Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 126: 1-12, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30408466

RESUMEN

Muscle atrophy F-box (MAFbx/atrogin-1), an E3 ubiquitin ligase, is a crucial mediator of skeletal muscle atrophy and cardiac hypertrophy in response to pressure overload and exercise. The role of MAFbx in the regulation of cardiac remodeling after myocardial infarction (MI) remains unclear. Permanent coronary ligation of the left coronary artery was performed on MAFbx knockout (KO) and wild-type (WT) mice and MAFbx expression in the WT mice was shown to be significantly increased in the left ventricles after MI. The mortality rate due to post-MI cardiac rupture was significantly decreased in MAFbx KO mice compared to that in the WT mice. DNA microarray and mRNA expression analyses revealed that the upregulation of genes involved in inflammatory processes and cell motility of leukocytes and neutrophils, including Mmp9, Il1b, Cxcl2, and Nlrp3, was significantly attenuated in MAFbx KO mice 1 day after MI. MAFbx downregulation inhibited nuclear factor-κB (Nfkb) activation after MI. Flow cytometry results demonstrated that the myocardial infiltration of neutrophils was suppressed in MAFbx KO mice 1 day after MI. Nlrp3 and Il1b protein levels were decreased in MAFbx KO mice compared with those in the WT mice. MAFbx downregulation significantly attenuated Tnfa-induced Cxcl2, Il1b, and Nlrp3 expression in cardiomyocytes. We conclude that MAFbx plays an important role in the mediation of excessive inflammation, including neutrophil infiltration, inflammasome formation, and production of proinflammatory cytokines through the activation of Nfkb, promoting cardiac rupture after MI.


Asunto(s)
Rotura Cardíaca Posinfarto/metabolismo , Proteínas Musculares/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Eliminación de Gen , Regulación de la Expresión Génica , Rotura Cardíaca Posinfarto/genética , Ventrículos Cardíacos/patología , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Infiltración Neutrófila , Ratas
2.
PLoS One ; 12(11): e0187894, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29121663

RESUMEN

BACKGROUND: Cardiac rupture is an important cause of death in the acute phase after myocardial infarction (MI). Macrophages play a pivotal role in cardiac remodeling after MI. Apoptosis inhibitor of macrophage (AIM) is secreted specifically by macrophages and contributes to macrophage accumulation in inflamed tissue by maintaining survival and recruiting macrophages. In this study, we evaluated the role of AIM in macrophage accumulation in the infarcted myocardium and cardiac rupture after MI. METHODS AND RESULTS: Wild-type (WT) and AIM‒/‒ mice underwent permanent left coronary artery ligation and were followed-up for 7 days. Macrophage accumulation and phenotypes (M1 pro-inflammatory macrophage or M2 anti-inflammatory macrophage) were evaluated by immunohistological analysis and RT-PCR. Matrix metalloproteinase (MMP) activity levels were measured by gelatin zymography. The survival rate was significantly higher (81.1% vs. 48.2%, P<0.05), and the cardiac rupture rate was significantly lower in AIM‒/‒ mice than in WT mice (10.8% vs. 31.5%, P<0.05). The number of M1 macrophages and the expression levels of M1 markers (iNOS and IL-6) in the infarcted myocardium were significantly lower in AIM‒/‒ mice than in WT mice. In contrast, there was no difference in the number of M2 macrophages and the expression of M2 markers (Arg-1, CD206 and TGF-ß1) between the two groups. The ratio of apoptotic macrophages in the total macrophages was significantly higher in AIM‒/‒ mice than in WT mice, although MCP-1 expression did not differ between the two groups. MMP-2 and 9 activity levels in the infarcted myocardium were significantly lower in AIM‒/‒ mice than in WT mice. CONCLUSIONS: These findings suggest that AIM depletion decreases the levels of M1 macrophages, which are a potent source of MMP-2 and 9, in the infarcted myocardium in the acute phase after MI by promoting macrophage apoptosis, and leads to a decrease in the incidence of cardiac rupture and improvements in survival rates.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/deficiencia , Rotura Cardíaca Posinfarto/epidemiología , Macrófagos/metabolismo , Infarto del Miocardio/metabolismo , Receptores Inmunológicos/deficiencia , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Modelos Animales de Enfermedad , Femenino , Rotura Cardíaca Posinfarto/genética , Rotura Cardíaca Posinfarto/metabolismo , Incidencia , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/inmunología , Masculino , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores Inmunológicos/genética , Receptores Depuradores , Tasa de Supervivencia
3.
Mol Ther ; 25(1): 192-204, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129114

RESUMEN

Inflammation plays an important role in cardiac injuries. Here, we examined the role of miRNA in regulating inflammation and cardiac injury during myocardial infarction. We showed that mir-155 expression was increased in the mouse heart after myocardial infarction. Upregulated mir-155 was primarily presented in macrophages and cardiac fibroblasts of injured hearts, while pri-mir-155 was only expressed in macrophages. mir-155 was also presented in exosomes derived from macrophages, and it can be transferred into cardiac fibroblasts by macrophage-derived exosomes. A mir-155 mimic or mir-155 containing exosomes inhibited cardiac fibroblast proliferation by downregulating Son of Sevenless 1 expression and promoted inflammation by decreasing Suppressor of Cytokine Signaling 1 expression. These effects were reversed by the addition of a mir-155 inhibitor. In vivo, mir-155-deficient mice showed a significant reduction of the incidence of cardiac rupture and an improved cardiac function compared with wild-type mice. Moreover, transfusion of wild-type macrophage exosomes to mir-155-/- mice exacerbated cardiac rupture. Finally, the mir-155-deficient mice exhibited elevated fibroblast proliferation and collagen production, along with reduced cardiac inflammation in injured heart. Taken together, our results demonstrate that activated macrophages secrete mir-155-enriched exosomes and identify macrophage-derived mir-155 as a paracrine regulator for fibroblast proliferation and inflammation; thus, a mir-155 inhibitor (i.e., mir-155 antagomir) has the potential to be a therapeutic agent for reducing acute myocardial-infarction-related adverse events.


Asunto(s)
Comunicación Celular , Exosomas/metabolismo , Fibroblastos/metabolismo , Macrófagos/metabolismo , MicroARNs/genética , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Expresión Génica , Regulación de la Expresión Génica , Rotura Cardíaca Posinfarto/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Activación de Macrófagos , Ratones , Modelos Biológicos , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Miocardio/metabolismo , Interferencia de ARN , Transporte de ARN , Proteína SOS1/genética
4.
Am J Physiol Heart Circ Physiol ; 311(6): H1485-H1497, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27769998

RESUMEN

We have reported that the Toll-like receptor 9 (TLR9) signaling pathway plays an important role in the development of pressure overload-induced inflammatory responses and heart failure. However, its role in cardiac remodeling after myocardial infarction has not been elucidated. TLR9-deficient and control C57Bl/6 wild-type mice were subjected to left coronary artery ligation. The survival rate 14 days postoperation was significantly lower in TLR9-deficient mice than that in wild-type mice with evidence of cardiac rupture in all dead mice. Cardiac magnetic resonance imaging showed no difference in infarct size and left ventricular wall thickness and function between TLR9-deficient and wild-type mice. There were no differences in the number of infiltrating inflammatory cells and the levels of inflammatory cytokine mRNA in infarct hearts between TLR9-deficient and wild-type mice. The number of α-smooth muscle actin (αSMA)-positive myofibroblasts and αSMA/Ki67-double-positive proliferative myofibroblasts was increased in the infarct and border areas in infarct hearts compared with those in sham-operated hearts in wild-type mice, but not in TLR9-deficient mice. The class B CpG oligonucleotide increased the phosphorylation level of NF-κB and the number of αSMA-positive and αSMA/Ki67-double-positive cells and these increases were attenuated by BAY1-7082, an NF-κB inhibitor, in cardiac fibroblasts isolated from wild-type hearts. The CpG oligonucleotide showed no effect on NF-κB activation or the number of αSMA-positive and αSMA/Ki67-double-positive cells in cardiac fibroblasts from TLR9-deficient hearts. Although the TLR9 signaling pathway is not involved in the acute inflammatory response in infarct hearts, it ameliorates cardiac rupture possibly by promoting proliferation and differentiation of cardiac fibroblasts.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Fibroblastos/citología , Rotura Cardíaca Posinfarto/genética , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptor Toll-Like 9/genética , Actinas/metabolismo , Animales , Western Blotting , Recuento de Células , Vasos Coronarios/cirugía , Citocinas/genética , Rotura Cardíaca Posinfarto/etiología , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/mortalidad , Inflamación , Antígeno Ki-67/metabolismo , Ligadura , Magnetoterapia , Masculino , Ratones , Ratones Noqueados , Infarto del Miocardio/complicaciones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Miocardio/patología , Miofibroblastos/citología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia
5.
Circ J ; 80(9): 1971-9, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27396441

RESUMEN

BACKGROUND: Inflammatory responses, especially by CD4(+)T cells activated by dendritic cells, are known to be important in the pathophysiology of cardiac repair after myocardial infarction (MI). Although co-stimulatory signals through B7 (CD80/86) and CD28 are necessary for CD4(+)T cell activation and survival, the roles of these signals in cardiac repair after MI are still unclear. METHODS AND RESULTS: C57BL/6 (Control) mice and CD28 knockout (CD28KO) mice were subjected to left coronary artery permanent ligation. The ratio of death by cardiac rupture within 5 days after MI was significantly higher in CD28KO mice compared with Control mice. Although there were no significant differences in the infarct size between the 2 groups, left ventricular end-diastolic and end-systolic diameters were significantly increased, and fractional shortening was significantly decreased in CD28KO mice compared with Control mice. Electron microscopic observation revealed that the extent of extracellular collagen fiber was significantly decreased in CD28KO mice compared with Control mice. The number of α-smooth muscle actin-positive myofibroblasts was significantly decreased, and matrix metalloproteinase-9 activity and the mRNA expression of interleukin-1ß were significantly increased in CD28KO mice compared with Control mice. CONCLUSIONS: Deletion of CD28 co-stimulatory signals exacerbates left ventricular remodeling and increases cardiac rupture after MI through prolongation of the inflammatory period and reduction of collagen fiber in the infarct scars. (Circ J 2016; 80: 1971-1979).


Asunto(s)
Antígenos CD28/deficiencia , Eliminación de Gen , Rotura Cardíaca Posinfarto/metabolismo , Infarto del Miocardio/metabolismo , Transducción de Señal , Remodelación Ventricular , Animales , Antígenos CD28/metabolismo , Regulación de la Expresión Génica , Rotura Cardíaca Posinfarto/genética , Rotura Cardíaca Posinfarto/patología , Rotura Cardíaca Posinfarto/fisiopatología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miofibroblastos/metabolismo , Miofibroblastos/ultraestructura
6.
J Mol Cell Cardiol ; 88: 55-63, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26393439

RESUMEN

Myocardial infarction is a leading cause of death, and cardiac rupture following myocardial infarction leads to extremely poor prognostic feature. A large body of evidence suggests that Akt is involved in several cardiac diseases. We previously reported that Akt-mediated Girdin phosphorylation is essential for angiogenesis and neointima formation. The role of Girdin expression and phosphorylation in myocardial infarction, however, is not understood. Therefore, we employed Girdin-deficient mice and Girdin S1416A knock-in (Girdin(SA/SA)) mice, replacing the Akt phosphorylation site with alanine, to address this question. We found that Girdin was expressed and phosphorylated in cardiac fibroblasts in vitro and that its phosphorylation was crucial for the proliferation and migration of cardiac fibroblasts. In vivo, Girdin was localized in non-cardiomyocyte interstitial cells and phosphorylated in α-smooth muscle actin-positive cells, which are likely to be cardiac myofibroblasts. In an acute myocardial infarction model, Girdin(SA/SA) suppressed the accumulation and proliferation of cardiac myofibroblasts in the infarcted area. Furthermore, lower collagen deposition in Girdin(SA/SA) mice impaired cardiac repair and resulted in increased mortality attributed to cardiac rupture. These findings suggest an important role of Girdin phosphorylation at serine 1416 in cardiac repair after acute myocardial infarction and provide insights into the complex mechanism of cardiac rupture through the Akt/Girdin-mediated regulation of cardiac myofibroblasts.


Asunto(s)
Rotura Cardíaca Posinfarto/metabolismo , Proteínas de Microfilamentos/metabolismo , Infarto del Miocardio/metabolismo , Miofibroblastos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Actinas/genética , Actinas/metabolismo , Sustitución de Aminoácidos , Animales , Animales Recién Nacidos , Proliferación Celular , Colágeno/genética , Colágeno/metabolismo , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Rotura Cardíaca Posinfarto/genética , Rotura Cardíaca Posinfarto/mortalidad , Rotura Cardíaca Posinfarto/patología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/genética , Infarto del Miocardio/genética , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/patología , Fosforilación , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Proteínas de Transporte Vesicular/genética
7.
Cardiol Rev ; 17(6): 293-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19829180

RESUMEN

Cardiac rupture after a myocardial infarction is an uncommon event with devastating consequences. Although the clinical features of rupture have been described, the genetic and molecular influences on this outcome in patients are less certain. In mice, at least 17 genetic models have been developed that enhance or suppress the likelihood of rupture postmyocardial infarction. The purpose of this review is to describe these recent advances, recognizing that nearly all of the information has been obtained from mouse models of free wall rupture. Although it is probable that the same genetic determinants apply to septal and papillary muscle rupture, the possibility remains that there are unique modulators of risk for rupture at differing anatomic sites within the heart. It is likely that the candidate genes also influence rupture in humans, although this conclusion must be confirmed. The mouse models will be helpful to direct future proteomic and genomic studies in patients and may already suggest certain fundamental pathways. For example, the essential role of collagen production and stabilization postmyocardial infarction may direct therapies to enhance collagen cross-linking and limit its degradation as a strategy to reduce rates of rupture and enhance myocardial healing.


Asunto(s)
Rotura Cardíaca Posinfarto/genética , Modelos Animales , Modelos Genéticos , Animales , Colágeno/genética , Colágeno/metabolismo , Rotura Cardíaca Posinfarto/metabolismo , Humanos , Ratones , Proteómica
8.
Cardiovasc Res ; 84(2): 273-82, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542177

RESUMEN

AIMS: Our objective was to study the effect of the genetic background on the wound healing process after myocardial infarction (MI) in mice. METHODS AND RESULTS: MI was induced in five different mouse strains (BalbC, C57Bl6, FVB, 129S6, and Swiss). At 3, 14, and 28 days after MI, cardiac dimensions were monitored by echocardiography and histology, whereas cardiac function was determined by direct intraventricular pressure measurements (dP/dt). Furthermore, matrix metalloproteinases were measured by zymography, and mRNA expression by quantitative PCR. Infarct rupture, which typically occurred at 3-6 days post-MI, was most frequent in 129S6 mice (62%), followed by C57Bl6 (36%), FVB (29%), Swiss (23%), and BalbC (5%). The high incidence of infarct rupture in 129S6 mice was associated with high systolic blood pressure and increased influx of inflammatory cells. Cardiac dilatation was most marked in Swiss mice and least prominent in 129S6 mice. The degree of dilatation was associated with a reduced ejection fraction and decreased dP/dt values at 14 and 28 days post-MI. At day 14 and 28 post-MI, secondary thinning of the infarct area was marked in BalbC, FVB, and Swiss, but absent in C57Bl6 and 129S6 mice. In the latter two groups, this was paralleled by the highest number of myofibroblasts at day 14 post-MI. CONCLUSION: The outcome of infarct healing in mice strongly depends on genetic background. On the basis of our results, we suggest that for studies on infarct rupture, the 129S6 mouse is the background of choice, whereas BalbC and Swiss mice are the preferred models to study infarct thinning post-MI.


Asunto(s)
Rotura Cardíaca Posinfarto/genética , Infarto del Miocardio/genética , Miocardio/patología , Cicatrización de Heridas/genética , Animales , Antihipertensivos/farmacología , Factor Natriurético Atrial/genética , Presión Sanguínea/genética , Modelos Animales de Enfermedad , Ecocardiografía , Regulación Enzimológica de la Expresión Génica , Rotura Cardíaca Posinfarto/patología , Rotura Cardíaca Posinfarto/fisiopatología , Rotura Cardíaca Posinfarto/prevención & control , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Masculino , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Péptido Natriurético Encefálico/genética , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Especificidad de la Especie , Volumen Sistólico/genética , Factores de Tiempo , Función Ventricular Izquierda/genética , Presión Ventricular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...