Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ESC Heart Fail ; 9(2): 1463-1470, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35118823

RESUMEN

AIMS: Heart failure (HF) represents a clinical syndrome resulting from different aetiologies and degrees of heart diseases. Among these, a key role is played by primary heart muscle disease (cardiomyopathies), which are the combination of multifactorial environmental insults in the presence or absence of a known genetic predisposition. The aim of the Maastricht Cardiomyopathy registry (mCMP-registry; NCT04976348) is to improve (early) diagnosis, risk stratification, and management of cardiomyopathy phenotypes beyond the limits of left ventricular ejection fraction (LVEF). METHODS AND RESULTS: The mCMP-registry is an investigator-initiated prospective registry including patient characteristics, diagnostic measurements performed as part of routine clinical care, treatment information, sequential biobanking, quality of life and economic impact assessment, and regular follow-up. All subjects aged ≥16 years referred to the cardiology department of the Maastricht University Medical Center (MUMC+) for HF-like symptoms or cardiac screening for cardiomyopathies are eligible for inclusion, irrespective of phenotype or underlying causes. Informed consented subjects will be followed up for 15 years. Two central approaches will be used to answer the research questions related to the aims of this registry: (i) a data-driven approach to predict clinical outcome and response to therapy and to identify clusters of patients who share underlying pathophysiological processes; and (ii) a hypothesis-driven approach in which clinical parameters are tested for their (incremental) diagnostic, prognostic, or therapeutic value. The study allows other centres to easily join this initiative, which will further boost research within this field. CONCLUSIONS: The broad inclusion criteria, systematic routine clinical care data-collection, extensive study-related data-collection, sequential biobanking, and multi-disciplinary approach gives the mCMP-registry a unique opportunity to improve diagnosis, risk stratification, and management of HF and (early) cardiomyopathy phenotypes beyond the LVEF limits.


Asunto(s)
Cardiomiopatías , Calidad de Vida , Bancos de Muestras Biológicas , Cardiomiopatías/diagnóstico , Cardiomiopatías/epidemiología , Cardiomiopatías/etiología , Humanos , Sistema de Registros , Medición de Riesgo , Volumen Sistólico/fisiología , Función Ventricular Izquierda/fisiología
2.
Sci Rep ; 9(1): 6055, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988323

RESUMEN

Heart failure (HF) is the leading cause of death in the Western world. Pathophysiological processes underlying HF development, including cardiac hypertrophy, fibrosis and inflammation, are controlled by specific microRNAs (miRNAs). Whereas most studies investigate miRNA function in one particular cardiac cell type, their multicellular function is poorly investigated. The present study probed 194 miRNAs -differentially expressed in cardiac inflammatory disease - for regulating cardiomyocyte size, cardiac fibroblasts collagen content, and macrophage polarization. Of the tested miRNAs, 13%, 26%, and 41% modulated cardiomyocyte size, fibroblast collagen production, and macrophage polarization, respectively. Seventeen miRNAs affected all three cellular processes, including miRNAs with established (miR-210) and unknown roles in cardiac pathophysiology (miR-145-3p). These miRNAs with a multi-cellular function commonly target various genes. In-depth analysis in vitro of previously unstudied miRNAs revealed that the observed phenotypical alterations concurred with changes in transcript and protein levels of hypertrophy-, fibrosis- and inflammation-related genes. MiR-145-3p and miR-891a-3p were identified to regulate the fibrotic response, whereas miR-223-3p, miR-486-3p, and miR-488-5p modulated macrophage activation and polarisation. In conclusion, miRNAs are multi-cellular regulators of different cellular processes underlying cardiac disease. We identified previously undescribed roles of miRNAs in hypertrophy, fibrosis, and inflammation, and attribute new cellular effects to various well-known miRNAs.


Asunto(s)
Cardiomegalia/patología , Insuficiencia Cardíaca/genética , MicroARNs/metabolismo , Miocarditis/inmunología , Miocardio/patología , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Cardiomegalia/inmunología , Células Cultivadas , Fibroblastos , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/patología , Humanos , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos , Ratones , Miocarditis/genética , Miocarditis/patología , Miocardio/citología , Miocardio/inmunología , Miocitos Cardíacos , Cultivo Primario de Células , Ratas
3.
Mol Ther Nucleic Acids ; 14: 424-437, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30731323

RESUMEN

MicroRNA-103/107 regulate systemic glucose metabolism and insulin sensitivity. For this reason, inhibitory strategies for these microRNAs are currently being tested in clinical trials. Given the high metabolic demands of the heart and the abundant cardiac expression of miR-103/107, we questioned whether antagomiR-mediated inhibition of miR-103/107 in C57BL/6J mice impacts on cardiac function. Notably, fractional shortening decreased after 6 weeks of antagomiR-103 and -107 treatment. This was paralleled by a prolonged systolic radial and circumferential time to peak and by a decreased global strain rate. Histology and electron microscopy showed reduced cardiomyocyte area and decreased mitochondrial volume and mitochondrial cristae density following antagomiR-103 and -107. In line, antagomiR-103 and -107 treatment decreased mitochondrial OXPHOS complexes' protein levels compared to scrambled, as assessed by mass spectrometry-based label-free quantitative proteomics. MiR-103/107 inhibition in primary cardiomyocytes did not affect glycolysis rates, but it decreased mitochondrial reserve capacity, reduced mitochondrial membrane potential, and altered mitochondrial network morphology, as assessed by live-cell imaging. Our data indicate that antagomiR-103 and -107 decrease cardiac function, cardiomyocyte size, and mitochondrial oxidative capacity in the absence of pathological stimuli. These data raise concern about the possible cardiac implications of the systemic use of antagomiR-103 and -107 in the clinical setting, and careful cardiac phenotyping within ongoing trials is highly recommended.

4.
Matrix Biol ; 74: 21-34, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29730504

RESUMEN

Myocardial damage as a consequence of cardiotropic viruses leads to a broad variety of clinical presentations and is still a complicated condition to diagnose and treat. Whereas the extracellular matrix protein Secreted Protein Acidic and Rich in Cysteine or SPARC has been implicated in hypertensive and ischemic heart disease by modulating collagen production and cross-linking, its role in cardiac inflammation and endothelial function is yet unknown. Absence of SPARC in mice resulted in increased cardiac inflammation and mortality, and reduced cardiac systolic function upon coxsackievirus-B3 induced myocarditis. Intra-vital microscopic imaging of the microvasculature of the cremaster muscle combined with electron microscopic imaging of the microvasculature of the cardiac muscle uncovered the significance of SPARC in maintaining endothelial glycocalyx integrity and subsequent barrier properties to stop inflammation. Moreover, systemic administration of recombinant SPARC restored the endothelial glycocalyx and consequently reversed the increase in inflammation and mortality observed in SPARC KO mice in response to viral exposure. Reducing the glycocalyx in vivo by systemic administration of hyaluronidase, an enzyme that degrades the endothelial glycocalyx, mimicked the barrier defects found in SPARC KO mice, which could be restored by subsequent administration of recombinant SPARC. In conclusion, the secreted glycoprotein SPARC protects against adverse cardiac inflammation and mortality by improving the glycocalyx function and resulting endothelial barrier function during viral myocarditis.


Asunto(s)
Infecciones por Coxsackievirus/metabolismo , Hialuronoglucosaminidasa/farmacología , Miocarditis/virología , Osteonectina/genética , Osteonectina/metabolismo , Músculos Abdominales/irrigación sanguínea , Músculos Abdominales/virología , Animales , Infecciones por Coxsackievirus/genética , Modelos Animales de Enfermedad , Enterovirus Humano B/patogenicidad , Técnicas de Inactivación de Genes , Glicocálix/química , Masculino , Ratones , Microscopía Electrónica , Miocarditis/genética , Miocarditis/metabolismo
5.
Matrix Biol ; 66: 110-124, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28958774

RESUMEN

The small leucine-rich proteoglycan osteoglycin has been implicated in matrix homeostasis in different organs, including the ischemic heart. However, whether osteoglycin modulates cardiac hypertrophy, fibrosis or inflammation in hypertensive heart disease and during aging remains unknown. Angiotensin-II-induced pressure overload increases cardiac osteoglycin expression, concomitant with the onset of inflammation and extracellular matrix deposition. Interestingly aging led to decreased cardiac levels of osteoglycin, yet absence of osteoglycin did not affect organ structure or cardiac function up to the age of 18months. However, Angiotensin-II infusion in combination with aging resulted in exaggerated cardiac fibrosis and inflammation in the osteoglycin null mice as compared to wild-type mice, resulting in increased diastolic dysfunction as determined by magnetic resonance imaging. In vitro, stimulation of bone marrow derived macrophages from osteoglycin null mice with Angiotensin-II resulted in significantly higher levels of ICAM-1 as well as pro-inflammatory cytokines and chemokines IL-1ß and MCP-1 as compared to WT cells. Further, stimulation of human cardiac fibroblasts with osteoglycin reduced cell proliferation and inhibited TGF-ß induced collagen gene expression. In mouse cardiac tissue, osteoglycin expression inversely correlated with TGF-ß expression and in cardiac biopsies of aortic stenosis patients, osteoglycin expression is significantly higher than in control biopsies. Interestingly, osteoglycin levels were higher in patients with less severe myocardial fibrosis and overall in the aortic stenosis patients osteoglycin levels negatively correlated with collagen content in the myocardium. In conclusion, osteoglycin expression is increased in the heart in response to pressure overload and its absence results in increased cardiac inflammation and fibrosis resulting in increased diastolic dysfunction.


Asunto(s)
Angiotensina II/farmacología , Estenosis de la Válvula Aórtica/metabolismo , Hipertensión/complicaciones , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Miocardio/patología , Envejecimiento , Animales , Estenosis de la Válvula Aórtica/genética , Células Cultivadas , Quimiocina CCL2/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Fibrosis , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones
6.
Hypertension ; 71(2): 280-288, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29255073

RESUMEN

Pressure overload causes cardiac fibroblast activation and transdifferentiation, leading to increased interstitial fibrosis formation and subsequently myocardial stiffness, diastolic and systolic dysfunction, and eventually heart failure. A better understanding of the molecular mechanisms underlying pressure overload-induced cardiac remodeling and fibrosis will have implications for heart failure treatment strategies. The microRNA (miRNA)-221/222 family, consisting of miR-221-3p and miR-222-3p, is differentially regulated in mouse and human cardiac pathology and inversely associated with kidney and liver fibrosis. We investigated the role of this miRNA family during pressure overload-induced cardiac remodeling. In myocardial biopsies of patients with severe fibrosis and dilated cardiomyopathy or aortic stenosis, we found significantly lower miRNA-221/222 levels as compared to matched patients with nonsevere fibrosis. In addition, miRNA-221/222 levels in aortic stenosis patients correlated negatively with the extent of myocardial fibrosis and with left ventricular stiffness. Inhibition of both miRNAs during AngII (angiotensin II)-mediated pressure overload in mice led to increased fibrosis and aggravated left ventricular dilation and dysfunction. In rat cardiac fibroblasts, inhibition of miRNA-221/222 derepressed TGF-ß (transforming growth factor-ß)-mediated profibrotic SMAD2 (mothers against decapentaplegic homolog 2) signaling and downstream gene expression, whereas overexpression of both miRNAs blunted TGF-ß-induced profibrotic signaling. We found that the miRNA-221/222 family may target several genes involved in TGF-ß signaling, including JNK1 (c-Jun N-terminal kinase 1), TGF-ß receptor 1 and TGF-ß receptor 2, and ETS-1 (ETS proto-oncogene 1). Our findings show that heart failure-associated downregulation of the miRNA-221/222 family enables profibrotic signaling in the pressure-overloaded heart.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Animales , Estenosis de la Válvula Aórtica/complicaciones , Estenosis de la Válvula Aórtica/metabolismo , Cardiomiopatías/metabolismo , Fibroblastos/metabolismo , Fibrosis/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Proto-Oncogenes Mas , Ratas , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
7.
Circulation ; 136(8): 747-761, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28611091

RESUMEN

BACKGROUND: Cardiovascular diseases remain the predominant cause of death worldwide, with the prevalence of heart failure continuing to increase. Despite increased knowledge of the metabolic alterations that occur in heart failure, novel therapies to treat the observed metabolic disturbances are still lacking. METHODS: Mice were subjected to pressure overload by means of angiotensin-II infusion or transversal aortic constriction. MicroRNA-146a was either genetically or pharmacologically knocked out or genetically overexpressed in cardiomyocytes. Furthermore, overexpression of dihydrolipoyl succinyltransferase (DLST) in the murine heart was performed by means of an adeno-associated virus. RESULTS: MicroRNA-146a was upregulated in whole heart tissue in multiple murine pressure overload models. Also, microRNA-146a levels were moderately increased in left ventricular biopsies of patients with aortic stenosis. Overexpression of microRNA-146a in cardiomyocytes provoked cardiac hypertrophy and left ventricular dysfunction in vivo, whereas genetic knockdown or pharmacological blockade of microRNA-146a blunted the hypertrophic response and attenuated cardiac dysfunction in vivo. Mechanistically, microRNA-146a reduced its target DLST-the E2 subcomponent of the α-ketoglutarate dehydrogenase complex, a rate-controlling tricarboxylic acid cycle enzyme. DLST protein levels significantly decreased on pressure overload in wild-type mice, paralleling a decreased oxidative metabolism, whereas DLST protein levels and hence oxidative metabolism were partially maintained in microRNA-146a knockout mice. Moreover, overexpression of DLST in wild-type mice protected against cardiac hypertrophy and dysfunction in vivo. CONCLUSIONS: Altogether we show that the microRNA-146a and its target DLST are important metabolic players in left ventricular dysfunction.


Asunto(s)
Aciltransferasas/biosíntesis , Cardiomegalia/metabolismo , Regulación Enzimológica de la Expresión Génica , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , Disfunción Ventricular Izquierda/metabolismo , Aciltransferasas/genética , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Cardiomegalia/prevención & control , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Ratas , Ratas Endogámicas Lew , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/prevención & control
8.
Eur Heart J ; 36(42): 2909-19, 2015 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-26206211

RESUMEN

AIMS: Viral myocarditis (VM) is an important cause of heart failure and sudden cardiac death in young healthy adults; it is also an aetiological precursor of dilated cardiomyopathy. We explored the role of the miR-221/-222 family that is up-regulated in VM. METHODS AND RESULTS: Here, we show that microRNA-221 (miR-221) and miR-222 levels are significantly elevated during acute VM caused by Coxsackievirus B3 (CVB3). Both miRs are expressed by different cardiac cells and by infiltrating inflammatory cells, but their up-regulation upon myocarditis is mostly exclusive for the cardiomyocyte. Systemic inhibition of miR-221/-222 in mice increased cardiac viral load, prolonged the viraemic state, and strongly aggravated cardiac injury and inflammation. Similarly, in vitro, overexpression of miR-221 and miR-222 inhibited enteroviral replication, whereas knockdown of this miR-cluster augmented viral replication. We identified and confirmed a number of miR-221/-222 targets that co-orchestrate the increased viral replication and inflammation, including ETS1/2, IRF2, BCL2L11, TOX, BMF, and CXCL12. In vitro inhibition of IRF2, TOX, or CXCL12 in cardiomyocytes significantly dampened their inflammatory response to CVB3 infection, confirming the functionality of these targets in VM and highlighting the importance of miR-221/-222 as regulators of the cardiac response to VM. CONCLUSIONS: The miR-221/-222 cluster orchestrates the antiviral and inflammatory immune response to viral infection of the heart. Its inhibition increases viral load, inflammation, and overall cardiac injury upon VM.


Asunto(s)
Infecciones por Coxsackievirus/virología , MicroARNs/fisiología , Miocarditis/virología , Animales , Infecciones por Coxsackievirus/inmunología , Humanos , Inmunidad Celular/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Miocarditis/inmunología , Miocitos Cardíacos/inmunología , Linfocitos T/inmunología , Regulación hacia Arriba , Carga Viral/inmunología , Replicación Viral/inmunología
9.
Circ Res ; 116(3): 425-36, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25520363

RESUMEN

RATIONALE: To maintain cardiac mechanical and structural integrity after an ischemic insult, profound alterations occur within the extracellular matrix. Osteoglycin is a small leucine-rich proteoglycan previously described as a marker of cardiac hypertrophy. OBJECTIVE: To establish whether osteoglycin may play a role in cardiac integrity and function after myocardial infarction (MI). METHODS AND RESULTS: Osteoglycin expression is associated with collagen deposition and scar formation in mouse and human MI. Absence of osteoglycin in mice resulted in significantly increased rupture-related mortality with tissue disruption, intramyocardial bleeding, and increased cardiac dysfunction, despite equal infarct sizes. Surviving osteoglycin null mice had greater infarct expansion in comparison with wild-type mice because of impaired collagen fibrillogenesis and maturation in the infarcts as revealed by electron microscopy and collagen polarization. Absence of osteoglycin did not affect cardiomyocyte hypertrophy in the remodeling remote myocardium. In cultured fibroblasts, osteoglycin knockdown or supplementation did not alter transforming growth factor-ß signaling. Adenoviral overexpression of osteoglycin in wild-type mice significantly improved collagen quality, thereby blunting cardiac dilatation and dysfunction after MI. In osteoglycin null mice, adenoviral overexpression of osteoglycin was unable to prevent rupture-related mortality because of insufficiently restoring osteoglycin protein levels in the heart. Finally, circulating osteoglycin levels in patients with heart failure were significantly increased in the patients with a previous history of MI compared with those with nonischemic heart failure and correlated with survival, left ventricular volumes, and other markers of fibrosis. CONCLUSIONS: Increased osteoglycin expression in the infarct scar promotes proper collagen maturation and protects against cardiac disruption and adverse remodeling after MI. In human heart failure, osteoglycin is a promising biomarker for ischemic heart failure.


Asunto(s)
Cardiomegalia/metabolismo , Colágeno/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Infarto del Miocardio/metabolismo , Animales , Cardiomegalia/patología , Cicatriz/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Linfotoxina-alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Ratas , Ratas Endogámicas Lew , Remodelación Ventricular
10.
Circulation ; 128(13): 1420-32, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-23956210

RESUMEN

BACKGROUND: Cardiac hypertrophy and subsequent heart failure triggered by chronic hypertension represent major challenges for cardiovascular research. Beyond neurohormonal and myocyte signaling pathways, growing evidence suggests inflammatory signaling pathways as therapeutically targetable contributors to this process. We recently reported that microRNA-155 is a key mediator of cardiac inflammation and injury in infectious myocarditis. Here, we investigated the impact of microRNA-155 manipulation in hypertensive heart disease. METHODS AND RESULTS: Genetic loss or pharmacological inhibition of the leukocyte-expressed microRNA-155 in mice markedly reduced cardiac inflammation, hypertrophy, and dysfunction on pressure overload. These alterations were macrophage dependent because in vivo cardiomyocyte-specific microRNA-155 manipulation did not affect cardiac hypertrophy or dysfunction, whereas bone marrow transplantation from wild-type mice into microRNA-155 knockout animals rescued the hypertrophic response of the cardiomyocytes and vice versa. In vitro, media from microRNA-155 knockout macrophages blocked the hypertrophic growth of stimulated cardiomyocytes, confirming that macrophages influence myocyte growth in a microRNA-155-dependent paracrine manner. These effects were at least partly mediated by the direct microRNA-155 target suppressor of cytokine signaling 1 (Socs1) because Socs1 knockdown in microRNA-155 knockout macrophages largely restored their hypertrophy-stimulating potency. CONCLUSIONS: Our findings reveal that microRNA-155 expression in macrophages promotes cardiac inflammation, hypertrophy, and failure in response to pressure overload. These data support the causative significance of inflammatory signaling in hypertrophic heart disease and demonstrate the feasibility of therapeutic microRNA targeting of inflammation in heart failure.


Asunto(s)
Cardiomegalia/patología , Insuficiencia Cardíaca/patología , Macrófagos/patología , MicroARNs/genética , Miocitos Cardíacos/patología , Animales , Cardiomegalia/genética , Células Cultivadas , Insuficiencia Cardíaca/genética , Humanos , Inflamación/genética , Inflamación/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Ratas
11.
Circ Res ; 111(4): 415-25, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22715471

RESUMEN

RATIONALE: Viral myocarditis results from an adverse immune response to cardiotropic viruses, which causes irreversible myocyte destruction and heart failure in previously healthy people. The involvement of microRNAs and their usefulness as therapeutic targets in this process are unknown. OBJECTIVE: To identify microRNAs involved in viral myocarditis pathogenesis and susceptibility. METHODS AND RESULTS: Cardiac microRNAs were profiled in both human myocarditis and in Coxsackievirus B3-injected mice, comparing myocarditis-susceptible with nonsusceptible mouse strains longitudinally. MicroRNA responses diverged depending on the susceptibility to myocarditis after viral infection in mice. MicroRNA-155, -146b, and -21 were consistently and strongly upregulated during acute myocarditis in both humans and susceptible mice. We found that microRNA-155 expression during myocarditis was localized primarily in infiltrating macrophages and T lymphocytes. Inhibition of microRNA-155 by a systemically delivered LNA-anti-miR attenuated cardiac infiltration by monocyte-macrophages, decreased T lymphocyte activation, and reduced myocardial damage during acute myocarditis in mice. These changes were accompanied by the derepression of the direct microRNA-155 target PU.1 in cardiac inflammatory cells. Beyond the acute phase, microRNA-155 inhibition reduced mortality and improved cardiac function during 7 weeks of follow-up. CONCLUSIONS: Our data show that cardiac microRNA dysregulation is a characteristic of both human and mouse viral myocarditis. The inflammatory microRNA-155 is upregulated during acute myocarditis, contributes to the adverse inflammatory response to viral infection of the heart, and is a potential therapeutic target for viral myocarditis.


Asunto(s)
Infecciones por Coxsackievirus/genética , Perfilación de la Expresión Génica , MicroARNs/metabolismo , Miocarditis/genética , Miocardio/metabolismo , Animales , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/fisiopatología , Infecciones por Coxsackievirus/terapia , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Enterovirus Humano B/patogenicidad , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Activación de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Miocarditis/inmunología , Miocarditis/patología , Miocarditis/fisiopatología , Miocarditis/terapia , Miocarditis/virología , Miocardio/inmunología , Miocardio/patología , Oligonucleótidos/administración & dosificación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Factores de Tiempo
12.
Aging Cell ; 10(5): 769-79, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21501375

RESUMEN

To understand the process of cardiac aging, it is of crucial importance to gain insight into the age-related changes in gene expression in the senescent failing heart. Age-related cardiac remodeling is known to be accompanied by changes in extracellular matrix (ECM) gene and protein levels. Small noncoding microRNAs regulate gene expression in cardiac development and disease and have been implicated in the aging process and in the regulation of ECM proteins. However, their role in age-related cardiac remodeling and heart failure is unknown. In this study, we investigated the aging-associated microRNA cluster 17-92, which targets the ECM proteins connective tissue growth factor (CTGF) and thrombospondin-1 (TSP-1). We employed aged mice with a failure-resistant (C57Bl6) and failure-prone (C57Bl6 × 129Sv) genetic background and extrapolated our findings to human age-associated heart failure. In aging-associated heart failure, we linked an aging-induced increase in the ECM proteins CTGF and TSP-1 to a decreased expression of their targeting microRNAs 18a, 19a, and 19b, all members of the miR-17-92 cluster. Failure-resistant mice showed an opposite expression pattern for both the ECM proteins and the microRNAs. We showed that these expression changes are specific for cardiomyocytes and are absent in cardiac fibroblasts. In cardiomyocytes, modulation of miR-18/19 changes the levels of ECM proteins CTGF and TSP-1 and collagens type 1 and 3. Together, our data support a role for cardiomyocyte-derived miR-18/19 during cardiac aging, in the fine-tuning of cardiac ECM protein levels. During aging, decreased miR-18/19 and increased CTGF and TSP-1 levels identify the failure-prone heart.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Insuficiencia Cardíaca/patología , MicroARNs/metabolismo , Trombospondina 1/metabolismo , Adulto , Anciano , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Biopsia , Colágeno/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Persona de Mediana Edad , Familia de Multigenes , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Ratas , Ratas Endogámicas Lew , Trombospondina 1/genética
13.
J Biol Chem ; 285(35): 27449-27456, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20566642

RESUMEN

Pathological forms of left ventricular hypertrophy (LVH) often progress to heart failure. Specific transcription factors have been identified that activate the gene program to induce pathological forms of LVH. It is likely that apart from activating transcriptional inducers of LVH, constitutive transcriptional repressors need to be removed during the development of cardiac hypertrophy. Here, we report that the constitutive presence of Krüppel-like factor 15 (KLF15) is lost in pathological hypertrophy and that this loss precedes progression toward heart failure. We show that transforming growth factor-beta-mediated activation of p38 MAPK is necessary and sufficient to decrease KLF15 expression. We further show that KLF15 robustly inhibits myocardin, a potent transcriptional activator. Loss of KLF15 during pathological LVH relieves the inhibitory effects on myocardin and stimulates the expression of serum response factor target genes, such as atrial natriuretic factor. This uncovers a novel mechanism where activated p38 MAPK decreases KLF15, an important constitutive transcriptional repressor whose removal seems a vital step to allow the induction of pathological LVH.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Hipertrofia Ventricular Izquierda/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Células COS , Chlorocebus aethiops , Activación Enzimática , Ratones , Ratas , Ratas Endogámicas Lew , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Hypertension ; 55(2): 249-56, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20048198

RESUMEN

Syndecan-1 (Synd1) is a transmembrane heparan sulfate proteoglycan that functions as a coreceptor for various growth factors and modulates signal transduction. The present study investigated whether Synd1, by affecting growth factor signaling, may play a role in hypertension-induced cardiac fibrosis and dysfunction. Expression of Synd1 was increased significantly in mouse hearts with angiotensin II-induced hypertension, which was spatially related to cardiac fibrosis. Angiotensin II significantly impaired fractional shortening and induced cardiac fibrosis in wild-type mice, whereas these effects were blunted in Synd1-null mice. Angiotensin II significantly increased cardiac expression of connective tissue growth factor and collagen type I and III in wild-type mice, which was blunted in Synd1-null mice. These findings were confirmed in vitro, where angiotensin II induced the expression of both connective tissue growth factor and collagen I in fibroblasts. The absence of Synd1 in either Synd1-null fibroblasts, after knockdown of Synd1 by short hairpin RNA, or after inhibition of heparan sulfates by protamine attenuated this increase, which was associated with reduced phosphorylation of Smad2. In conclusion, loss of Synd1 reduces cardiac fibrosis and dysfunction during angiotensin II-induced hypertension.


Asunto(s)
Angiotensina II/farmacología , Miocardio/patología , Proteína Smad2/metabolismo , Sindecano-1/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Fibrosis/patología , Regulación de la Expresión Génica , Hipertensión/inducido químicamente , Hipertensión/complicaciones , Masculino , Ratones , Ratones Endogámicos BALB C , Probabilidad , ARN Mensajero/análisis , Distribución Aleatoria , Proteína Smad2/efectos de los fármacos , Proteína Smad2/genética , Sindecano-1/genética
15.
Circ Res ; 104(2): 170-8, 6p following 178, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19096030

RESUMEN

The myocardium of the failing heart undergoes a number of structural alterations, most notably hypertrophy of cardiac myocytes and an increase in extracellular matrix proteins, often seen as primary fibrosis. Connective tissue growth factor (CTGF) is a key molecule in the process of fibrosis and therefore seems an attractive therapeutic target. Regulation of CTGF expression at the promoter level has been studied extensively, but it is unknown how CTGF transcripts are regulated at the posttranscriptional level. Here we provide several lines of evidence to show that CTGF is importantly regulated by 2 major cardiac microRNAs (miRNAs), miR-133 and miR-30. First, the expression of both miRNAs was inversely related to the amount of CTGF in 2 rodent models of heart disease and in human pathological left ventricular hypertrophy. Second, in cultured cardiomyocytes and fibroblasts, knockdown of these miRNAs increased CTGF levels. Third, overexpression of miR-133 or miR-30c decreased CTGF levels, which was accompanied by decreased production of collagens. Fourth, we show that CTGF is a direct target of these miRNAs, because they directly interact with the 3' untranslated region of CTGF. Taken together, our results indicate that miR-133 and miR-30 importantly limit the production of CTGF. We also provide evidence that the decrease of these 2 miRNAs in pathological left ventricular hypertrophy allows CTGF levels to increase, which contributes to collagen synthesis. In conclusion, our results show that both miR-133 and miR-30 directly downregulate CTGF, a key profibrotic protein, and thereby establish an important role for these miRNAs in the control of structural changes in the extracellular matrix of the myocardium.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/metabolismo , Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Procesamiento Postranscripcional del ARN , Remodelación Ventricular , Regiones no Traducidas 3' , Animales , Animales Recién Nacidos , Secuencia de Bases , Células Cultivadas , Biología Computacional , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Femenino , Fibrosis , Técnicas de Silenciamiento del Gen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Miocardio/patología , Filogenia , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Renina/genética , Renina/metabolismo , Regulación hacia Arriba , Remodelación Ventricular/genética
16.
J Exp Med ; 206(1): 113-23, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19103879

RESUMEN

The matricellular protein SPARC (secreted protein, acidic and rich in cysteine, also known as osteonectin) mediates cell-matrix interactions during wound healing and regulates the production and/or assembly of the extracellular matrix (ECM). This study investigated whether SPARC functions in infarct healing and ECM maturation after myocardial infarction (MI). In comparison with wild-type (WT) mice, animals with a targeted inactivation of SPARC exhibited a fourfold increase in mortality that resulted from an increased incidence of cardiac rupture and failure after MI. SPARC-null infarcts had a disorganized granulation tissue and immature collagenous ECM. In contrast, adenoviral overexpression of SPARC in WT mice improved the collagen maturation and prevented cardiac dilatation and dysfunction after MI. In cardiac fibroblasts in vitro, reduction of SPARC by short hairpin RNA attenuated transforming growth factor beta (TGF)-mediated increase of Smad2 phosphorylation, whereas addition of recombinant SPARC increased Smad2 phosphorylation concordant with increased Smad2 phosphorylation in SPARC-treated mice. Importantly, infusion of TGF-beta rescued cardiac rupture in SPARC-null mice but did not significantly alter infarct healing in WT mice. These findings indicate that local production of SPARC is essential for maintenance of the integrity of cardiac ECM after MI. The protective effects of SPARC emphasize the potential therapeutic applications of this protein to prevent cardiac dilatation and dysfunction after MI.


Asunto(s)
Rotura Cardíaca Posinfarto/metabolismo , Infarto del Miocardio/metabolismo , Osteonectina/deficiencia , Animales , Proteínas de Unión al Calcio/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Tejido de Granulación/efectos de los fármacos , Tejido de Granulación/metabolismo , Tejido de Granulación/patología , Corazón/fisiopatología , Rotura Cardíaca Posinfarto/fisiopatología , Rotura Cardíaca Posinfarto/prevención & control , Hemodinámica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Osteonectina/genética , Osteonectina/fisiología , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteína Smad2/genética , Análisis de Supervivencia , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/fisiología , Factor de Crecimiento Transformador beta/uso terapéutico
17.
J Exp Med ; 204(5): 1227-35, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17485520

RESUMEN

The intercalated disc (ID) of cardiac myocytes is emerging as a crucial structure in the heart. Loss of ID proteins like N-cadherin causes lethal cardiac abnormalities, and mutations in ID proteins cause human cardiomyopathy. A comprehensive screen for novel mechanisms in failing hearts demonstrated that expression of the lysosomal integral membrane protein 2 (LIMP-2) is increased in cardiac hypertrophy and heart failure in both rat and human myocardium. Complete loss of LIMP-2 in genetically engineered mice did not affect cardiac development; however, these LIMP-2 null mice failed to mount a hypertrophic response to increased blood pressure but developed cardiomyopathy. Disturbed cadherin localization in these hearts suggested that LIMP-2 has important functions outside lysosomes. Indeed, we also find LIMP-2 in the ID, where it associates with cadherin. RNAi-mediated knockdown of LIMP-2 decreases the binding of phosphorylated beta-catenin to cadherin, whereas overexpression of LIMP-2 has the opposite effect. Collectively, our data show that LIMP-2 is crucial to mount the adaptive hypertrophic response to cardiac loading. We demonstrate a novel role for LIMP-2 as an important mediator of the ID.


Asunto(s)
Antígenos CD36/metabolismo , Cardiomiopatía Dilatada/metabolismo , Hipertensión/complicaciones , Proteínas de Membrana de los Lisosomas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Estenosis de la Válvula Aórtica/metabolismo , Antígenos CD36/genética , Cadherinas/metabolismo , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/genética , Cartilla de ADN , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas de Membrana de los Lisosomas/genética , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , beta Catenina/metabolismo
18.
Hypertension ; 47(3): 467-74, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16432052

RESUMEN

Imatinib specifically inhibits receptor tyrosine kinase signaling and is clinically used to treat leukemia. Receptor tyrosine kinases not only mediate tumor growth but also initiate adverse signaling in heart failure. We investigated whether imatinib, by inhibiting the platelet-derived growth factor receptor-beta (PDGFRbeta), prevents cardiac and renal damage in TGR(mRen2)27 (Ren2) rats. Eight-week-old male homozygous Ren2 and Sprague Dawley rats were treated either with imatinib (30 mg/kg; STI-571) or placebo for 8 weeks (Ren2 n=12 for each group; Sprague Dawley n=6 for each group). Imatinib did not affect blood pressure or left ventricular (LV) hypertrophy in both groups. Imatinib attenuated the decline in fractional shortening (imatinib versus Ren2 placebo 45+/-4.5% versus 32+/-3%; n=7-11; P<0.05) and in diastolic function in Ren2 rats (baseline diastolic dP/dt corrected for systolic blood pressure Ren2 imatinib versus Ren2 placebo 38.6+/-0.67 versus 35.3+/-0.41 [1 . s(-1)]; n=7-11; P<0.05). This was associated with decreased cardiac fibrosis and decreased activation of PDGFRbeta and extracellular signal-regulated kinase 1/2. Renal microvascular hypertrophy and perivascular fibrosis in Ren2 rats were significantly decreased by imatinib. In vitro, imatinib blocked angiotensin II-induced activation of the PDGFRbeta and significantly decreased fibroblast proliferation and collagen production. In conclusion, imatinib did not affect LV hypertrophy but attenuated the decline in cardiac function and reduced renal microvascular damage associated with reduced activation of the PDGFRbeta. The simultaneous improvement in both heart and kidneys suggests that inhibition of the PDGFRbeta has broad protective effects that may provide novel avenues for a blood pressure-independent protection against end-organ damage.


Asunto(s)
Cardiopatías/prevención & control , Homocigoto , Hipertensión/genética , Enfermedades Renales/prevención & control , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Renina/genética , Animales , Animales Modificados Genéticamente , Arteriolas/efectos de los fármacos , Arteriolas/patología , Benzamidas , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Fibrosis , Hemodinámica/efectos de los fármacos , Hipertensión/complicaciones , Mesilato de Imatinib , Riñón/irrigación sanguínea , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
19.
Circ Res ; 95(5): 515-22, 2004 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-15284191

RESUMEN

Cardiac hypertrophy can lead to heart failure (HF), but it is unpredictable which hypertrophied myocardium will progress to HF. We surmised that apart from hypertrophy-related genes, failure-related genes are expressed before the onset of failure, permitting molecular prediction of HF. Hearts from hypertensive homozygous renin-overexpressing (Ren-2) rats that had progressed to early HF were compared by microarray analysis to Ren-2 rats that had remained compensated. To identify which HF-related genes preceded failure, cardiac biopsy specimens were taken during compensated hypertrophy and we then monitored whether the rat progressed to HF or remained compensated. Among 48 genes overexpressed in failing hearts, we focused on thrombospondin-2 (TSP2). TSP2 was selectively overexpressed only in biopsy specimens from rats that later progressed to HF. Moreover, expression of TSP2 was increased in human hypertrophied hearts with decreased (0.19+/-0.01) versus normal ejection fraction (0.11+/-0.03 [arbitrary units]; P<0.05). Angiotensin II induced fatal cardiac rupture in 70% of TSP2 knockout mice, with cardiac failure in the surviving mice; this was not seen in wild-type mice. In TSP2 knockout mice, angiotensin II increased matrix metalloproteinase (MMP)-2 and MMP-9 activity by 120% and 390% compared with wild-type mice (P<0.05). In conclusion, we identify TSP2 as a crucial regulator of the integrity of the cardiac matrix that is necessary for the myocardium to cope with increased loading and that may function by its regulation of MMP activity. This suggests that expression of TSP2 marks an early-stage molecular program that is activated uniquely in hypertrophied hearts that are prone to fail.


Asunto(s)
Gasto Cardíaco Bajo/etiología , Matriz Extracelular/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Miocardio/metabolismo , Trombospondinas/biosíntesis , Angiotensina II/antagonistas & inhibidores , Angiotensina II/toxicidad , Animales , Animales Modificados Genéticamente , Gasto Cardíaco Bajo/genética , Gasto Cardíaco Bajo/metabolismo , Cardiomiopatías/inducido químicamente , Colagenasas/metabolismo , Progresión de la Enfermedad , Precursores Enzimáticos/metabolismo , Gelatinasas/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Rotura Cardíaca/inducido químicamente , Rotura Cardíaca/patología , Humanos , Hipertensión/complicaciones , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/genética , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Renina/genética , Volumen Sistólico , Trombospondinas/genética , Trombospondinas/fisiología , Regulación hacia Arriba
20.
Biochem Pharmacol ; 64(10): 1483-91, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12417261

RESUMEN

In the present study, we have analyzed the response of human smooth muscle cell (SMC)s to oxidative stress, in terms of recruitment of key elements of the stress-activated protein kinase (SAPK) pathway, such as Rac(1), p38, and the small heat shock protein (HSP)27. The level of expression of three small HSPs, alphaB-crystallin, HSP20, HSP27, as well as the phosphorylation levels of HSP27 and p38, were higher in cultured, asynchronously growing SMCs originating from left interior mammary artery (LIMA) than those originating from aorta, saphenous vein, and umbilical vein, validating the choice of SMCs from LIMA as a model system in our study. In synchronized, quiescent SMCs from LIMA, oxidative stress (H(2)O(2) stimulation)-induced membrane translocation of Rac(1), p38 phosphorylation, membrane translocation, and phosphorylation of HSP27. In these cells, simvastatin (S), an HMG-CoA reductase inhibitor, blocked, in a mevalonate-dependent way, oxidative stress-induced membrane translocation of Rac(1). However, S pretreatment prior to oxidative stress increased the levels of p38 phosphorylation, HSP27 membrane translocation/phosphorylation, actin polymerization, and apoptosis in these cells, in a mevalonate-dependent way. These results establish that S pretreatment has a stimulatory effect on the stress-activated p38/HSP27 pathway, despite its blocking effect on Rac(1) activation.


Asunto(s)
Proteínas de Choque Térmico , Músculo Liso/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Simvastatina/farmacología , Proteína de Unión al GTP rac1/metabolismo , Citoesqueleto de Actina/metabolismo , Anticolesterolemiantes/farmacología , Apoptosis , Células Cultivadas , Proteínas de Choque Térmico HSP27 , Humanos , Arterias Mamarias/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Chaperonas Moleculares , Músculo Liso/metabolismo , Estrés Oxidativo/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...