harvested from both animal and human subjects. To investigate cell type-specific expression of Nogo in the heart, immunofluorescence and confocal microscopy were utilized. Nogo expression appears to be most clearly associated with cardiac fibroblasts. To our knowledge, this is the first report to demonstrate the relationship between Nogo expression and HF, including cell-type specificity, in both mouse and human HF and phenotypic rescue.


Asunto(s)
Insuficiencia Cardíaca/patología , Proteínas Musculares/genética , Proteínas de la Mielina/genética , Miocardio/patología , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Eliminación de Gen , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Humanos , Inmunohistoquímica , Proteínas con Dominio LIM , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Proteínas de la Mielina/metabolismo , Miocardio/metabolismo , Proteínas Nogo , Análisis de Secuencia por Matrices de Oligonucleótidos , Quinasas de Receptores Adrenérgicos beta/genética , Quinasas de Receptores Adrenérgicos beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA