Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 317(3): H581-H596, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31322426

RESUMEN

The adaptive immune response is key for cardiac wound healing post-myocardial infarction (MI) despite low T-cell numbers. We hypothesized that CD8+ T-cells regulate the inflammatory response, leading to decreased survival and cardiac function post-MI. We performed permanent occlusion of the left anterior descending coronary artery on C57BL/6J and CD8atm1mak mice (deficient in functional CD8+ T-cells). CD8atm1mak mice had increased survival at 7 days post-MI compared with that of the wild-type (WT) and improved cardiac physiology at day 7 post-MI. Despite having less mortality, 100% of the CD8atm1mak group died because of cardiac rupture compared with only 33% of the WT. Picrosirius red staining and collagen immunoblotting indicated an acceleration of fibrosis in the infarct area as well as remote area in the CD8atm1mak mice; however, this increase was due to elevated soluble collagen implicating poor scar formation. Plasma and tissue inflammation were exacerbated as indicated by higher levels of Cxcl1, Ccl11, matrix metalloproteinase (MMP)-2, and MMP-9. Immunohistochemistry and flow cytometry indicated that the CD8atm1mak group had augmented numbers of neutrophils and macrophages at post-MI day 3 and increased mast cell markers at post-MI day 7. Cleavage of tyrosine-protein kinase MER was increased in the CD8atm1mak mice, resulting in delayed removal of necrotic tissue. In conclusion, despite having improved cardiac physiology and overall survival, CD8atm1mak mice had increased innate inflammation and poor scar formation, leading to higher incidence of cardiac rupture. Our data suggest that the role of CD8+ T-cells in post-MI recovery may be both beneficial and detrimental to cardiac remodeling and is mediated via a cell-specific mechanism.NEW & NOTEWORTHY We identified new mechanisms implicating CD8+ T-cells as regulators of the post-myocardial infarction (MI) wound healing process. Mice without functional CD8+ T-cells had improved cardiac physiology and less mortality 7 days post MI compared with wild-type animals. Despite having better overall survival, animals lacking functional CD8+ T-cells had delayed removal of necrotic tissue, leading to poor scar formation and increased cardiac rupture, suggesting that CD8+ T-cells play a dual role in the cardiac remodeling process.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Innata , Inflamación/inmunología , Infarto del Miocardio/inmunología , Miocardio/inmunología , Animales , Antígenos CD8/genética , Linfocitos T CD8-positivos/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/metabolismo , Rotura Cardíaca Posinfarto/patología , Rotura Cardíaca Posinfarto/fisiopatología , Inflamación/metabolismo , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal , Remodelación Ventricular
2.
Cardiovasc Res ; 115(1): 154-167, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982352

RESUMEN

Aims: Inadequate healing after myocardial infarction (MI) leads to heart failure and fatal ventricular rupture, while optimal healing requires timely induction and resolution of inflammation. This study tested the hypothesis that heat shock protein B1 (HSPB1), which limits myocardial inflammation during endotoxemia, modulates wound healing after MI. Methods and results: To test this hypothesis, cardiomyocyte-specific HSPB1 knockout (Hspb1-/-) mice were generated using the Cre-LoxP recombination system. MI was induced by ligation of the left anterior descending coronary artery in Hspb1-/- and wild-type (WT) littermates. HSPB1 was up-regulated in cardiomyocytes of WT animals in response to MI, and deficiency of cardiomyocyte HSPB1 increased MI-induced cardiac rupture and mortality within 21 days after MI. Serial echocardiography showed more aggravated remodelling and cardiac dysfunction in Hspb1-/- mice than in WT mice at 1, 3, and 7 days after MI. Decreased collagen deposition and angiogenesis, as well as increased MMP2 and MMP9 activity, were also observed in Hspb1-/- mice compared with WT controls after MI, using immunofluorescence, polarized light microscopy, and zymographic analyses. Notably, Hspb1-/- hearts exhibited enhanced and prolonged leucocyte infiltration, enhanced expression of inflammatory cytokines, and enhanced TLR4/MyD88/NFκB activation compared with WT controls after MI. In-depth molecular analyses in both mice and primary cardiomyocytes demonstrated that cardiomyocyte-specific knockout of HSPB1 increased nuclear factor-κB (NFκB) activation, which promoted the expression of proinflammatory mediators. This led to increased leucocyte recruitment, thereby to excessive inflammation, ultimately resulting in adverse remodelling, cardiac dysfunction, and cardiac rupture following MI. Conclusion: These data suggest that HSPB1 acts as a negative regulator of NFκB-mediated leucocyte recruitment and the subsequent inflammation in cardiomyocytes. Cardiomyocyte HSPB1 is required for wound healing after MI and could be a target for myocardial repair in MI patients.


Asunto(s)
Quimiotaxis de Leucocito , Rotura Cardíaca Posinfarto/metabolismo , Proteínas de Choque Térmico/deficiencia , Leucocitos/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Remodelación Ventricular , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Choque Térmico HSP27/deficiencia , Proteínas de Choque Térmico HSP27/genética , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/patología , Rotura Cardíaca Posinfarto/fisiopatología , Proteínas de Choque Térmico/genética , Leucocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Ratas Sprague-Dawley , Transducción de Señal , Cicatrización de Heridas
3.
FASEB J ; 32(1): 254-264, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860151

RESUMEN

Phagocytosis after myocardial infarction (MI) is a prerequisite to cardiac repair. Recruited monocytes clear necrotic cardiomyocytes and differentiate into cardiac macrophages. Some studies have linked apoptotic cell receptors on cardiac macrophages to tissue repair; however, the contribution of precursor monocyte phagocytic receptors, which are the first to interact with the cardiac parenchyma, is unclear. The scavenger receptor cluster of differentiation (CD)36 protein was detected on cardiac Ly6cHI monocytes, and bone marrow-derived Cd36 was essential for both early phagocytosis of dying cardiomyocytes and for smaller infarct sizes in female and male mice after permanent coronary ligation. Cd36 deficiency led to reduced expression of phagocytosis receptor Mertk and nuclear receptor Nr4a1 in cardiac macrophages, the latter previously shown to be required for phagocyte survival. Nr4a1 was required for phagocytosis-induced Mertk expression, and Nr4a1 protein directly bound to Mertk gene regulatory elements. To test the overall contribution of the Cd36-Mertk axis, MI was induced in Cd36-/- Mertk-/- double-knockout mice and led to increases in myocardial rupture. These data implicate monocyte CD36 in the mitigation of early infarct size and transition to Mertk-dependent macrophage function. Increased myocardial rupture in the absence of both Cd36 and Mertk underscore the physiologic significance of phagocytosis during tissue injury.-Dehn, S., Thorp, E. B. Myeloid receptor CD36 is required for early phagocytosis of myocardial infarcts and induction of Nr4a1-dependent mechanisms of cardiac repair.


Asunto(s)
Antígenos CD36/inmunología , Infarto del Miocardio/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Fagocitosis/inmunología , Animales , Apoptosis/inmunología , Antígenos CD36/deficiencia , Antígenos CD36/genética , Gasto Cardíaco , Células Cultivadas , Femenino , Rotura Cardíaca Posinfarto/etiología , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/patología , Humanos , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/agonistas , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo
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.
J Mol Cell Cardiol ; 69: 32-42, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24508700

RESUMEN

Myocardial infarction (MI) provokes regional inflammation which facilitates the healing, whereas excessive inflammation leads to adverse cardiac remodelling. Our aim was to determine the role of macrophage migration inhibitory factor (MIF) in inflammation and cardiac remodelling following MI. Wild type (WT) or global MIF deficient (MIFKO) mice were subjected to coronary artery occlusion. Compared to WT mice, MIFKO mice had a significantly lower incidence of post-MI cardiac rupture (27% vs. 53%) and amelioration of cardiac remodelling. These were associated with suppressed myocardial leukocyte infiltration, inflammatory mediators' expression, and reduced activity of MMP-2, MMP-9, p38 and JNK MAPK. Infarct myocardium-derived or exogenous MIF mediated macrophage chemotaxis in vitro that was suppressed by inhibition of p38 MAPK or NF-κB. To further dissect the role of MIF derived from different cellular sources in post-MI cardiac remodelling, we generated chimeric mice with MIF deficiency either in bone marrow derived-cells (WT(KO)) or in somatic-cells (KO(WT)). Compared to WT and KO(WT) mice, WT(KO) mice had reduced rupture risk and ameliorated cardiac remodelling, associated with attenuated regional leukocyte infiltration and expression of inflammatory mediators. In contrast, KO(WT) mice had delayed healing and enhanced expression of M1 macrophage markers, but diminished expression of M2 markers during the early healing phase. In conclusion, global MIF deletion protects the heart from post-infarct cardiac rupture and remodelling through suppression of leukocyte infiltration and inflammation. Leukocyte-derived MIF promotes inflammatory responses after MI, whereas cardiac-derived MIF affects early but not ultimate healing process.


Asunto(s)
Rotura Cardíaca Posinfarto/inmunología , Inflamación/inmunología , Oxidorreductasas Intramoleculares/fisiología , Leucocitos/inmunología , Factores Inhibidores de la Migración de Macrófagos/fisiología , Infarto del Miocardio/inmunología , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Rotura Cardíaca Posinfarto/metabolismo , Rotura Cardíaca Posinfarto/patología , Técnicas para Inmunoenzimas , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
PLoS One ; 8(10): e76206, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098445

RESUMEN

OBJECTIVES: Macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine, has been implicated in the pathogenesis of multiple inflammatory disorders. We determined changes in circulating MIF levels, explored the cellular source of MIF, and studied the role of MIF in mediating inflammatory responses following acute myocardial infarction (MI). METHODS AND RESULTS: We recruited 15 patients with MI, 10 patients with stable angina and 10 healthy volunteers and measured temporal changes of MIF in plasma. Expression of MIF, matrix metalloproteinase-9 (MMP-9) and interleukin-6 (IL-6) in cultured peripheral blood mononuclear cells (PBMCs) and the media were measured by ELISA or real-time PCR. Compared to controls, plasma levels of MIF and IL-6 were significantly elevated at admission and 72 h post-MI. In contrast, expression of MIF, MMP-9 and IL-6 by PBMCs from MI patients was unchanged at admission, but significantly increased at 72 h. Addition of MIF activated cultured PBMCs by upregulating expression of inflammatory molecules and also synergistically enhanced stimulatory action of IL-1ß which were inhibited by anti-MIF interventions. In a mouse MI model we observed similar changes in circulating MIF as seen in patients, with reciprocal significant increases in plasma MIF and reduction of MIF content in the infarct myocardium at 3 h after MI. MIF content in the infarct myocardium was restored at 72 h post-MI and was associated with robust macrophage infiltration. Further, anti-MIF intervention significantly reduced inflammatory cell infiltration and expression of monocyte chemoattractant protein-1 at 24 h and incidence of cardiac rupture in mice post-MI. CONCLUSION: MI leads to a rapid release of MIF from the myocardium into circulation. Subsequently MIF facilitates PBMC production of pro-inflammatory mediators and myocardial inflammatory infiltration. Attenuation of these events, and post-MI cardiac rupture, by anti-MIF interventions suggests that MIF could be a potential therapeutic target following MI.


Asunto(s)
Activación de Macrófagos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos/metabolismo , Infarto del Miocardio/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/farmacología , Modelos Animales de Enfermedad , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/metabolismo , Rotura Cardíaca Posinfarto/patología , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-6/sangre , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/sangre , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Modelos Biológicos , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Factores de Riesgo , Factores de Tiempo
7.
Arterioscler Thromb Vasc Biol ; 32(9): 2206-13, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22796582

RESUMEN

OBJECTIVE: Leukocyte infiltration in ischemic areas is a hallmark of myocardial infarction, and overwhelming infiltration of innate immune cells has been shown to promote adverse remodeling and cardiac rupture. Recruitment of inflammatory cells in the ischemic heart depends highly on the family of CC-chemokines and their receptors. Here, we hypothesized that the chemokine decoy receptor D6, which specifically binds and scavenges inflammatory CC-chemokines, might limit inflammation and adverse cardiac remodeling after infarction. METHODS AND RESULTS: D6 was expressed in human and murine infarcted myocardium. In a murine model of myocardial infarction, D6 deficiency led to increased chemokine (C-C motif) ligand 2 and chemokine (C-C motif) ligand 3 levels in the ischemic heart. D6-deficient (D6(-/-)) infarcts displayed increased infiltration of pathogenic neutrophils and Ly6Chi monocytes, associated with strong matrix metalloproteinase-9 and matrix metalloproteinase-2 activities in the ischemic heart. D6(-/-) mice were cardiac rupture prone after myocardial infarction, and functional analysis revealed that D6(-/-) hearts had features of adverse remodeling with left ventricle dilation and reduced ejection fraction. Bone marrow chimera experiments showed that leukocyte-borne D6 had no role in this setting, and that leukocyte-specific chemokine (C-C motif) receptor 2 deficiency rescued the adverse phenotype observed in D6(-/-) mice. CONCLUSIONS: We show for the first time that the chemokine decoy receptor D6 limits CC-chemokine-dependent pathogenic inflammation and is required for adequate cardiac remodeling after myocardial infarction.


Asunto(s)
Inflamación/prevención & control , Infarto del Miocardio/inmunología , Miocardio/inmunología , Receptores CCR10/metabolismo , Receptores de Quimiocina/metabolismo , Remodelación Ventricular , Animales , Antígenos Ly/metabolismo , Trasplante de Médula Ósea , Quimiocina CCL2/metabolismo , Quimiocina CCL3/metabolismo , Quimiotaxis , Modelos Animales de Enfermedad , Genotipo , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/patología , Humanos , Hipertrofia Ventricular Izquierda/inmunología , Hipertrofia Ventricular Izquierda/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Infiltración Neutrófila , Neutrófilos/inmunología , Fenotipo , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Transducción de Señal , Volumen Sistólico , Ultrasonografía , Función Ventricular Izquierda , Receptor de Quimiocina D6
8.
Dis Markers ; 31(5): 259-65, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22048267

RESUMEN

MicroRNAs are non-coding RNAs, functionioning as post-transcriptional regulators of gene expression. Some microRNAs have been demonstrated to play a role in regulation of innate immunity. After myocardial infarction (MI), innate immunity is activated leading to an acute inflammatory reaction. There is evidence that an intense inflammatory reaction might contribute to the development of ventricular rupture (VR) after MI. Using real-time PCR, we analysed the expression of miR-146a, miR-150, and miR-155 in autopsy samples of infarcted heart tissue from 50 patients with MI (23 with VR and 27 without VR). An altered expression of all three microRNAs was found in MI compared to the normal hearts. Comparing MI patients with VR and those without VR, we found miR-146a up-regulation, and miR-150 and miR-155 down-regulation in patients with VR. In conclusion, our study demonstrated an altered expression of miR-146a, miR-150, and miR-155 in MI compared to the normal hearts. These microRNAs are involved in regulation of the innate immunity. Differential expression of these microRNAs in MI patients with VR in comparison to those without VR provides further evidence that innate immunity resulting in an intense inflammatory reaction plays an important role in the pathogenesis of VR after MI in humans.


Asunto(s)
Rotura Cardíaca Posinfarto/metabolismo , Ventrículos Cardíacos/patología , Inmunidad Innata/genética , MicroARNs/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Expresión Génica , Estudios de Asociación Genética , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/patología , Ventrículos Cardíacos/inmunología , Ventrículos Cardíacos/metabolismo , Humanos , Inflamación/metabolismo , Masculino , MicroARNs/genética , Persona de Mediana Edad , Miocardio/metabolismo , Miocardio/patología , Infiltración Neutrófila/genética , Adulto Joven
9.
Arterioscler Thromb Vasc Biol ; 31(4): 834-41, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21252067

RESUMEN

OBJECTIVE: The goal of this study was to investigate the role of platelets in systemic and cardiac inflammatory responses and the development of postinfarct ventricular complications, as well as the efficacy of antiplatelet interventions. METHODS AND RESULTS: Using a mouse myocardial infarction (MI) model, we determined platelet accumulation and severity of inflammation within the infarcted myocardium by immunohistochemistry and biochemical assays, analyzed peripheral blood platelet-leukocyte conjugation using flow cytometry, and tested antiplatelet interventions, including thienopyridines and platelet depletion. Platelets accumulated within the infarcted region early post-MI and colocalized with inflammatory cells. MI evoked early increase in circulating platelet-leukocyte conjugation mediated by P-selectin/P-selectin glycoprotein ligand-1. Antiplatelet interventions inhibited platelet-leukocyte conjugation in peripheral blood, inflammatory infiltration, content of matrix metalloproteinases or plasminogen activation, and expression of inflammatory mediators in the infarcted myocardium (all P<0.05) and lowered rupture incidence (P<0.01). Clopidogrel therapy alleviated the extent of chronic ventricular dilatation by serial echocardiography. CONCLUSIONS: Platelets play a pivotal role in promoting systemic and cardiac inflammatory responses post-MI. Platelets accumulate within the infarcted myocardium, contributing to regional inflammation, ventricular remodeling, and rupture. Antiplatelet therapy reduces the severity of inflammation and risk of post-MI complications, demonstrating a previously unrecognized protective action.


Asunto(s)
Plaquetas/metabolismo , Rotura Cardíaca Posinfarto/etiología , Mediadores de Inflamación/sangre , Inflamación/etiología , Infarto del Miocardio/complicaciones , Miocardio/metabolismo , Activación Plaquetaria , Remodelación Ventricular , Animales , Antiinflamatorios/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Rotura Cardíaca Posinfarto/sangre , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/patología , Rotura Cardíaca Posinfarto/prevención & control , Inmunohistoquímica , Inflamación/sangre , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Leucocitos/inmunología , Masculino , Glicoproteínas de Membrana/sangre , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/sangre , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Miocardio/inmunología , Miocardio/patología , Selectina-P/sangre , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Factores de Tiempo , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Remodelación Ventricular/efectos de los fármacos
10.
Int J Cardiol ; 143(1): 20-8, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-19195725

RESUMEN

BACKGROUND/OBJECTIVES: Infarct size (IS) is a determinant of pathophysiological events after myocardial infarction (MI), but its relation to the risk of cardiac rupture remains undefined. METHODS: MI was induced in 129sv and C57Bl/6 mice. Left ventricular (LV) remodelling was examined by echocardiography prior to the onset of rupture. Changes in muscle tensile strength and expression of inflammatory factors were determined. Autopsy was performed and IS measured. RESULTS: Rupture incidence was higher in 129sv than C57Bl/6 mice (62% vs. 33%, P<0.001). Rupture occurred in mice with IS over a threshold, which was smaller in 129sv than C57Bl/6 mice (20% vs. 30%). 129sv mice with IS>30% had a higher incidence of rupture than those with IS 20-30%. Echocardiography revealed IS-dependent LV remodelling and dysfunction and 129sv mice had a better-preserved function compared with C57Bl/6 counterparts. 129sv but not C57Bl/6 mice that subsequently developed rupture showed more severe regional dysfunction and remodelling compared with IS-matched non-ruptured hearts. Tensile strength of the infarcted myocardium was reduced significantly, which was IS-related. 129sv mice had higher expression levels of inflammatory mediators in the infarcted myocardium or circulating inflammatory cells, underlying the higher risk of rupture in this strain than C57Bl/6. CONCLUSIONS: A critical IS level is necessary for post-MI rupture and IS correlates with the reduction in muscle tensile strength. Strain differences exist in global function and regional or systemic inflammation that explain the different risk of rupture or heart failure between strains. Limiting IS or minimizing inflammation would lower the risk of ventricular rupture.


Asunto(s)
Rotura Cardíaca Posinfarto , Infarto del Miocardio , Miocarditis , Índice de Severidad de la Enfermedad , Animales , Oclusión Coronaria/diagnóstico por imagen , Oclusión Coronaria/epidemiología , Oclusión Coronaria/cirugía , Modelos Animales de Enfermedad , Expresión Génica/inmunología , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/epidemiología , Insuficiencia Cardíaca/inmunología , Rotura Cardíaca Posinfarto/diagnóstico por imagen , Rotura Cardíaca Posinfarto/epidemiología , Rotura Cardíaca Posinfarto/inmunología , Incidencia , Mediadores de Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/epidemiología , Infarto del Miocardio/inmunología , Miocarditis/diagnóstico por imagen , Miocarditis/epidemiología , Miocarditis/inmunología , Factores de Riesgo , Resistencia a la Tracción , Ultrasonografía , Remodelación Ventricular/inmunología
11.
Cardiovasc Pathol ; 14(5): 247-50, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16168897

RESUMEN

INTRODUCTION: Experimental studies have shown that neutrophils might play an important role in the pathogenesis of ischemic and reperfusion injury in myocardial infarction (MI). Our aim was to compare histologic characteristics of MI with and without rupture of the free wall (RFW), with emphasis on the density of interstitial neutrophil infiltration. METHODS: Autopsy samples of infarcted heart tissue from 110 patients with MI (50 with and 60 without RFW) were included. On the basis of histologic changes and clinical data, all cases were divided into three groups according to the duration of MI (

Asunto(s)
Rotura Cardíaca Posinfarto/inmunología , Infarto del Miocardio/inmunología , Infiltración Neutrófila/inmunología , Anciano , Femenino , Rotura Cardíaca Posinfarto/patología , Humanos , Inmunohistoquímica , Antígeno Lewis X/metabolismo , Masculino , Infarto del Miocardio/patología , Neutrófilos/inmunología
12.
Life Sci ; 74(12): 1561-72, 2004 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-14729404

RESUMEN

We previously found that male mice with myocardial infarction (MI) had a high rate of cardiac rupture, which generally occurred at 3 to 5 days after MI. Since matrix metalloproteinases (MMPs) play an important role in infarct healing, tissue repair and extracellular matrix (ECM) remodeling post-MI, we studied the temporal relationship of MMP expression and inflammatory response to cardiac rupture after acute MI. Male C57BL/6J mice were subjected to MI (induced by ligating the left anterior descending coronary artery) and killed 1, 2, 4, 7 or 14 days after MI. MMP-2 and MMP-9 activity in the heart were measured by zymography. Collagen content was measured by hydroxyproline assay. We found that after MI, MMP-9 activity increased as early as 1 day and reached a maximum by 2-4 days, associated with a similar increase in neutrophil and macrophage infiltration in the infarct area. MMP-2 started to increase rapidly within 4 days, reaching a maximum by 7 days and remaining high even at 14 days. Intense macrophage infiltration appeared by 4 days after MI and then gradually decreased within 7 to 14 days. Collagen content was unchanged until 4 days after MI, at which point it increased and remained high thereafter. Our data suggest that in mice, overexpression of MMP-2 and MMP-9 (possibly expressed mainly by neutrophils and macrophages) may lead to excessive ECM degradation in the early phase of MI, impairing infarct healing and aggravating early remodeling which in turn causes cardiac rupture.


Asunto(s)
Rotura Cardíaca Posinfarto/enzimología , Rotura Cardíaca Posinfarto/inmunología , Inflamación/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Biomarcadores , Colágeno/metabolismo , Rotura Cardíaca , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Neutrófilos/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA