Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Clin Lab Anal ; 34(9): e23367, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32441412

ABSTRACT

BACKGROUND: Eosinophil levels predict prognosis in ST-segment elevation myocardial infarction (STEMI) patients. Both eosinophils and high-sensitivity C-reactive protein (hs-CRP) play a major role in the acute inflammatory response of myocardial infarction. The purpose of this study was to evaluate eosinophil percentage (EOS%) and hs-CRP as prognostic markers for in-hospital adverse events in STEMI patients undergoing primary percutaneous coronary intervention. METHODS: We retrospectively analyzed the clinical data of 518 patients. Major adverse cardiac events (MACEs) were defined as cardiac rupture, cardiac arrest, malignant arrhythmia, and cardiac death. Based on the receiver operating characteristic (ROC) analysis, all patients were regrouped into 3 groups (None, One, and Two) according to cutoff EOS% value (≤0.3%) and hs-CRP value (>11.8 mg/L). Both Cox regression analyses and the KM (Kaplan-Meier) survival curve were used to examine the prognostic role of combined hs-CRP and EOS% in cardiovascular events. RESULTS: Of the 518 STEMI patients, 50 of them developed MACEs. Patients who developed MACEs had a significantly lower EOS% and higher hs-CRP than patients who remained MACE-free. In the multivariable Cox regression analysis, the highest risk of in-hospital MACEs was constantly observed in patients with a combined low EOS% and elevated hs-CRP. Patients with reduced EOS% and high hs-CRP had significantly higher incidence rates of cardiac rupture (P = .001), cardiac arrest (P = .001), and malignant arrhythmia (P < .001); furthermore, they had the worst cumulative survival compared with the other two groups. CONCLUSION: Combined reduced EOS% and elevated hs-CRP were valuable tools for identifying patients at risk of in-hospital MACEs.


Subject(s)
Biomarkers/analysis , C-Reactive Protein/analysis , Eosinophils/pathology , Heart Rupture, Post-Infarction/diagnosis , Hospital Mortality/trends , Percutaneous Coronary Intervention/adverse effects , ST Elevation Myocardial Infarction/therapy , Aged , Female , Follow-Up Studies , Heart Rupture, Post-Infarction/etiology , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/mortality , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , ST Elevation Myocardial Infarction/pathology , Survival Rate
2.
Am J Physiol Heart Circ Physiol ; 317(3): H581-H596, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31322426

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Innate , Inflammation/immunology , Myocardial Infarction/immunology , Myocardium/immunology , Animals , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Collagen/metabolism , Disease Models, Animal , Female , Fibrosis , Heart Rupture, Post-Infarction/immunology , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Ventricular Remodeling
3.
Cardiovasc Res ; 115(1): 154-167, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29982352

ABSTRACT

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.


Subject(s)
Chemotaxis, Leukocyte , Heart Rupture, Post-Infarction/metabolism , Heat-Shock Proteins/deficiency , Leukocytes/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Ventricular Remodeling , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , HSP27 Heat-Shock Proteins/deficiency , HSP27 Heat-Shock Proteins/genetics , Heart Rupture, Post-Infarction/immunology , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Heat-Shock Proteins/genetics , Leukocytes/immunology , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Rats, Sprague-Dawley , Signal Transduction , Wound Healing
4.
J Mol Cell Cardiol ; 126: 1-12, 2019 01.
Article in English | MEDLINE | ID: mdl-30408466

ABSTRACT

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.


Subject(s)
Heart Rupture, Post-Infarction/metabolism , Muscle Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Gene Deletion , Gene Expression Regulation , Heart Rupture, Post-Infarction/genetics , Heart Ventricles/pathology , Inflammasomes/metabolism , Inflammation/genetics , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Neutrophil Infiltration , Rats
5.
Med Sci Monit ; 24: 6989-7000, 2018 Oct 02.
Article in English | MEDLINE | ID: mdl-30275441

ABSTRACT

BACKGROUND Cardiac rupture often occurs after acute myocardial infarction due to complex and unclear pathogenesis. This study investigated whether metformin increases the incidence of cardiac rupture after myocardial infarction through the AMPK-MTOR/PGC-1α signaling pathway. MATERIAL AND METHODS An acute myocardial infarction (MI) mouse model was established. A series of experiments involving RT-qPCR, Western blot, TUNEL staining, and Masson staining were performed in this study. RESULTS Myocardial infarction occurred, resulting in the cardiac rupture, and the expression level of PGC-1α increased in the cardiac myocardium. Meanwhile, the proportion of myocardial NT-PGC-1α/PGC-1α decreased. The expression level of myocardial PGC-1α in MI mice with cardiac rupture after MI was significantly higher than that in the mice without cardiac rupture, and the ratio of myocardial NT-PGC-1α/PGC-1α was low. In addition, increasing the dose of metformin significantly increased the incidence of cardiac rupture after myocardial infarction in MI mice. High-dose metformin caused cardiac rupture in MI mice. Moreover, high-dose metformin (Met 2.0 nM) reduces the proportion of NT-PGC-1α/PGC-1α in primary cardiomyocytes of SD mice (SD-NRVCs [Neonatal rat ventricular cardiomyocytes]), and its effect was inhibited by Compound C (AMPK inhibitor). Further, after 3 days of treatment with high-dose metformin in MI mice, myocardial fibrin synthesis decreased and fibrosis was significantly inhibited. Meanwhile, cardiomyocyte apoptosis increased significantly. With the increase in metformin concentration, the expression level of myocardial LC3b gradually increased in MI mice, suggesting that metformin enhances the autophagy of cardiomyocytes. CONCLUSIONS These results suggest that metformin increases cardiac rupture after myocardial infarction through the AMPK-MTOR/PGC-1α signaling pathway.


Subject(s)
Heart Rupture, Post-Infarction/chemically induced , Heart Rupture, Post-Infarction/metabolism , Metformin/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Disease Models, Animal , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , TOR Serine-Threonine Kinases/metabolism
6.
PLoS One ; 12(11): e0187894, 2017.
Article in English | MEDLINE | ID: mdl-29121663

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Heart Rupture, Post-Infarction/epidemiology , Macrophages/metabolism , Myocardial Infarction/metabolism , Receptors, Immunologic/deficiency , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Disease Models, Animal , Female , Heart Rupture, Post-Infarction/genetics , Heart Rupture, Post-Infarction/metabolism , Incidence , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/immunology , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptors, Immunologic/genetics , Receptors, Scavenger , Survival Rate
7.
Cardiovasc Res ; 113(5): 475-487, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28339742

ABSTRACT

AIMS: Monocytes/macrophages response plays a key role in post-infarction inflammation that contributes greatly to post-infarction ventricular remodelling and cardiac rupture. Therapeutic targeting of the GABAA receptor, which is enriched in monocytes/macrophages but not expressed in the myocardium, may be possible after myocardial infarction (MI). METHODS AND RESULTS: After MI was induced by ligation of the coronary artery, C57BL/6 mice were intraperitoneally administered with one specific agonist or antagonist of the GABAA receptor (topiramate or bicuculline), in the setting of presence or depletion of monocytes/macrophages. Our data showed that within the first 2 weeks after MI, when monocytes/macrophages dominated, in contrast with bicuculline, topiramate treatment significantly reduced Ly-6Chigh monocyte numbers by regulating splenic monocytopoiesis and promoted foetal derived macrophages preservation and conversion of M1 to M2 or Ly-6Chigh to Ly-6Clow macrophage phenotype in the infarcted heart, though GABAAergic drugs failed to affect M1/M2 or Ly-6Chigh/Ly-6Clow macrophage polarization directly. Accordingly, pro-inflammatory activities mediated by M1 or Ly-6Chigh macrophages were decreased and reparative processes mediated by M2 or Ly-6Clow macrophages were augmented. As a result, post-infarction ventricular remodelling was attenuated, as reflected by reduced infarct size and increased collagen density within infarcts. Echocardiographic indices, mortality and rupture rates were reduced. After depletion of monocytes/macrophages by clodronate liposomes, GABAAergic drugs exhibited no effect on cardiac dysfunction and surrogate clinical outcomes. CONCLUSION: Control of the GABAA receptor activity in monocytes/macrophages can potently modulate post-infarction inflammation. Topiramate emerges as a promising drug, which may be feasible to translate for MI therapy in the future.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Fructose/analogs & derivatives , GABA Agonists/pharmacology , Heart Ventricles/drug effects , Macrophages/drug effects , Monocytes/drug effects , Myocardial Infarction/drug therapy , Myocarditis/prevention & control , Receptors, GABA/drug effects , Animals , Antigens, Ly/metabolism , Collagen/metabolism , Disease Models, Animal , Fibrosis , Fructose/pharmacology , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/physiopathology , Heart Rupture, Post-Infarction/prevention & control , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/metabolism , Myocardium/pathology , Phenotype , Receptors, GABA/metabolism , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Time Factors , Topiramate , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
8.
Cardiovasc Drugs Ther ; 31(2): 145-156, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28204966

ABSTRACT

PURPOSE: Inhibition of the renin-angiotensin system (RAS) is beneficial in patient management after myocardial infarction (MI). However, whether RAS inhibition also provides cardiac protection in the acute phase of MI is unclear. METHODS: Male 129sv mice underwent coronary artery occlusion to induce MI, followed by treatment with losartan (L, 20 and 60 mg/kg), perindopril (P, 2 and 6 mg/kg), amlodipine (20 mg/kg as a BP-lowering agent) or vehicle as control. Drug effects on hemodynamics were examined. Effects of treatments on incidence of cardiac rupture, haematological profile, monocyte and neutrophil population in the spleen and the heart, cardiac leukocyte density, expression of inflammatory genes and activity of MMPs were studied after MI. RESULTS: Incidence of cardiac rupture within 2 weeks was significantly and similarly reduced by both losartan (L) and perindopril (P) in a dose-dependent manner [75% (27/36) in vehicle, 40-45% in low-dose (L 10/22, P 8/20) and 16-20% (L 5/32, P 4/20) in high-dose groups, all P < 0.05]. This action was independent of their BP-lowering action, as amlodipine reduced BP to a similar degree without effect on rupture (70%, 21/30). Compared to the control group, high dose losartan and perindopril decreased counts of white blood cells, neutrophils and lymphocytes (all P < 0.05), and inhibited splenic monocyte and neutrophil release into the circulation. Consequently, monocyte, neutrophil and leukocyte infiltration, inflammatory gene expressions (IL-1ß, IL-6, MMP9, MCP-1, TNF-α and TGFß1) and activity of MMP2 and MMP9 in the infarct tissue were attenuated by losartan and/or perindopril treatment (all P < 0.05). CONCLUSIONS: RAS inhibition by losartan or perindopril prevented cardiac rupture at the acute phase of MI through blockade of splenic release of monocytes and neutrophils and consequently attenuation of systemic and regional inflammatory responses.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , Heart Rupture, Post-Infarction/prevention & control , Inflammation/prevention & control , Losartan/pharmacology , Myocardial Infarction/drug therapy , Myocardium/metabolism , Perindopril/pharmacology , Renin-Angiotensin System/drug effects , Amlodipine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Calcium Channel Blockers/pharmacology , Chemotaxis, Leukocyte/drug effects , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Heart Rupture, Post-Infarction/etiology , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Mice, 129 Strain , Monocytes/drug effects , Monocytes/metabolism , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Spleen/drug effects , Spleen/metabolism , Time Factors
9.
Circ J ; 80(9): 1971-9, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27396441

ABSTRACT

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).


Subject(s)
CD28 Antigens/deficiency , Gene Deletion , Heart Rupture, Post-Infarction/metabolism , Myocardial Infarction/metabolism , Signal Transduction , Ventricular Remodeling , Animals , CD28 Antigens/metabolism , Gene Expression Regulation , Heart Rupture, Post-Infarction/genetics , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure
10.
Am J Physiol Heart Circ Physiol ; 309(11): H1883-93, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26453333

ABSTRACT

The activation of the calpain system is involved in the repair process following myocardial infarction (MI). However, the impact of the inhibition of calpain by calpastatin, its natural inhibitor, on scar healing and left ventricular (LV) remodeling is elusive. Male mice ubiquitously overexpressing calpastatin (TG) and wild-type (WT) controls were subjected to an anterior coronary artery ligation. Mortality at 6 wk was higher in TG mice (24% in WT vs. 44% in TG, P < 0.05) driven by a significantly higher incidence of cardiac rupture during the first week post-MI, despite comparable infarct size and LV dysfunction and dilatation. Calpain activation post-MI was blunted in TG myocardium. In TG mice, inflammatory cell infiltration and activation were reduced in the infarct zone (IZ), particularly affecting M2 macrophages and CD4(+) T cells, which are crucial for scar healing. To elucidate the role of calpastatin overexpression in macrophages, we stimulated peritoneal macrophages obtained from TG and WT mice in vitro with IL-4, yielding an abrogated M2 polarization in TG but not in WT cells. Lymphopenic Rag1(-/-) mice receiving TG splenocytes before MI demonstrated decreased T-cell recruitment and M2 macrophage activation in the IZ day 5 after MI compared with those receiving WT splenocytes. Calpastatin overexpression prevented the activation of the calpain system after MI. It also impaired scar healing, promoted LV rupture, and increased mortality. Defective scar formation was associated with blunted CD4(+) T-cell and M2-macrophage recruitment.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Calcium-Binding Proteins/metabolism , Lymphocyte Activation , Macrophage Activation , Macrophages/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Ventricular Remodeling , Wound Healing , Animals , CD4-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/genetics , Calpain/metabolism , Chemotaxis, Leukocyte , Disease Models, Animal , Enzyme Activation , Genotype , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/immunology , Myocardium/pathology , Phenotype , Time Factors , Up-Regulation , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
11.
J Mol Cell Cardiol ; 88: 55-63, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26393439

ABSTRACT

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.


Subject(s)
Heart Rupture, Post-Infarction/metabolism , Microfilament Proteins/metabolism , Myocardial Infarction/metabolism , Myofibroblasts/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vesicular Transport Proteins/metabolism , Actins/genetics , Actins/metabolism , Amino Acid Substitution , Animals , Animals, Newborn , Cell Proliferation , Collagen/genetics , Collagen/metabolism , Gene Expression Regulation , Gene Knock-In Techniques , Heart Rupture, Post-Infarction/genetics , Heart Rupture, Post-Infarction/mortality , Heart Rupture, Post-Infarction/pathology , Mice , Mice, Knockout , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Myocardial Infarction/genetics , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/pathology , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
12.
Am J Physiol Heart Circ Physiol ; 307(5): H741-51, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25015963

ABSTRACT

Thymosin-ß4 (Tß4) promotes cell survival, angiogenesis, and tissue regeneration and reduces inflammation. Cardiac rupture after myocardial infarction (MI) is mainly the consequence of excessive regional inflammation, whereas cardiac dysfunction after MI results from a massive cardiomyocyte loss and cardiac fibrosis. It is possible that Tß4 reduces the incidence of cardiac rupture post-MI via anti-inflammatory actions and that it decreases adverse cardiac remodeling and improves cardiac function by promoting cardiac cell survival and cardiac repair. C57BL/6 mice were subjected to MI and treated with either vehicle or Tß4 (1.6 mg·kg(-1)·day(-1) ip via osmotic minipump) for 7 days or 5 wk. Mice were assessed for 1) cardiac remodeling and function by echocardiography; 2) inflammatory cell infiltration, capillary density, myocyte apoptosis, and interstitial collagen fraction histopathologically; 3) gelatinolytic activity by in situ zymography; and 4) expression of ICAM-1 and p53 by immunoblot analysis. Tß4 reduced cardiac rupture that was associated with a decrease in the numbers of infiltrating inflammatory cells and apoptotic myocytes, a decrease in gelatinolytic activity and ICAM-1 and p53 expression, and an increase in the numbers of CD31-positive cells. Five-week treatment with Tß4 ameliorated left ventricular dilation, improved cardiac function, markedly reduced interstitial collagen fraction, and increased capillary density. In a murine model of acute MI, Tß4 not only decreased mortality rate as a result of cardiac rupture but also significantly improved cardiac function after MI. Thus, the use of Tß4 could be explored as an alternative therapy in preventing cardiac rupture and restoring cardiac function in patients with MI.


Subject(s)
Heart Rupture, Post-Infarction/prevention & control , Thymosin/therapeutic use , Ventricular Function, Left , Animals , Apoptosis , Echocardiography , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/physiopathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Thymosin/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
J Mol Cell Cardiol ; 69: 32-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24508700

ABSTRACT

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.


Subject(s)
Heart Rupture, Post-Infarction/immunology , Inflammation/immunology , Intramolecular Oxidoreductases/physiology , Leukocytes/immunology , Macrophage Migration-Inhibitory Factors/physiology , Myocardial Infarction/immunology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Leukocytes/metabolism , Leukocytes/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
PLoS One ; 8(10): e76206, 2013.
Article in English | MEDLINE | ID: mdl-24098445

ABSTRACT

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.


Subject(s)
Macrophage Activation , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/metabolism , Myocardial Infarction/metabolism , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Heart Rupture, Post-Infarction/immunology , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Humans , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukin-6/blood , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/blood , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Middle Aged , Models, Biological , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Risk Factors , Time Factors
15.
Dis Markers ; 31(5): 259-65, 2011.
Article in English | MEDLINE | ID: mdl-22048267

ABSTRACT

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.


Subject(s)
Heart Rupture, Post-Infarction/metabolism , Heart Ventricles/pathology , Immunity, Innate/genetics , MicroRNAs/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Gene Expression , Genetic Association Studies , Heart Rupture, Post-Infarction/immunology , Heart Rupture, Post-Infarction/pathology , Heart Ventricles/immunology , Heart Ventricles/metabolism , Humans , Inflammation/metabolism , Male , MicroRNAs/genetics , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Neutrophil Infiltration/genetics , Young Adult
16.
Gend Med ; 8(4): 239-51, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21664194

ABSTRACT

OBJECTIVE: The objective of this study was to test the hypothesis that gender has a significant impact on cardiac inflammation, fibrosis, and survival after myocardial infarction (MI) in a murine model of left coronary artery ligation (CAL) by investigating the underlying cellular-molecular mechanisms. METHODS: Age-matched male and female mice were randomly assigned into 6 groups: sham-operated males, sham-operated females, intact males with CAL, intact females with CAL, castrated males with CAL, and oophorectomized females with CAL. The animals were sacrificed 14 days postoperatively. The hearts from each group were harvested for morphologic studies (n = 6) (infarct and fibrotic area, inflammatory cell markers CD40 and CD68) and mRNA expression analyses (n = 6) of pro- and antiinflammatory molecules, including matrix metalloproteinase (MMP)-9, plasminogen activator inhibitor (PAI)-1, interleukin (IL)-10, transforming growth factor (TGF)-ß, and endothelial nitric oxide synthase (eNOS). RESULTS: Intact males with CAL had significantly lower 14-day survival compared with intact females with CAL. Similarly, the infarct areas in intact males with CAL were largest compared with other CAL animals. The fibrotic area was also larger in intact males with CAL than in intact females with CAL. Numbers of CD40(+)/CD68(+) cells and MMP-9 expression were higher in intact males with CAL than in intact females with CAL and castrated males with CAL. IL-10, eNOS, and TGF-ß were significantly suppressed in oophorectomized females with CAL compared with intact females with CAL. Intact females with CAL and castrated males with CAL exhibited notably enhanced post-MI PAI-1 expression. CONCLUSIONS: Male gender (compared with female) may be an unfavorable prognostic factor after MI in terms of enhanced inflammation and fibrosis in a murine model. Although castration seemed to be significantly antiinflammatory and antifibrotic after MI, oophorectomy had no significant impact on survival, suggesting that factors other than estrogen may account for favorable outcome after MI in the female gender. Furthermore, enhanced postinfarct PAI-1 expression in castrated and female mice may contribute to suppressed MMP-9 expression and survival advantage.


Subject(s)
Heart Rupture/metabolism , Inflammation/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Plasminogen Inactivators/metabolism , Animals , Female , Heart Rupture/pathology , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Inflammation/pathology , Interleukin-10/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardium/pathology , Nitric Oxide Synthase Type III/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Transforming Growth Factor beta/metabolism
17.
Am J Physiol Heart Circ Physiol ; 301(2): H497-505, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21602472

ABSTRACT

Secreted protein, acidic, and rich in cysteine (SPARC) is a matricellular protein that functions in the extracellular processing of newly synthesized collagen. Collagen deposition to form a scar is a key event following a myocardial infarction (MI). Because the roles of SPARC in the early post-MI setting have not been defined, we examined age-matched wild-type (WT; n=22) and SPARC-deficient (null; n=25) mice at day 3 post-MI. Day 0 WT (n=28) and null (n=20) mice served as controls. Infarct size was 52 ± 2% for WT and 47 ± 2% for SPARC null (P=NS), indicating that the MI injury was comparable in the two groups. By echocardiography, WT mice increased end-diastolic volumes from 45 ± 2 to 83 ± 5 µl (P < 0.05). SPARC null mice also increased end-diastolic volumes but to a lesser extent than WT (39 ± 3 to 63 ± 5 µl; P < 0.05 vs. day 0 controls and vs. WT day 3 MI). Ejection fraction fell post-MI in WT mice from 57 ± 2 to 19 ± 1%. The decrease in ejection fraction was attenuated in the absence of SPARC (65 ± 2 to 28 ± 2%). Fibroblasts isolated from SPARC null left ventricle (LV) showed differences in the expression of 22 genes encoding extracellular matrix and adhesion molecule genes, including fibronectin, connective tissue growth factor (CTGF; CCN2), matrix metalloproteinase-3 (MMP-3), and tissue inhibitor of metalloproteinase-2 (TIMP-2). The change in fibroblast gene expression levels was mirrored in tissue protein extracts for fibronectin, CTGF, and MMP-3 but not TIMP-2. Combined, the results of this study indicate that SPARC deletion preserves LV function at day 3 post-MI but may be detrimental for the long-term response due to impaired fibroblast activation.


Subject(s)
Extracellular Matrix Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Osteonectin/metabolism , Ventricular Function, Left , Ventricular Remodeling , Analysis of Variance , Animals , Blotting, Western , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Female , Fibroblasts/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardium/pathology , Oligonucleotide Array Sequence Analysis , Osteonectin/deficiency , Osteonectin/genetics , Stroke Volume , Time Factors , Ultrasonography , Ventricular Remodeling/genetics
18.
J Mol Cell Cardiol ; 50(3): 426-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20807543

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is an oxidant-sensitive protease inhibitor that is inactivated by oxidation and has a critical role in ventricular remodeling post myocardial infarction (MI). PAI-1 knockout (KO) mice die within 7days of myocardial infarction post MI due to increased plasmin activity leading to ventricular rupture. The goal of this study was to assess the relevant pathways of leukocyte-derived oxidants post MI that alter PAI-1 activity. Transplantation of wild-type (WT) bone marrow into PAI-1 null mice prolonged survival after MI (WT marrow: 41.66% vs. PAI-1 KO marrow: 0% in PAI-1 KO mice at day 7 (p<0.02). To determine relevant enzyme systems, we transplanted marrow from mice with specific deletions relevant to leukocyte-derived oxidants (NAD(P)H oxidase, iNOS, myeloperoxidase (MPO)) to determine which deletion controls PAI-1 oxidative inactivation and prolongs survival. MI was induced by ligation of the left anterior descending artery (LAD) and the incidence of cardiac rupture was monitored. PAI-1 KO transplanted with MPO KO, or iNOS KO bone marrow died within 9 days after MI. PAI-1 KO mice transplanted with p47(phox) KO marrow exhibited prolonged survival 21 days after MI (30% survival, p<0.03, n=10) compared to WT marrow (8.3%, n=12). Three days after MI, PAI-1 KO mice transplanted with p47(phox) KO marrow had increased PAI-1 activity and decreased nitration of PAI-1 in myocardial tissue compared to PAI-1 KO mice transplanted with WT marrow. These data suggest that modulating O(2)(•-) generation by NAD(P)H oxidase appears to be a therapeutically relevant target for increasing myocardial PAI-1 levels after MI, whereas downstream enzymes like MPO and iNOS may not be.


Subject(s)
Heart Rupture, Post-Infarction/metabolism , Heart Rupture/metabolism , Heart Ventricles/pathology , Leukocytes/metabolism , NADPH Oxidases/blood , Plasminogen Activator Inhibitor 1/metabolism , Animals , Bone Marrow Transplantation , Heart Rupture/enzymology , Heart Rupture/pathology , Heart Rupture, Post-Infarction/blood , Heart Rupture, Post-Infarction/enzymology , Heart Rupture, Post-Infarction/pathology , Leukocytes/enzymology , Male , Mice , Mice, Knockout , NADPH Oxidases/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Peroxidase/metabolism , Plasminogen Activator Inhibitor 1/blood , Ventricular Remodeling/physiology
19.
Cardiol Rev ; 17(6): 293-9, 2009.
Article in English | MEDLINE | ID: mdl-19829180

ABSTRACT

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.


Subject(s)
Heart Rupture, Post-Infarction/genetics , Models, Animal , Models, Genetic , Animals , Collagen/genetics , Collagen/metabolism , Heart Rupture, Post-Infarction/metabolism , Humans , Mice , Proteomics
20.
J Exp Med ; 206(1): 113-23, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19103879

ABSTRACT

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


Subject(s)
Heart Rupture, Post-Infarction/metabolism , Myocardial Infarction/metabolism , Osteonectin/deficiency , Animals , Calcium-Binding Proteins/genetics , Extracellular Matrix Proteins/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Granulation Tissue/drug effects , Granulation Tissue/metabolism , Granulation Tissue/pathology , Heart/physiopathology , Heart Rupture, Post-Infarction/physiopathology , Heart Rupture, Post-Infarction/prevention & control , Hemodynamics/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Osteonectin/genetics , Osteonectin/physiology , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Smad2 Protein/genetics , Survival Analysis , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...