Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(3): 545-562.e23, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32621799

RESUMEN

Scar tissue size following myocardial infarction is an independent predictor of cardiovascular outcomes, yet little is known about factors regulating scar size. We demonstrate that collagen V, a minor constituent of heart scars, regulates the size of heart scars after ischemic injury. Depletion of collagen V led to a paradoxical increase in post-infarction scar size with worsening of heart function. A systems genetics approach across 100 in-bred strains of mice demonstrated that collagen V is a critical driver of postinjury heart function. We show that collagen V deficiency alters the mechanical properties of scar tissue, and altered reciprocal feedback between matrix and cells induces expression of mechanosensitive integrins that drive fibroblast activation and increase scar size. Cilengitide, an inhibitor of specific integrins, rescues the phenotype of increased post-injury scarring in collagen-V-deficient mice. These observations demonstrate that collagen V regulates scar size in an integrin-dependent manner.


Asunto(s)
Cicatriz/metabolismo , Colágeno Tipo V/deficiencia , Colágeno Tipo V/metabolismo , Lesiones Cardíacas/metabolismo , Contracción Miocárdica/genética , Miofibroblastos/metabolismo , Animales , Cicatriz/genética , Cicatriz/fisiopatología , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Colágeno Tipo V/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Fibrosis/genética , Fibrosis/metabolismo , Regulación de la Expresión Génica/genética , Integrinas/antagonistas & inhibidores , Integrinas/genética , Integrinas/metabolismo , Isoproterenol/farmacología , Masculino , Mecanotransducción Celular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía de Fuerza Atómica/instrumentación , Microscopía Electrónica de Transmisión , Contracción Miocárdica/efectos de los fármacos , Miofibroblastos/citología , Miofibroblastos/patología , Miofibroblastos/ultraestructura , Análisis de Componente Principal , Proteómica , RNA-Seq , Análisis de la Célula Individual
2.
Proc Natl Acad Sci U S A ; 116(13): 6172-6180, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30867288

RESUMEN

Heart performance relies on highly coordinated excitation-contraction (EC) coupling, and defects in this critical process may be exacerbated by additional genetic defects and/or environmental insults to cause eventual heart failure. Here we report a regulatory pathway consisting of the RNA binding protein RBFox2, a stress-induced microRNA miR-34a, and the essential EC coupler JPH2. In this pathway, initial cardiac defects diminish RBFox2 expression, which induces transcriptional repression of miR-34a, and elevated miR-34a targets Jph2 to impair EC coupling, which further manifests heart dysfunction, leading to progressive heart failure. The key contribution of miR-34a to this process is further established by administrating its mimic, which is sufficient to induce cardiac defects, and by using its antagomir to alleviate RBFox2 depletion-induced heart dysfunction. These findings elucidate a potential feed-forward mechanism to account for a critical transition to cardiac decompensation and suggest a potential therapeutic avenue against heart failure.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Corazón/fisiopatología , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Proteínas Musculares/metabolismo , Factores de Empalme de ARN/metabolismo , Animales , Regulación hacia Abajo , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología
3.
J Mol Cell Cardiol ; 160: 73-86, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34273410

RESUMEN

The temporal nature of chromatin structural changes underpinning pathologic transcription are poorly understood. We measured chromatin accessibility and DNA methylation to study the contribution of chromatin remodeling at different stages of cardiac hypertrophy and failure. ATAC-seq and reduced representation bisulfite sequencing were performed in cardiac myocytes after transverse aortic constriction (TAC) or depletion of the chromatin structural protein CTCF. Early compensation to pressure overload showed changes in chromatin accessibility and DNA methylation preferentially localized to intergenic and intronic regions. Most methylation and accessibility changes observed in enhancers and promoters at the late phase (3 weeks after TAC) were established at an earlier time point (3 days after TAC), before heart failure manifests. Enhancers were paired with genes based on chromatin conformation capture data: while enhancer accessibility generally correlated with changes in gene expression, this feature, nor DNA methylation, was alone sufficient to predict transcription of all enhancer interacting genes. Enrichment of transcription factors and active histone marks at these regions suggests that enhancer activity coordinates with other epigenetic factors to determine gene transcription. In support of this hypothesis, ChIP-qPCR demonstrated increased enhancer and promoter occupancy of GATA4 and NKX2.5 at Itga9 and Nppa, respectively, concomitant with increased transcription of these genes in the diseased heart. Lastly, we demonstrate that accessibility and DNA methylation are imperfect predictors of chromatin structure at the scale of A/B compartmentalization-rather, accessibility, DNA methylation, transcription factors and other histone marks work within these domains to determine gene expression. These studies establish that chromatin reorganization during early compensation after pathologic stimuli is maintained into the later decompensatory phases of heart failure. The findings reveal the rules for how local chromatin features govern gene expression in the context of global genomic structure and identify chromatin remodeling events for therapeutic targeting in disease.


Asunto(s)
Cardiomegalia/genética , Cardiomegalia/metabolismo , Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , Cromatina/metabolismo , Epigénesis Genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Fenotipo , Animales , Metilación de ADN/genética , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos/genética , Expresión Génica , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas/genética , Ratas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
J Mol Cell Cardiol ; 128: 198-211, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30742811

RESUMEN

Heart failure is associated with hypertrophying of cardiomyocytes and changes in transcriptional activity. Studies from rapidly dividing cells in culture have suggested that transcription may be compartmentalized into factories within the nucleus, but this phenomenon has not been tested in vivo and the role of nuclear architecture in cardiac gene regulation is unknown. While alterations to transcription have been linked to disease, little is known about the regulation of the spatial organization of transcription and its properties in the pathological setting. In the present study, we investigate the structural features of endogenous transcription factories in the heart and determine the principles connecting chromatin structure to transcriptional regulation in vivo. Super-resolution imaging of endogenous RNA polymerase II clusters in neonatal and adult cardiomyocytes revealed distinct properties of transcription factories in response to pathological stress: neonatal nuclei demonstrated changes in number of clusters, with parallel increases in nuclear area, while the adult nuclei underwent changes in size and intensity of RNA polymerase II foci. Fluorescence in situ hybridization-based labeling of genes revealed locus-specific relationships between expression change and anatomical localization-with respect to nuclear periphery and heterochromatin regions, both sites associated with gene silencing-in the nuclei of cardiomyocytes in hearts (but not liver hepatocytes) of mice subjected to pathologic stimuli that induce heart failure. These findings demonstrate a role for chromatin organization and rearrangement of nuclear architecture for cell type-specific transcription in vivo during disease. RNA polymerase II ChIP and chromatin conformation capture studies in the same model system demonstrate formation and reorganization of distinct nuclear compartments regulating gene expression. These findings reveal locus-specific compartmentalization of stress-activated, housekeeping and silenced genes in the anatomical context of the endogenous nucleus, revealing basic principles of global chromatin structure and nuclear architecture in the regulation of gene expression in healthy and diseased conditions.


Asunto(s)
Insuficiencia Cardíaca/genética , Corazón/diagnóstico por imagen , ARN Polimerasa II/genética , Transcripción Genética/genética , Animales , Animales Recién Nacidos , Cromatina/genética , Cromatina/aislamiento & purificación , Regulación de la Expresión Génica , Corazón/fisiopatología , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/diagnóstico por imagen , Humanos , Hibridación Fluorescente in Situ , Ratones , Imagen Molecular/métodos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Polimerasa II/aislamiento & purificación , Activación Transcripcional/genética
5.
J Biol Chem ; 293(25): 9652-9661, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29769316

RESUMEN

Heart failure is associated with induction of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). The serine/threonine protein kinase/endoribonuclease IRE1α is a key protein in ER stress signal transduction. IRE1α activity can induce both protective UPR and apoptotic downstream signaling events, but the specific role for IRE1α activity in the heart is unknown. A major aim of this study was to characterize the specific contribution of IRE1α in cardiac physiology and pathogenesis. We used both cultured myocytes and a transgenic mouse line with inducible and cardiomyocyte-specific IRE1α overexpression as experimental models to achieve targeted IRE1α activation. IRE1α expression induced a potent but transient ER stress response in cardiomyocytes and did not cause significant effects in the intact heart under normal physiological conditions. Furthermore, the IRE1α-activated transgenic heart responding to pressure overload exhibited preserved function and reduced fibrotic area, associated with increased adaptive UPR signaling and with blunted inflammatory and pathological gene expression. Therefore, we conclude that IRE1α induces transient ER stress signaling and confers a protective effect against pressure overload-induced pathological remodeling in the heart. To our knowledge, this report provides first direct evidence of a specific and protective role for IRE1α in the heart and reveals an interaction between ER stress signaling and inflammatory regulation in the pathologically stressed heart.


Asunto(s)
Estrés del Retículo Endoplásmico , Endorribonucleasas/fisiología , Insuficiencia Cardíaca/prevención & control , Insulinoma/prevención & control , Presión/efectos adversos , Sustancias Protectoras/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Apoptosis , Células Cultivadas , Femenino , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Insulinoma/metabolismo , Insulinoma/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Transducción de Señal , Respuesta de Proteína Desplegada
6.
PLoS Genet ; 12(7): e1006038, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27385019

RESUMEN

We aimed to understand the genetic control of cardiac remodeling using an isoproterenol-induced heart failure model in mice, which allowed control of confounding factors in an experimental setting. We characterized the changes in cardiac structure and function in response to chronic isoproterenol infusion using echocardiography in a panel of 104 inbred mouse strains. We showed that cardiac structure and function, whether under normal or stress conditions, has a strong genetic component, with heritability estimates of left ventricular mass between 61% and 81%. Association analyses of cardiac remodeling traits, corrected for population structure, body size and heart rate, revealed 17 genome-wide significant loci, including several loci containing previously implicated genes. Cardiac tissue gene expression profiling, expression quantitative trait loci, expression-phenotype correlation, and coding sequence variation analyses were performed to prioritize candidate genes and to generate hypotheses for downstream mechanistic studies. Using this approach, we have validated a novel gene, Myh14, as a negative regulator of ISO-induced left ventricular mass hypertrophy in an in vivo mouse model and demonstrated the up-regulation of immediate early gene Myc, fetal gene Nppb, and fibrosis gene Lgals3 in ISO-treated Myh14 deficient hearts compared to controls.


Asunto(s)
Galectina 3/biosíntesis , Insuficiencia Cardíaca/genética , Hipertrofia Ventricular Izquierda/genética , Cadenas Pesadas de Miosina/biosíntesis , Miosina Tipo II/biosíntesis , Péptido Natriurético Encefálico/biosíntesis , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Galectina 3/genética , Regulación de la Expresión Génica , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/patología , Frecuencia Cardíaca/genética , Humanos , Hipertrofia Ventricular Izquierda/inducido químicamente , Hipertrofia Ventricular Izquierda/patología , Isoproterenol/toxicidad , Ratones , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Péptido Natriurético Encefálico/genética , Sitios de Carácter Cuantitativo/genética , Remodelación Ventricular/genética
7.
J Biol Chem ; 292(31): 12787-12800, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28637870

RESUMEN

Stress-induced p38 mitogen-activated protein kinase (MAPK) activity is implicated in pathological remodeling in the heart. For example, constitutive p38 MAPK activation in cardiomyocytes induces pathological features, including myocyte hypertrophy, apoptosis, contractile dysfunction, and fetal gene expression. However, the physiological function of cardiomyocyte p38 MAPK activity in beneficial compensatory vascular remodeling is unclear. This report investigated the functional role and the underlying mechanisms of cardiomyocyte p38 MAPK activity in cardiac remodeling induced by chronic stress. Using both in vitro and in vivo model systems, we found that p38 MAPK activity is required for hypoxia-induced pro-angiogenic activity from cardiomyocytes and that p38 MAPK activation in cardiomyocyte is sufficient to promote paracrine signaling-mediated, pro-angiogenic activity. We further demonstrate that VEGF is a paracrine factor responsible for the p38 MAPK-mediated pro-angiogenic activity from cardiomyocytes and that p38 MAPK pathway activation is sufficient for inducing VEGF secretion from cardiomyocytes in an Sp1-dependent manner. More significantly, cardiomyocyte-specific inactivation of p38α in mouse heart impaired compensatory angiogenesis after pressure overload and promoted early onset of heart failure. In summary, p38αMAPK has a critical role in the cross-talk between cardiomyocytes and vasculature by regulating stress-induced VEGF expression and secretion in cardiomyocytes. We conclude that as part of a stress-induced signaling pathway, p38 MAPK activity significantly contributes to both pathological and compensatory remodeling in the heart.


Asunto(s)
Endotelio Vascular/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Isquemia Miocárdica/metabolismo , Revascularización Miocárdica , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Hipoxia de la Célula , Células Cultivadas , Cruzamientos Genéticos , Endotelio Vascular/citología , Endotelio Vascular/patología , Activación Enzimática , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/genética , Isquemia Miocárdica/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Interferencia de ARN , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Sus scrofa , Factor A de Crecimiento Endotelial Vascular/agonistas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/agonistas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Circulation ; 136(17): 1613-1625, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-28802249

RESUMEN

BACKGROUND: Cardiovascular disease is associated with epigenomic changes in the heart; however, the endogenous structure of cardiac myocyte chromatin has never been determined. METHODS: To investigate the mechanisms of epigenomic function in the heart, genome-wide chromatin conformation capture (Hi-C) and DNA sequencing were performed in adult cardiac myocytes following development of pressure overload-induced hypertrophy. Mice with cardiac-specific deletion of CTCF (a ubiquitous chromatin structural protein) were generated to explore the role of this protein in chromatin structure and cardiac phenotype. Transcriptome analyses by RNA-seq were conducted as a functional readout of the epigenomic structural changes. RESULTS: Depletion of CTCF was sufficient to induce heart failure in mice, and human patients with heart failure receiving mechanical unloading via left ventricular assist devices show increased CTCF abundance. Chromatin structural analyses revealed interactions within the cardiac myocyte genome at 5-kb resolution, enabling examination of intra- and interchromosomal events, and providing a resource for future cardiac epigenomic investigations. Pressure overload or CTCF depletion selectively altered boundary strength between topologically associating domains and A/B compartmentalization, measurements of genome accessibility. Heart failure involved decreased stability of chromatin interactions around disease-causing genes. In addition, pressure overload or CTCF depletion remodeled long-range interactions of cardiac enhancers, resulting in a significant decrease in local chromatin interactions around these functional elements. CONCLUSIONS: These findings provide a high-resolution chromatin architecture resource for cardiac epigenomic investigations and demonstrate that global structural remodeling of chromatin underpins heart failure. The newly identified principles of endogenous chromatin structure have key implications for epigenetic therapy.


Asunto(s)
Cardiomegalia/metabolismo , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Epigénesis Genética , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cromatina/genética , Cromatina/patología , Estudio de Asociación del Genoma Completo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología
9.
Am J Physiol Heart Circ Physiol ; 315(3): H634-H643, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29775411

RESUMEN

The chemotherapeutic effect of doxorubicin (Dox) is limited by cumulative dose-dependent cardiotoxicity in cancer survivors. Dexrazoxane (DRZ) is approved to prevent Dox-induced cardiotoxicity. Humanin and its synthetic analog HNG have a cytoprotective effect on the heart. To investigate the cardioprotective efficacy of HNG alone or in combination with DRZ against Dox-induced cardiotoxicity, 80 adult male mice were randomly divided into 8 groups to receive the following treatments via intraperitoneal injection: saline dailym HNG (5 mg/kg) daily, DRZ (60 mg/kg) weekly, Dox (3 mg/kg) weekly, DRZ + HNG, Dox + HNG, Dox + DRZ, and Dox + HNG + DRZ. Echocardiograms were performed before and at 4, 8, and 9.5 wk after the beginning of treatment. All mice were euthanized at 10 wk. In the absence of Dox, HNG, DRZ, or DRZ + HNG had no adverse effect on the heart. Dox treatment caused decreases in ejection fraction and cardiac mass and increases in cardiomyocyte apoptosis and intracardiac fibrosis. HNG or DRZ alone blunted the Dox-induced decrease in left ventricle posterior wall thickness and modestly ameliorated the Dox-induced decrease in ejection fraction. HNG + DRZ significantly ameliorated Dox-induced decreases in ejection function, cardiac fibrosis, and cardiac mass. Using a targeted analysis for the mitochondrial gene array and protein expression in heart tissues, we demonstrated that HNG + DRZ reversed DOX-induced altered transcripts that were biomarkers of cardiac damage and uncoupling protein-2. We conclude that HNG enhances the cardiac protective effect of DRZ against Dox-induced cardiotoxicity. HNG + DRZ protects mitochondria from Dox-induced cardiac damage and blunts the onset of cardiac dysfunction. Thus, HNG may be an adjuvant to DRZ in preventing Dox-induced cardiotoxicity. NEW & NOTEWORTHY Doxorubicin (Dox) is commonly used for treating a wide range of human cancers. However, cumulative dosage-dependent carditoxicity often limits its clinical applications. We demonstrated in this study that treating young adult male mice with synthetic humanin analog enhanced the cardiac protective effect of dexrazoxane against chemotherapeutic agent Dox-induced cardiac dysfunction. Thus, humanin analog can potentially serve as an adjuvant to dexrazoxane in more effectively preventing Dox-induced cardiac dysfunction and cardiomyopathy.


Asunto(s)
Cardiotónicos/farmacología , Dexrazoxano/farmacología , Péptidos y Proteínas de Señalización Intracelular/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Cardiotónicos/administración & dosificación , Cardiotoxicidad , Dexrazoxano/administración & dosificación , Doxorrubicina/toxicidad , Sinergismo Farmacológico , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo
10.
Circ Res ; 118(5): 786-97, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26838786

RESUMEN

RATIONALE: Only a small portion of the known heritability of cardiovascular diseases, such as heart failure, can be explained based on single-gene mutations. Chromatin structure and regulation provide a substrate through which genetic differences in noncoding regions may affect cellular function and response to disease, but the mechanisms are unknown. OBJECTIVE: We conducted genome-wide measurements of DNA methylation in different strains of mice that are susceptible and resistant to isoproterenol-induced dysfunction to test the hypothesis that this epigenetic mark may play a causal role in the development of heart failure. METHODS AND RESULTS: BALB/cJ and BUB/BnJ mice, determined to be susceptible and resistant to isoproterenol-induced heart failure, respectively, were administered the drug for 3 weeks via osmotic minipump. Reduced representational bisulfite sequencing was then used to compare the differences between the cardiac DNA methylomes in the basal state between strains and then after isoproterenol treatment. Single-base resolution DNA methylation measurements were obtained and revealed a bimodal distribution of methylation in the heart, enriched in lone intergenic CpGs and depleted from CpG islands around genes. Isoproterenol induced global decreases in methylation in both strains; however, the basal methylation pattern between strains shows striking differences that may be predictive of disease progression before environmental stress. The global correlation between promoter methylation and gene expression (as measured by microarray) was modest and revealed itself only with focused analyses of transcription start site and gene body regions (in contrast to when gene methylation was examined in toto). Modules of comethylated genes displayed correlation with other protein-based epigenetic marks, supporting the hypothesis that chromatin modifications act in a combinatorial manner to specify transcriptional phenotypes in the heart. CONCLUSIONS: This study provides the first single-base resolution map of the mammalian cardiac DNA methylome and the first case-control analysis of the changes in DNA methylation with heart failure. The findings demonstrate marked genetic differences in DNA methylation that are associated with disease progression.


Asunto(s)
Cromatina/fisiología , Metilación de ADN/fisiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Isoproterenol/toxicidad , Animales , Cardiotónicos/toxicidad , Islas de CpG/fisiología , Susceptibilidad a Enfermedades , Femenino , Insuficiencia Cardíaca/inducido químicamente , Ratones , Ratones Endogámicos BALB C , Especificidad de la Especie
11.
Circulation ; 133(21): 2038-49, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27059949

RESUMEN

BACKGROUND: Although metabolic reprogramming is critical in the pathogenesis of heart failure, studies to date have focused principally on fatty acid and glucose metabolism. Contribution of amino acid metabolic regulation in the disease remains understudied. METHODS AND RESULTS: Transcriptomic and metabolomic analyses were performed in mouse failing heart induced by pressure overload. Suppression of branched-chain amino acid (BCAA) catabolic gene expression along with concomitant tissue accumulation of branched-chain α-keto acids was identified as a significant signature of metabolic reprogramming in mouse failing hearts and validated to be shared in human cardiomyopathy hearts. Molecular and genetic evidence identified the transcription factor Krüppel-like factor 15 as a key upstream regulator of the BCAA catabolic regulation in the heart. Studies using a genetic mouse model revealed that BCAA catabolic defect promoted heart failure associated with induced oxidative stress and metabolic disturbance in response to mechanical overload. Mechanistically, elevated branched-chain α-keto acids directly suppressed respiration and induced superoxide production in isolated mitochondria. Finally, pharmacological enhancement of branched-chain α-keto acid dehydrogenase activity significantly blunted cardiac dysfunction after pressure overload. CONCLUSIONS: BCAA catabolic defect is a metabolic hallmark of failing heart resulting from Krüppel-like factor 15-mediated transcriptional reprogramming. BCAA catabolic defect imposes a previously unappreciated significant contribution to heart failure.


Asunto(s)
Aminoácidos de Cadena Ramificada/genética , Aminoácidos de Cadena Ramificada/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Animales , Insuficiencia Cardíaca/patología , Humanos , Masculino , Metabolismo/fisiología , Metabolómica , Ratones , Ratones Noqueados , Transcriptoma
12.
Am J Physiol Heart Circ Physiol ; 311(5): H1234-H1247, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27663768

RESUMEN

All terminally differentiated organs face two challenges, maintaining their cellular identity and restricting organ size. The molecular mechanisms responsible for these decisions are of critical importance to organismal development, and perturbations in their normal balance can lead to disease. A hallmark of heart failure, a condition affecting millions of people worldwide, is hypertrophic growth of cardiomyocytes. The various forms of heart failure in human and animal models share conserved transcriptome remodeling events that lead to expression of genes normally silenced in the healthy adult heart. However, the chromatin remodeling events that maintain cell and organ size are incompletely understood; insights into these mechanisms could provide new targets for heart failure therapy. Using a quantitative proteomics approach to identify muscle-specific chromatin regulators in a mouse model of hypertrophy and heart failure, we identified upregulation of the histone methyltransferase Smyd1 during disease. Inducible loss-of-function studies in vivo demonstrate that Smyd1 is responsible for restricting growth in the adult heart, with its absence leading to cellular hypertrophy, organ remodeling, and fulminate heart failure. Molecular studies reveal Smyd1 to be a muscle-specific regulator of gene expression and indicate that Smyd1 modulates expression of gene isoforms whose expression is associated with cardiac pathology. Importantly, activation of Smyd1 can prevent pathological cell growth. These findings have basic implications for our understanding of cardiac pathologies and open new avenues to the treatment of cardiac hypertrophy and failure by modulating Smyd1.


Asunto(s)
Cardiomegalia/genética , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Insuficiencia Cardíaca/genética , Proteínas Musculares/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética , Animales , Western Blotting , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/metabolismo , Aumento de la Célula , Ecocardiografía , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Células HeLa , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos/patología , Proteómica , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba , Remodelación Ventricular/genética
13.
J Mol Cell Cardiol ; 79: 115-22, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25446185

RESUMEN

Defect in mitochondrial biogenesis and cardiac energy metabolism is a critical contributing factor to cardiac hypertrophy and heart failure. Sentrin/SUMO specific protease 1 (SENP1) mediated regulation of PGC-1α transcriptional activity plays an essential role in mitochondrial biogenesis and mitochondrial function. However, whether SENP1 plays a role in cardiac hypertrophy and failure is unknown. We investigated whether alteration in SENP1 expression affects cardiomyopathy and the underlying mechanism. In our present study, we found that the expression of SENP1 was induced in mouse and human failing hearts associated with induced expression of mitochondrial genes. SENP1 expression in cardiomyocytes was induced by hypertrophic stimuli through calcium/calcineurin-NFAT3. SENP1 regulated mitochondrial gene expression by de-SUMOylation of MEF-2C, which enhanced MEF-2C-mediated PGC-1α transcription. Genetic induction of SENP1 led to mitochondrial dysregulation and cardiac dysfunction in vivo. Our data showed that pathogenesis of cardiomyopathy is attributed by SENP1 mediated regulation of mitochondrial abnormities. SENP1 up-regulation in diseased heart is mediated via calcineurin-NFAT/MEF2C-PGC-1α pathway.


Asunto(s)
Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Endopeptidasas/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Animales , Calcineurina/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Cisteína Endopeptidasas , Endopeptidasas/genética , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Factores de Transcripción MEF2/metabolismo , Ratones , Mitocondrias/ultraestructura , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Factores de Transcripción NFATC/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Sumoilación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
14.
Development ; 139(23): 4449-60, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23095891

RESUMEN

The Notch signaling pathway is an important contributor to the development and homeostasis of the cardiovascular system. Not surprisingly, mutations in Notch receptors and ligands have been linked to a variety of hereditary diseases that impact both the heart and the vasculature. In particular, mutations in the gene encoding the human Notch ligand jagged 1 result in a multisystem autosomal dominant disorder called Alagille syndrome, which includes tetralogy of Fallot among its more severe cardiac pathologies. Jagged 1 is expressed throughout the developing embryo, particularly in endothelial cells. Here, we demonstrate that endothelial-specific deletion of Jag1 leads to cardiovascular defects in both embryonic and adult mice that are reminiscent of those in Alagille syndrome. Mutant mice display right ventricular hypertrophy, overriding aorta, ventricular septal defects, coronary vessel abnormalities and valve defects. Examination of mid-gestational embryos revealed that the loss of Jag1, similar to the loss of Notch1, disrupts endothelial-to-mesenchymal transition during endocardial cushion formation. Furthermore, adult mutant mice exhibit cardiac valve calcifications associated with abnormal matrix remodeling and induction of bone morphogenesis. This work shows that the endothelium is responsible for the wide spectrum of cardiac phenotypes displayed in Alagille Syndrome and it demonstrates a crucial role for Jag1 in valve morphogenesis.


Asunto(s)
Síndrome de Alagille/genética , Calcinosis/genética , Proteínas de Unión al Calcio/genética , Cardiomiopatías/genética , Cardiopatías Congénitas/genética , Enfermedades de las Válvulas Cardíacas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/metabolismo , Anomalías de los Vasos Coronarios/genética , Anomalías de los Vasos Coronarios/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interventricular/genética , Defectos del Tabique Interventricular/metabolismo , Enfermedades de las Válvulas Cardíacas/metabolismo , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/metabolismo , Proteína Jagged-1 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Técnicas de Cultivo de Órganos , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Serrate-Jagged
15.
Acta Pharmacol Sin ; 36(12): 1480-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26592513

RESUMEN

AIM: The mitochondrial targeted 2C-type serine/threonine protein phosphatase (PP2Cm) is encoded by the gene PPM1K and is highly conserved among vertebrates. PP2Cm plays a critical role in branched-chain amino acid catabolism and regulates cell survival. Its expression is dynamically regulated by the nutrient environment and pathological stresses. However, little is known about the molecular mechanism underlying the regulation of PPM1K gene expression. In this study, we aimed to reveal how PPM1K expression is affected by miRNA-mediated post-transcriptional regulation. METHODS: Computational analysis based on conserved miRNA binding motifs was applied to predict the candidate miRNAs that potentially affect PPM1K expression. Dual-luciferase reporter assay was performed to verify the miRNAs' binding sites in the PPM1K gene and their influence on PPM1K 3'UTR activity. We further over-expressed the mimics of these miRNAs in human and mouse cells to examine whether miRNAs affected the mRNA level of PPM1K. RESULTS: Computational analysis identified numerous miRNAs potentially targeting PPM1K. Luciferase reporter assays demonstrated that the 3'UTR of PPM1K gene contained the recognition sites of miR-204 and miR-211. Overexpression of these miRNAs in human and mouse cells diminished the 3'UTR activity and the endogenous mRNA level of PPM1K. However, the miR-22 binding site was found only in human and not mouse PPM1K 3'UTR. Accordingly, PPM1K 3'UTR activity was suppressed by miR-22 overexpression in human but not mouse cells. CONCLUSION: These data suggest that different miRNAs contribute to the regulation of PP2Cm expression in a species-specific manner. miR-204 and miR-211 are efficient in both mouse and human cells, while miR-22 regulates PP2Cm expression only in human cells.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Fosfoproteínas Fosfatasas/genética , Regiones no Traducidas 3' , Animales , Línea Celular , Humanos , Ratones , Proteína Fosfatasa 2C , ARN Mensajero/genética
16.
Circ Res ; 110(2): 265-74, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22179058

RESUMEN

RATIONALE: Phosphorylation of ß(2)-adrenergic receptor (ß(2)AR) by a family of serine/threonine kinases known as G protein-coupled receptor kinase (GRK) and protein kinase A (PKA) is a critical determinant of cardiac function. Upregulation of G protein-coupled receptor kinase 2 (GRK2) is a well-established causal factor of heart failure, but the underlying mechanism is poorly understood. OBJECTIVE: We sought to determine the relative contribution of PKA- and GRK-mediated phosphorylation of ß(2)AR to the receptor coupling to G(i) signaling that attenuates cardiac reserve and contributes to the pathogenesis of heart failure in response to pressure overload. METHODS AND RESULTS: Overexpression of GRK2 led to a G(i)-dependent decrease of contractile response to ßAR stimulation in cultured mouse cardiomyocytes and in vivo. Importantly, cardiac-specific transgenic overexpression of a mutant ß(2)AR lacking PKA phosphorylation sites (PKA-TG) but not the wild-type ß(2)AR (WT-TG) or a mutant ß(2)AR lacking GRK sites (GRK-TG) led to exaggerated cardiac response to pressure overload, as manifested by markedly exacerbated cardiac maladaptive remodeling and failure and early mortality. Furthermore, inhibition of G(i) signaling with pertussis toxin restores cardiac function in heart failure associated with increased ß(2)AR to G(i) coupling induced by removing PKA phosphorylation of the receptor and in GRK2 transgenic mice, indicating that enhanced phosphorylation of ß(2)AR by GRK and resultant increase in G(i)-biased ß(2)AR signaling play an important role in the development of heart failure. CONCLUSIONS: Our data show that enhanced ß(2)AR phosphorylation by GRK, in addition to PKA, leads the receptor to G(i)-biased signaling, which, in turn, contributes to the pathogenesis of heart failure, marking G(i)-biased ß(2)AR signaling as a primary event linking upregulation of GRK to cardiac maladaptive remodeling, failure and cardiodepression.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Insuficiencia Cardíaca/enzimología , Miocitos Cardíacos/enzimología , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Agonistas Adrenérgicos beta/farmacología , Animales , Cardiomegalia/enzimología , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Contracción Miocárdica , Miocitos Cardíacos/efectos de los fármacos , Toxina del Pertussis/farmacología , Fosforilación , Receptores Adrenérgicos beta 2/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Regulación hacia Arriba , Función Ventricular Izquierda , Presión Ventricular , Remodelación Ventricular
17.
Mol Cell Proteomics ; 11(6): M111.014258, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22270000

RESUMEN

A fundamental question in biology is how genome-wide changes in gene expression are enacted in response to a finite stimulus. Recent studies have mapped changes in nucleosome localization, determined the binding preferences for individual transcription factors, and shown that the genome adopts a nonrandom structure in vivo. What remains unclear is how global changes in the proteins bound to DNA alter chromatin structure and gene expression. We have addressed this question in the mouse heart, a system in which global gene expression and massive phenotypic changes occur without cardiac cell division, making the mechanisms of chromatin remodeling centrally important. To determine factors controlling genomic plasticity, we used mass spectrometry to measure chromatin-associated proteins. We have characterized the abundance of 305 chromatin-associated proteins in normal cells and measured changes in 108 proteins that accompany the progression of heart disease. These studies were conducted on a high mass accuracy instrument and confirmed in multiple biological replicates, facilitating statistical analysis and allowing us to interrogate the data bioinformatically for modules of proteins involved in similar processes. Our studies reveal general principles for global shifts in chromatin accessibility: altered linker to core histone ratio; differing abundance of chromatin structural proteins; and reprogrammed histone post-translational modifications. Using small interfering RNA-mediated loss-of-function in isolated cells, we demonstrate that the non-histone chromatin structural protein HMGB2 (but not HMGB1) suppresses pathologic cell growth in vivo and controls a gene expression program responsible for hypertrophic cell growth. Our findings reveal the basis for alterations in chromatin structure necessary for genome-wide changes in gene expression. These studies have fundamental implications for understanding how global chromatin remodeling occurs with specificity and accuracy, demonstrating that isoform-specific alterations in chromatin structural proteins can impart these features.


Asunto(s)
Cardiomegalia/metabolismo , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteína HMGB2/metabolismo , Proteoma/metabolismo , Animales , Aumento de la Célula , Análisis por Conglomerados , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteína HMGB2/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Miocardio/metabolismo , Miocardio/patología , Interferencia de ARN
18.
Hum Mol Genet ; 20(3): 436-44, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21088111

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is caused by a mutant prelamin A, progerin, that terminates with a farnesylcysteine. HGPS knock-in mice (Lmna(HG/+)) develop severe progeria-like disease phenotypes. These phenotypes can be ameliorated with a protein farnesyltransferase inhibitor (FTI), suggesting that progerin's farnesyl lipid is important for disease pathogenesis and raising the possibility that FTIs could be useful for treating humans with HGPS. Subsequent studies showed that mice expressing non-farnesylated progerin (Lmna(nHG/+) mice, in which progerin's carboxyl-terminal -CSIM motif was changed to -SSIM) also develop severe progeria, raising doubts about whether any treatment targeting protein prenylation would be particularly effective. We suspected that those doubts might be premature and hypothesized that the persistent disease in Lmna(nHG/+) mice could be an unanticipated consequence of the cysteine-to-serine substitution that was used to eliminate farnesylation. To test this hypothesis, we generated a second knock-in allele yielding non-farnesylated progerin (Lmna(csmHG)) in which the carboxyl-terminal -CSIM motif was changed to -CSM. We then compared disease phenotypes in mice harboring the Lmna(nHG) or Lmna(csmHG) allele. As expected, Lmna(nHG/+) and Lmna(nHG/nHG) mice developed severe progeria-like disease phenotypes, including osteolytic lesions and rib fractures, osteoporosis, slow growth and reduced survival. In contrast, Lmna(csmHG/+) and Lmna(csmHG/csmHG) mice exhibited no bone disease and displayed entirely normal body weights and survival. The frequencies of misshapen cell nuclei were lower in Lmna(csmHG/+) and Lmna(csmHG/csmHG) fibroblasts. These studies show that the ability of non-farnesylated progerin to elicit disease depends on the carboxyl-terminal mutation used to eliminate protein prenylation.


Asunto(s)
Farnesiltransferasa/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progeria/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Sustitución de Aminoácidos , Animales , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Femenino , Técnicas de Sustitución del Gen , Imidazoles/farmacología , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Ratones , Mutación , Proteínas Nucleares/química , Fenotipo , Progeria/metabolismo , Progeria/patología , Progeria/fisiopatología , Precursores de Proteínas/química , Prenilación de Proteína
19.
Am J Physiol Heart Circ Physiol ; 305(11): H1624-38, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24077883

RESUMEN

Myocyte hypertrophy antecedent to heart failure involves changes in global gene expression, although the preceding mechanisms to coordinate DNA accessibility on a genomic scale are unknown. Chromatin-associated proteins alter chromatin structure by changing their association with DNA, thereby altering the gene expression profile. Little is known about the global changes in chromatin subproteomes that accompany heart failure, and the mechanisms by which these proteins alter chromatin structure. The present study tests the fundamental hypothesis that cardiac growth and plasticity in the setting of disease recapitulates conserved developmental chromatin remodeling events. We used quantitative proteomics to identify chromatin-associated proteins extracted via detergent and to quantify changes in their abundance during disease. Our study identified 321 proteins in this subproteome, demonstrating it to have modest conservation (37%) with that revealed using strong acid. Of these proteins, 176 exhibited altered expression during cardiac hypertrophy and failure; we conducted extensive functional characterization of one of these proteins, Nucleolin. Morpholino-based knockdown of nucleolin nearly abolished protein expression but surprisingly had little impact on gross morphological development. However, hearts of fish lacking Nucleolin displayed severe developmental impairment, abnormal chamber patterning and functional deficits, ostensibly due to defects in cardiac looping and myocyte differentiation. The mechanisms underlying these defects involve perturbed bone morphogenetic protein 4 expression, decreased rRNA transcription, and a shift to more heterochromatic chromatin. This study reports the quantitative analysis of a new chromatin subproteome in the normal and diseased mouse heart. Validation studies in the complementary model system of zebrafish examine the role of Nucleolin to orchestrate genomic reprogramming events shared between development and disease.


Asunto(s)
Cardiomegalia/metabolismo , Cromatina/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Proteómica , Proteínas de Unión al ARN/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Células Cultivadas , Ensamble y Desensamble de Cromatina , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Técnicas de Silenciamiento del Gen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Miocitos Cardíacos/patología , Fosfoproteínas/genética , Proteómica/métodos , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/genética , Ratas , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Pez Cebra , Proteínas de Pez Cebra/genética , Nucleolina
20.
Circ Res ; 109(12): 1332-41, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22034492

RESUMEN

RATIONALE: Accurate and comprehensive de novo transcriptome profiling in heart is a central issue to better understand cardiac physiology and diseases. Although significant progress has been made in genome-wide profiling for quantitative changes in cardiac gene expression, current knowledge offers limited insights to the total complexity in cardiac transcriptome at individual exon level. OBJECTIVE: To develop more robust bioinformatic approaches to analyze high-throughput RNA sequencing (RNA-Seq) data, with the focus on the investigation of transcriptome complexity at individual exon and transcript levels. METHODS AND RESULTS: In addition to overall gene expression analysis, the methods developed in this study were used to analyze RNA-Seq data with respect to individual transcript isoforms, novel spliced exons, novel alternative terminal exons, novel transcript clusters (ie, novel genes), and long noncoding RNA genes. We applied these approaches to RNA-Seq data obtained from mouse hearts after pressure-overload-induced by transaortic constriction. Based on experimental validations, analyses of the features of the identified exons/transcripts, and expression analyses including previously published RNA-Seq data, we demonstrate that the methods are highly effective in detecting and quantifying individual exons and transcripts. Novel insights inferred from the examined aspects of the cardiac transcriptome open ways to further experimental investigations. CONCLUSIONS: Our work provided a comprehensive set of methods to analyze mouse cardiac transcriptome complexity at individual exon and transcript levels. Applications of the methods may infer important new insights to gene regulation in normal and disease hearts in terms of exon utilization and potential involvement of novel components of cardiac transcriptome.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Corazón/fisiología , Análisis de Secuencia de ARN/métodos , Animales , Modelos Animales de Enfermedad , Exones/genética , Exones/fisiología , Regulación de la Expresión Génica/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Transcripción Genética/genética , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA