Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Nat Commun ; 13(1): 81, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013172

RESUMEN

Despite the high prevalence of ischemic heart diseases worldwide, no antibody-based treatment currently exists. Starting from the evidence that a specific isoform of the Bone Morphogenetic Protein 1 (BMP1.3) is particularly elevated in both patients and animal models of myocardial infarction, here we assess whether its inhibition by a specific monoclonal antibody reduces cardiac fibrosis. We find that this treatment reduces collagen deposition and cross-linking, paralleled by enhanced cardiomyocyte survival, both in vivo and in primary cultures of cardiac cells. Mechanistically, we show that the anti-BMP1.3 monoclonal antibody inhibits Transforming Growth Factor ß pathway, thus reducing myofibroblast activation and inducing cardioprotection through BMP5. Collectively, these data support the therapeutic use of anti-BMP1.3 antibodies to prevent cardiomyocyte apoptosis, reduce collagen deposition and preserve cardiac function after ischemia.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Proteína Morfogenética Ósea 1/genética , Cardiotónicos/farmacología , Cicatriz/genética , Fibrosis Endomiocárdica/genética , Infarto del Miocardio/genética , Animales , Proteína Morfogenética Ósea 1/antagonistas & inhibidores , Proteína Morfogenética Ósea 1/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 5/genética , Proteína Morfogenética Ósea 5/metabolismo , Estudios de Casos y Controles , Supervivencia Celular/efectos de los fármacos , Cicatriz/etiología , Cicatriz/metabolismo , Cicatriz/prevención & control , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/prevención & control , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Troponina T/genética , Troponina T/metabolismo
2.
Nat Commun ; 13(1): 149, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013221

RESUMEN

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Asunto(s)
Caquexia/genética , Fibrosis Endomiocárdica/genética , Insuficiencia Cardíaca/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Factores de Transcripción/genética , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Caquexia/metabolismo , Caquexia/fisiopatología , Caquexia/prevención & control , Estudios de Casos y Controles , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/fisiopatología , Fibrosis Endomiocárdica/prevención & control , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Pruebas de Función Cardíaca , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/agonistas , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/deficiencia , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Atrofia Muscular/prevención & control , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción/agonistas , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/deficiencia
3.
Cells ; 10(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34572150

RESUMEN

Atrial fibrillation is very common among the elderly and/or obese. While myocardial fibrosis is associated with atrial fibrillation, the exact mechanisms within atrial myocytes and surrounding non-myocytes are not fully understood. This review considers the potential roles of myocardial fibroblasts and myofibroblasts in fibrosis and modulating myocyte electrophysiology through electrotonic interactions. Coupling with (myo)fibroblasts in vitro and in silico prolonged myocyte action potential duration and caused resting depolarization; an optogenetic study has verified in vivo that fibroblasts depolarized when coupled myocytes produced action potentials. This review also introduces another non-myocyte which may modulate both myocardial (myo)fibroblasts and myocytes: epicardial adipose tissue. Epicardial adipocytes are in intimate contact with myocytes and (myo)fibroblasts and may infiltrate the myocardium. Adipocytes secrete numerous adipokines which modulate (myo)fibroblast and myocyte physiology. These adipokines are protective in healthy hearts, preventing inflammation and fibrosis. However, adipokines secreted from adipocytes may switch to pro-inflammatory and pro-fibrotic, associated with reactive oxygen species generation. Pro-fibrotic adipokines stimulate myofibroblast differentiation, causing pronounced fibrosis in the epicardial adipose tissue and the myocardium. Adipose tissue also influences myocyte electrophysiology, via the adipokines and/or through electrotonic interactions. Deeper understanding of the interactions between myocytes and non-myocytes is important to understand and manage atrial fibrillation.


Asunto(s)
Tejido Adiposo/metabolismo , Fibrilación Atrial/patología , Fibrosis Endomiocárdica/patología , Potenciales de Acción/fisiología , Adipocitos/fisiología , Adipoquinas/fisiología , Tejido Adiposo/patología , Fibrilación Atrial/metabolismo , Cardiomiopatías/patología , Fenómenos Electrofisiológicos , Fibrosis Endomiocárdica/metabolismo , Mapeo Epicárdico/métodos , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibrosis/patología , Corazón/fisiología , Atrios Cardíacos/patología , Humanos , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Miofibroblastos , Pericardio/patología
4.
Biomolecules ; 11(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34439775

RESUMEN

Galectin-3 (gal-3) is a fibrosis marker and may play a role in fibrosis of the left atrium (LA). Left atrial wall fibrosis may influence the transition from paroxysmal to non-paroxysmal atrial fibrillation (AF). In this study, we assessed the correlation of gal-3 concentration with the main echocardio-graphic parameters evaluating dimensions, volume, compliance, and left atrial contractility during AF and after successful electrical cardioversion (DCCV). The study included 63 patients with left atrial enlargement who qualified for DCCV due to persistent AF. The procedure recovered sinus rhythm in 43 (68.3%) patients. The concentration of gal-3 was negatively correlated with the echocardiographic parameters of LA including dimensions (LA length pre, rho = -0.38; p = 0.003), volume (LAV pre, rho = -0.39; p = 0.003), compliance (LASr mean post, rho = -0.33) and contractility (pLASRct mean post, rho = -0.33; p = 0.038). Negative correlations of gal-3 concentration were also observed in relation to the volume and contractility of the left ventricle. The concentration of gal-3 significantly negatively correlates with the size, systolic function, and compliance of the LA wall in patients with persistent AF. Determining gal-3 concentration in patients with persistent AF may help in the assessment of remodeling of the LA wall.


Asunto(s)
Fibrilación Atrial/metabolismo , Remodelación Atrial , Proteínas Sanguíneas/metabolismo , Fibrosis Endomiocárdica/metabolismo , Galectinas/metabolismo , Atrios Cardíacos/metabolismo , Anciano , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/terapia , Biomarcadores/metabolismo , Proteínas Sanguíneas/genética , Ecocardiografía , Cardioversión Eléctrica/métodos , Fibrosis Endomiocárdica/diagnóstico por imagen , Fibrosis Endomiocárdica/fisiopatología , Fibrosis Endomiocárdica/terapia , Femenino , Galectinas/genética , Expresión Génica , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Contracción Miocárdica , Función Ventricular Izquierda
5.
Cells ; 10(7)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34359886

RESUMEN

Cardiac fibrosis is the excess deposition of extracellular matrix (ECM), such as collagen. Myofibroblasts are major players in the production of collagen, and are differentiated primarily from resident fibroblasts. Collagen can compensate for the dead cells produced by injury. The appropriate production of collagen is beneficial for preserving the structural integrity of the heart, and protects the heart from cardiac rupture. However, excessive deposition of collagen causes cardiac dysfunction. Recent studies have demonstrated that myofibroblasts can change their phenotypes. In addition, myofibroblasts are found to have functions other than ECM production. Myofibroblasts have macrophage-like functions, in which they engulf dead cells and secrete anti-inflammatory cytokines. Research into fibroblasts has been delayed due to the lack of selective markers for the identification of fibroblasts. In recent years, it has become possible to genetically label fibroblasts and perform sequencing at single-cell levels. Based on new technologies, the origins of fibroblasts and myofibroblasts, time-dependent changes in fibroblast states after injury, and fibroblast heterogeneity have been demonstrated. In this paper, recent advances in fibroblast and myofibroblast research are reviewed.


Asunto(s)
Fibrosis Endomiocárdica/patología , Matriz Extracelular/patología , Fibroblastos/patología , Macrófagos/patología , Miofibroblastos/patología , Animales , Cardiotónicos/uso terapéutico , Diferenciación Celular , Linaje de la Célula , Colágeno/genética , Colágeno/metabolismo , Citocinas/genética , Citocinas/metabolismo , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Fibrosis Endomiocárdica/tratamiento farmacológico , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/clasificación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Transducción de Señal
6.
Bull Exp Biol Med ; 171(3): 305-311, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34302205

RESUMEN

We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-ß pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.


Asunto(s)
Angiotensina II/farmacología , Antifibróticos/farmacología , Proteína Morfogenética Ósea 4/genética , Fibroblastos/efectos de los fármacos , MAP Quinasa Quinasa 4/genética , Alcaloides de la Vinca/farmacología , Animales , Animales Recién Nacidos , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/metabolismo , Colágeno/genética , Colágeno/metabolismo , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Ratas , Ratas Wistar , Transducción de Señal , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Am Heart Assoc ; 10(15): e021069, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34320813

RESUMEN

Background Cardiac fibrosis plays a crucial role in the pathogenesis of dilated cardiomyopathy (DCM). HE4 (human epididymis protein 4) is a secretory protein expressed in activated fibroblasts that exacerbates tissue fibrosis. In the present study, we investigated the clinical utility of HE4 measurement in patients with DCM and its pathophysiological role in preclinical experiments in vivo and in vitro. Methods and Results We measured serum HE4 levels of 87 patients with DCM. Endomyocardial biopsy expressed severe fibrosis only in the high HE4 group (P<0.0001). Echocardiography showed that left ventricular end-diastolic diameter tends to decrease over time (58±7.3 to 51±6.6 mm; P<0.0001) in the low HE4 group (<59.65 pmol/L [median value]). HE4 was significantly associated with risk reduction of mortality and cardiovascular hospitalization in multivariate Cox model. In vivo, HE4 was highly expressed in kidney and lung tissue of mouse, and scarcely expressed in heart. In genetically induced DCM mouse model, HE4 expression increased in kidney but not in heart and lung. In vitro, supernatant from HE4-transfected human embryonic kidney 293T cells enhanced transdifferentiation of rat neonatal fibroblasts and increased expression of fibrosis-related genes, and this was accompanied by the activation of extracellular signal-regulated kinase signaling in cardiac fibroblasts. Treatment with an inhibitor of upstream signal of extracellular signal-regulated kinase or a neutralizing HE4 antibody canceled the profibrotic properties of HE4. Conclusions HE4 functions as a secretory factor, activating cardiac fibroblasts, thereby inducing cardiac interstitial fibrosis. HE4 could be a promising biomarker for assessing ongoing fibrosis and a novel therapeutic target in DCM. Registration URL: https://upload.umin.ac.jp/cgi-open-bin/ctr; Unique identifier: UMIN000043062.


Asunto(s)
Cardiomiopatía Dilatada , Fibrosis Endomiocárdica , Ventrículos Cardíacos , Riñón , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Biopsia/métodos , Síndrome Cardiorrenal/metabolismo , Síndrome Cardiorrenal/patología , Cardiomiopatía Dilatada/diagnóstico por imagen , Cardiomiopatía Dilatada/mortalidad , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/terapia , Transdiferenciación Celular , Descubrimiento de Drogas , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Hospitalización/estadística & datos numéricos , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/fisiología , Valor Predictivo de las Pruebas , Ratas , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP/antagonistas & inhibidores , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP/inmunología , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP/metabolismo
8.
J Biol Chem ; 297(1): 100893, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34153319

RESUMEN

Fibrosis is a pronounced feature of heart disease and the result of dysregulated activation of resident cardiac fibroblasts (CFs). Recent work identified stress-induced degradation of the cytoskeletal protein ßIV-spectrin as an important step in CF activation and cardiac fibrosis. Furthermore, loss of ßIV-spectrin was found to depend on Ca2+/calmodulin-dependent kinase II (CaMKII). Therefore, we sought to determine the mechanism for CaMKII-dependent regulation of ßIV-spectrin and CF activity. Computational screening and MS revealed a critical serine residue (S2250 in mouse and S2254 in human) in ßIV-spectrin phosphorylated by CaMKII. Disruption of ßIV-spectrin/CaMKII interaction or alanine substitution of ßIV-spectrin Ser2250 (ßIV-S2254A) prevented CaMKII-induced degradation, whereas a phosphomimetic construct (ßIV-spectrin with glutamic acid substitution at serine 2254 [ßIV-S2254E]) showed accelerated degradation in the absence of CaMKII. To assess the physiological significance of this phosphorylation event, we expressed exogenous ßIV-S2254A and ßIV-S2254E constructs in ßIV-spectrin-deficient CFs, which have increased proliferation and fibrotic gene expression compared with WT CFs. ßIV-S2254A but not ßIV-S2254E normalized CF proliferation, gene expression, and contractility. Pathophysiological targeting of ßIV-spectrin phosphorylation and subsequent degradation was identified in CFs activated with the profibrotic ligand angiotensin II, resulting in increased proliferation and signal transducer and activation of transcription 3 nuclear accumulation. While therapeutic delivery of exogenous WT ßIV-spectrin partially reversed these trends, ßIV-S2254A completely negated increased CF proliferation and signal transducer and activation of transcription 3 translocation. Moreover, we observed ßIV-spectrin phosphorylation and associated loss in total protein within human heart tissue following heart failure. Together, these data illustrate a considerable role for the ßIV-spectrin/CaMKII interaction in activating profibrotic signaling.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Fibrosis Endomiocárdica/metabolismo , Miofibroblastos/metabolismo , Espectrina/metabolismo , Sustitución de Aminoácidos , Animales , Células COS , Proliferación Celular , Células Cultivadas , Chlorocebus aethiops , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/fisiología , Fosforilación , Espectrina/genética
9.
Pflugers Arch ; 472(12): 1719-1732, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33047172

RESUMEN

Cardiac fibroblasts play an important role in cardiac matrix turnover and are involved in cardiac fibrosis development. Ca2+ is a driving belt in this phenomenon. This study evaluates the functional expression and contribution of the Ca2+-activated channel TRPM4 in atrial fibroblast phenotype. Molecular and electrophysiological investigations were conducted in human atrial fibroblasts in primary culture and in atrial fibroblasts obtained from wild-type and transgenic mice with disrupted Trpm4 gene (Trpm4-/-). A typical TRPM4 current was recorded on human cells (equal selectivity for Na+ and K+, activation by internal Ca2+, voltage sensitivity, conductance of 23.2 pS, inhibition by 9-phenanthrol (IC50 = 6.1 × 10-6 mol L-1)). Its detection rate was 13% on patches at days 2-4 in culture but raised to 100% on patches at day 28. By the same time, a cell growth was observed. This growth was smaller when cells were maintained in the presence of 9-phenanthrol. Similar cell growth was measured on wild-type mice atrial fibroblasts during culture. However, this growth was minimized on Trpm4-/- mice fibroblasts compared to control animals. In addition, the expression of alpha smooth muscle actin increased during culture of atrial fibroblasts from wild-type mice. This was not observed in Trpm4-/- mice fibroblasts. It is concluded that TRPM4 participates in fibroblast growth and could thus be involved in cardiac fibrosis.


Asunto(s)
Fibrosis Endomiocárdica/metabolismo , Miofibroblastos/metabolismo , Canales Catiónicos TRPM/metabolismo , Potenciales de Acción , Anciano , Animales , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Miocardio/citología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/fisiología , Fenantrenos
10.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998408

RESUMEN

Despite relevant advances made in therapies for cardiovascular diseases (CVDs), they still represent the first cause of death worldwide. Cardiac fibrosis and excessive extracellular matrix (ECM) remodeling are common end-organ features in diseased hearts, leading to tissue stiffness, impaired myocardial functional, and progression to heart failure. Although fibrosis has been largely recognized to accompany and complicate various CVDs, events and mechanisms driving and governing fibrosis are still not entirely elucidated, and clinical interventions targeting cardiac fibrosis are not yet available. Immune cell types, both from innate and adaptive immunity, are involved not just in the classical response to pathogens, but they take an active part in "sterile" inflammation, in response to ischemia and other forms of injury. In this context, different cell types infiltrate the injured heart and release distinct pro-inflammatory cytokines that initiate the fibrotic response by triggering myofibroblast activation. The complex interplay between immune cells, fibroblasts, and other non-immune/host-derived cells is now considered as the major driving force of cardiac fibrosis. Here, we review and discuss the contribution of inflammatory cells of innate immunity, including neutrophils, macrophages, natural killer cells, eosinophils and mast cells, in modulating the myocardial microenvironment, by orchestrating the fibrogenic process in response to tissue injury. A better understanding of the time frame, sequences of events during immune cells infiltration, and their action in the injured inflammatory heart environment, may provide a rationale to design new and more efficacious therapeutic interventions to reduce cardiac fibrosis.


Asunto(s)
Comunicación Celular/inmunología , Fibrosis Endomiocárdica/inmunología , Inmunidad Innata , Daño por Reperfusión Miocárdica/inmunología , Miocardio/inmunología , Miofibroblastos/inmunología , Inmunidad Adaptativa , Animales , Citocinas/inmunología , Citocinas/metabolismo , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Eosinófilos/inmunología , Eosinófilos/metabolismo , Eosinófilos/patología , Matriz Extracelular/química , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Humanos , Inflamación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Mastocitos/inmunología , Mastocitos/metabolismo , Mastocitos/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología
11.
Am J Physiol Cell Physiol ; 319(5): C781-C796, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32845719

RESUMEN

Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.


Asunto(s)
Cardiomiopatía Dilatada/genética , Fibrosis Endomiocárdica/genética , Síndrome Mucocutáneo Linfonodular/genética , Infarto del Miocardio/genética , Miocarditis/genética , Tenascina/genética , Animales , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Regulación de la Expresión Génica , Humanos , Inflamación , Ratones , Ratones Noqueados , Síndrome Mucocutáneo Linfonodular/metabolismo , Síndrome Mucocutáneo Linfonodular/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/metabolismo , Miocardio/patología , Tenascina/metabolismo , Remodelación Ventricular/genética , Cicatrización de Heridas/genética
12.
Matrix Biol ; 91-92: 75-91, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32446910

RESUMEN

The cardiac stroma plays essential roles in health and following cardiac damage. The major player of the stroma with respect to extracellular matrix deposition, maintenance and remodeling is the poorly defined fibroblast. It has long been recognized that there is considerable variability to the fibroblast phenotype. With the advent of new, high throughput analytical methods our understanding and appreciation of this heterogeneity has grown dramatically. This review aims to explore the diversity of cardiac fibroblasts and highlights new insights into the diverse nature of these cells and their progenitors as revealed by single cell sequencing and fate mapping studies. We propose that at least in part the observed heterogeneity is related to the existence of a differentiation cascade within stromal cells. Beyond in-organ heterogeneity, we also discuss how the stromal response to damage differs between non-regenerating organs such as the heart and regenerating organs such as skeletal muscle. In exploring possible causes for these differences, we outline that although fibrogenic cells from different organs overlap in many properties, they still possess organ-specific transcriptional signatures and differentiation biases that make them functionally distinct.


Asunto(s)
Fibrosis Endomiocárdica/genética , Proteínas de la Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Miofibroblastos/metabolismo , Células del Estroma/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula/genética , Rastreo Celular/métodos , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miocardio/citología , Miocardio/metabolismo , Miofibroblastos/citología , Especificidad de Órganos , Transducción de Señal , Análisis de la Célula Individual/métodos , Células del Estroma/citología , Transcripción Genética
13.
Matrix Biol ; 91-92: 117-135, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32416242

RESUMEN

Fibroblasts are the primary regulator of cardiac extracellular matrix (ECM). In response to disease stimuli cardiac fibroblasts undergo cell state transitions to a myofibroblast phenotype, which underlies the fibrotic response in the heart and other organs. Identifying regulators of fibroblast state transitions would inform which pathways could be therapeutically modulated to tactically control maladaptive extracellular matrix remodeling. Indeed, a deeper understanding of fibroblast cell state and plasticity is necessary for controlling its fate for therapeutic benefit. p38 mitogen activated protein kinase (MAPK), which is part of the noncanonical transforming growth factor ß (TGFß) pathway, is a central regulator of fibroblast to myofibroblast cell state transitions that is activated by chemical and mechanical stress signals. Fibroblast intrinsic signaling, local and global cardiac mechanics, and multicellular interactions individually and synergistically impact these state transitions and hence the ECM, which will be reviewed here in the context of cardiac fibrosis.


Asunto(s)
Fibrosis Endomiocárdica/genética , Proteínas de la Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Infarto del Miocardio/genética , Miofibroblastos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Animales , Diferenciación Celular , Linaje de la Célula/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Matriz Extracelular/química , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/patología , Transducción de Señal , Transcripción Genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Matrix Biol ; 91-92: 167-175, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32438054

RESUMEN

Inflammation contributes to the development of heart failure (HF) through multiple mechanisms including regulating extracellular matrix (ECM) degradation and deposition. Interactions between cells in the myocardium orchestrates the magnitude and duration of inflammatory cell recruitment and ECM remodeling events associated with HF. More recently, studies have shown T-cells have signficant roles in post-MI wound healing. T-cell biology in HF illustrates the complexity of cross-talk between inflammatory cell types and resident fibroblasts. This review will focus on T-cell recruitment to the myocardium and T-cell specific factors that might influence cardiac wound healing and fibrosis in the heart with consideration of age and sex as important factors in T-cell activity.


Asunto(s)
Fibrosis Endomiocárdica/inmunología , Matriz Extracelular/inmunología , Fibroblastos/inmunología , Insuficiencia Cardíaca/inmunología , Infarto del Miocardio/inmunología , Linfocitos T/inmunología , Factores de Edad , Antígenos CD/genética , Antígenos CD/inmunología , Comunicación Celular/genética , Comunicación Celular/inmunología , Citocinas/genética , Citocinas/inmunología , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/patología , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Inflamación , Macrófagos/inmunología , Macrófagos/patología , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Transducción de Señal , Linfocitos T/clasificación , Linfocitos T/patología
15.
Bull Exp Biol Med ; 168(5): 658-661, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32246376

RESUMEN

Reduced expression of the key regulator of cardiac metabolism, transcription factor PPARα, in surgical samples of the auricles from patients with coronary heart disease and heart failure was detected by real-time quantitative PCR. These changes indicate reduced activity of this factor and a shift of energy metabolism from oxidative phosphorylation to glycolysis typical of dedifferentiated cells. Electron microscopy revealed dedifferentiated cardiomyocytes with disassembled contractile apparatus and disorganized sarcomeres. In the examined specimens from patients with heart failure, severe myocardial fibrosis was revealed.


Asunto(s)
Metabolismo Energético/fisiología , Corazón/fisiología , Miocitos Cardíacos/metabolismo , PPAR alfa/fisiología , Regeneración/fisiología , Biopsia , Desdiferenciación Celular/genética , Enfermedad Coronaria/genética , Enfermedad Coronaria/metabolismo , Enfermedad Coronaria/patología , Enfermedad Coronaria/fisiopatología , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/fisiopatología , Metabolismo Energético/genética , Regulación de la Expresión Génica , Glucólisis/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Fosforilación Oxidativa , PPAR alfa/genética , PPAR alfa/metabolismo
16.
Mol Biol Rep ; 47(4): 2605-2617, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32130618

RESUMEN

Atrial fibrillation (AF) is a commonly encountered heart arrhythmia and a risk factor for cardiovascular system. The purpose of the present study was to explore the role of long non-coding RNA myocardial infarction-associated transcript (MIAT) in AF and AF-induced myocardial fibrosis and the possible mechanisms involved in this process. We successfully induced an AF rat model. Expression of MIAT presented a dramatic increase, while microRNA (miR)-133a-3p presented a dramatic decrease in atrium tissues of rats with AF induction. In addition, we also found that MIAT was highly expressed and miR-133a-3p was significantly reduced in peripheral blood leukocyte of AF patients. For biological function exploration of MIAT/miR-133a-3p axis, MIAT was knocked down using small hairpin RNA (shRNA) lentivirus injection and the rescue experiments were performed simultaneously by inhibiting miR-133a-3p using anti-miR-133a-3p lentivirus injection in rats with AF. MIAT downregulation significantly alleviated AF, increased atrial effective refractory period (AERP), and reduced the duration of AF as well as cardiomyocytes apoptosis. Whereas these effects of MIAT downregulation on AF were reversed by anti-miR-133a-3p administration. Luciferase reporter revealed that miR-133a-3p was directly regulated by MIAT. Moreover, MIAT knockdown effectively reduced AF-induced atrial fibrosis by detecting reduced collagen in the right atria and inhibited expression of fibrosis-related gene expression of collagen I, collagen III, connective tissue growth factor (CTGF) and transforming growth factor-ß1 (TGF-ß1) in rats with AF, these findings were in contrast with the findings for rats with inhibition of miR-133a-3p. In conclusion, our study demonstrated the role of MIAT downregulation in alleviating AF and AF-induced myocardial fibrosis, and the functional regulatory pathway of MIAT targeting miR-133a-3p.


Asunto(s)
Fibrilación Atrial/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Animales , Apoptosis/genética , Fibrilación Atrial/fisiopatología , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , China , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Femenino , Fibrosis/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , ARN Largo no Codificante/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
17.
Int Heart J ; 60(3): 648-655, 2019 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-31019180

RESUMEN

Apelin was proved to attenuate cardiac interstitial fibrosis. However, the association between apelin level and myocardial fibrosis in patients with hypertrophic cardiomyopathy (HCM) is still unclear.This study aims to determine whether apelin is associated with myocardial fibrosis in HCM and investigate the predictive values of apelin for myocardial fibrosis in HCM.One hundred sixteen patients with HCM were enrolled in this study. Plasma apelin-13 and high-sensitivity cardiac troponin I (cTNI) were measured. The cardiac systolic and diastolic functions were evaluated by echocardiography, and the presence and extent of cardiac fibrosis were assessed by cardiac magnetic resonance. All statistical data were analyzed by SPSS version 21.0.The percentage of late gadolinium enhancement (LGE) was negatively correlated with apelin and positively correlated with cTNI, maximum wall thickness (MWT), and left ventricular mass index in the overall patients with HCM and LGE. Apelin, cTNI, MWT, and left ventricular ejection fraction were independent predictors of the presence of LGE. The cutoff values of apelin, cTNI, and MWT were 1.24 pg/mL, 0.031 ng/mL, and 19 mm, respectively, for the prediction of LGE. The combined measurements of MWT ≥ 19 mm and/or apelin ≤ 1.24 pg/mL, as well as the combined measurements of MWT ≥ 19 mm and/or cTNI ≥ 0.031 ng/mL, obtained higher specificity and higher sensitivity, thus, indicating the presence of LGE.Plasma apelin and cTNI are independent predictors of myocardial fibrosis. The combined measurements of serum apelin and MWT, as well as cTNI and MWT, showed higher predictive values for predicting myocardial fibrosis in patients with HCM.


Asunto(s)
Apelina/sangre , Cardiomiopatía Hipertrófica/complicaciones , Fibrosis Endomiocárdica/diagnóstico por imagen , Troponina I/sangre , Adulto , Biomarcadores/sangre , Cardiomiopatía Hipertrófica/metabolismo , Fibrosis Endomiocárdica/metabolismo , Femenino , Gadolinio/metabolismo , Humanos , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas
18.
Aging Cell ; 18(3): e12926, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30834643

RESUMEN

Biological aging dynamically alters normal immune and cardiac function, favoring the production of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) and increased instances of cardiac distress. Cardiac failure is the primary reason for hospitalization of the elderly (65+ years). The elderly are also increasingly susceptible to developing chronic bacterial infections due to aging associated immune abnormalities. Since bacterial infections compound the rates of cardiac failure in the elderly, and this phenomenon is not entirely understood, the interplay between the immune system and cardiovascular function in the elderly is of great interest. Using Mycobacterium avium, an opportunistic pathogen, we investigated the effect of mycobacteria on cardiac function in aged mice. Young (2-3 months) and old (18-20 months) C57BL/6 mice were intranasally infected with M. avium strain 104, and we compared the bacterial burden, immune status, cardiac electrical activity, pathology, and function of infected mice against uninfected age-matched controls. Herein, we show that biological aging may predispose old mice infected with M. avium to mycobacterial dissemination into the heart tissue and this leads to cardiac dysfunction. M. avium infected old mice had significant dysrhythmia, cardiac hypertrophy, increased recruitment of CD45+ leukocytes, cardiac fibrosis, and increased expression of inflammatory genes in isolated heart tissue. This is the first study to report the effect of mycobacteria on cardiac function in an aged model. Our findings are critical to understanding how nontuberculous mycobacterium (NTM) and other mycobacterial infections contribute to cardiac dysfunction in the elderly population.


Asunto(s)
Arritmias Cardíacas/microbiología , Cardiomegalia/microbiología , Fibrosis Endomiocárdica/microbiología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Micobacterias no Tuberculosas , Envejecimiento/inmunología , Envejecimiento/patología , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Susceptibilidad a Enfermedades , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Inflamación/microbiología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Antígenos Comunes de Leucocito/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Mycobacterium no Tuberculosas/patología , Mycobacterium avium , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
19.
PLoS One ; 14(2): e0199802, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30789913

RESUMEN

INTRODUCTION: This study aimed to investigate the effect of aerobic exercise on the expression of neitrin-1,DCC receptor and myocardial fibrosis in rats with acute myocardial infarction. METHODS: Twenty-four rats were randomly divided into three groups: the sham group (n = 8), the acute myocardial infarction (AMI) model group (n = 8), and the aerobic exercise treatment after acute myocardial infarction group (ET) (n = 8). After 10 weeks, the serum levels of netrin-1, tumor necrosis factor alpha α (TNF-α), and interleukin 6 (IL-6) were measured. The expression of matrix metalloproteinase 2 and 9 (MMP2, 9), and their inhibitor, tissue inhibitor of metalloproteinase 2 (TIMP2), myocardial netrin-1, and the deleted in colorectal cancer (DCC) receptor were evaluated. Histopathological results were also evaluated. The collagen volume fraction of the myocardial tissues was also calculated. RESULTS: Compared with the sham group, in the AMI and ET groups, left ventricular end diastolic pressure (LVEDP) were increased, while left ventricular systolic pressure (LVSP), and left ventricular pressure maximal rate of rise and fall (± dp/dtmax) were significantly decreased (P<0.05,). Compared with the AMI group, in the ET group, LVSP, and ±dp/dtmax were significantly increased while LVEDP was decreased (P<0.05). Compared with the sham group, the AMI group and ET groups showed increased levels of serum TNF-α, IL-6 and significantly reduced levels of netrin-1. Levels of TNF-α and IL-6 were significantly reduced in the ET group compared with the AMI group, whereas the level of netrin-1 was increased. The expression of myocardial MMP2 and MMP9 was significantly increased in the AMI group compared with the sham group, whereas that of myocardial netrin-1, TIMP2 and the DCC receptor, was significantly reduced. Compared with the AMI group, the ET group showed reduced expression of myocardial MMP2 and MMP9 proteins, whereas expression of myocardial netrin-1, TIMP2 and the DCC receptor, was significantly increased. The collagen volume fraction of the myocardial tissues was significantly increased in the AMI group and the ET group compared with the sham group, with a greater increase in the AMI group. CONCLUSIONS: Aerobic exercise increased levels of serum netrin-1, myocardial netrin-1, and the DCC receptor and reduced the expression of myocardial MMP2 and MMP9 proteins, to improve the degree of fibrosis following myocardial infarction in rats.


Asunto(s)
Fibrosis Endomiocárdica/metabolismo , Infarto del Miocardio/metabolismo , Netrina-1/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Cardiomiopatías/metabolismo , Receptor DCC/sangre , Fibrosis Endomiocárdica/sangre , Fibrosis/metabolismo , Interleucina-6/sangre , Masculino , Metaloproteinasa 2 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/sangre , Infarto del Miocardio/sangre , Miocardio/metabolismo , Miocardio/patología , Netrina-1/sangre , Condicionamiento Físico Animal/métodos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/sangre , Función Ventricular Izquierda , Remodelación Ventricular
20.
Mol Aspects Med ; 65: 70-99, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30056242

RESUMEN

Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-ß family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.


Asunto(s)
Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/metabolismo , Animales , Biomarcadores , Microambiente Celular , Susceptibilidad a Enfermedades , Fibrosis Endomiocárdica/diagnóstico , Fibrosis Endomiocárdica/terapia , Epigénesis Genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica , Humanos , Miofibroblastos/metabolismo , Transducción de Señal , Remodelación Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA