Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Crit Care ; 12(1): R2, 2008.
Article in English | MEDLINE | ID: mdl-18173852

ABSTRACT

INTRODUCTION: Cardiomyocytes exposed to inflammatory processes express intracellular adhesion molecule-1 (ICAM-1). We investigated whether fibrinogen and fibrinogen degradation products, including D-dimer, could alter cardiomyocyte contractile function through interaction with ICAM-1 found on inflamed cardiomyocytes. METHODS: In vivo, rats were injected with endotoxin to model systemic inflammation, whereas isolated rat cardiomyocytes were treated with tumor necrosis factor-alpha to model the inflammatory environment seen following exposure to bacterial products such as lipopolysaccharide. RESULTS: In vivo, endotoxin administration profoundly decreased cardiac contractile function associated with a large increase in intracardiac ICAM-1 and perivascular fibrinogen. Confocal microscopy with double-staining of isolated rat cardiomyocytes demonstrated colocalization of ICAM-1 and fibrinogen. This interaction was disrupted through pre-treatment of the cells with an ICAM-1-blocking antibody. Functionally, isolated rat cardiomyocyte preparations exhibited decreased fractional shortening when incubated with fibrinogen, and through the use of synthetic peptides, we determined that residues 117-133 of the fibrinogen gamma chain are responsible for this interaction with ICAM-1. Despite having crosslinked gamma chains, D-dimer retained the ability to decrease cardiomyocyte contractility. CONCLUSION: Site 117-133 of the fibrinogen gamma chain is able to depress cardiomyocyte contractility through binding ICAM-1.


Subject(s)
Endotoxins/pharmacology , Fibrinogen/pharmacology , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Animals , Fibrinogen/metabolism , Intercellular Adhesion Molecule-1/physiology , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
2.
Cardiovasc Res ; 72(1): 134-42, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16934241

ABSTRACT

OBJECTIVE: Binding of ICAM-1 expressed on cardiomyocytes decreases cardiomyocyte contractility in vitro by altering the intracellular Ca2+ transient. We tested the hypothesis that signaling via ICAM-1 contributes to decreased left ventricular contractility in an in vivo model of systemic inflammation. METHODS: C57B6 wild-type mice and ICAM-1 knock-out mice were treated with intraperitoneal lipopolysaccharide (LPS) then left ventricular contractility was measured 6 h later using a volume-conductance micromanometer catheter. We repeated this experiment in chimeric mice lacking ICAM-1 expression in bone marrow-derived cells (M-) and/or lacking ICAM-1 expression in the heart and other tissues (H-). RESULTS: In C57B6 wild-type mice LPS injection significantly increased cardiac ICAM-1 expression and decreased in vivo measures of left ventricular contractility (end-systolic elastance, Ees decreased 58 +/- 4%, p < 0.05, [dP/dtmax]/EDV decreased 60 +/- 6%, p < 0.05). Cyclophosphamide pretreatment to decrease leukocyte count prevented the LPS-induced decrease in contractility. In ICAM-1 knock-out mice LPS did not decrease any measure of contractility. LPS did not decrease left ventricular contractility in M+/H- mice but decreased contractility in M+/H+ and M-/H+ mice to the same extent as in C57B6 wild-type mice implicating the importance of cardiac ICAM-1. CONCLUSIONS: We conclude that signaling via cardiac ICAM-1 is necessary to mediate leukocyte-dependent decreases of left ventricular contractility in endotoxemic mice.


Subject(s)
Intercellular Adhesion Molecule-1/physiology , Leukocytes/physiology , Myocardium/immunology , Sepsis/physiopathology , Signal Transduction/physiology , Ventricular Dysfunction, Left/immunology , Animals , Cyclophosphamide/pharmacology , Extracellular Fluid/chemistry , Fibrinogen/analysis , Gene Expression/drug effects , Intercellular Adhesion Molecule-1/genetics , Leukocyte Count , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/chemistry , Myocardium/metabolism , Sepsis/metabolism , Ventricular Dysfunction, Left/metabolism
3.
Am J Physiol Heart Circ Physiol ; 287(3): H1013-22, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15087287

ABSTRACT

ICAM-1 mediates interaction of cardiomyocytes with the extracellular matrix and leukocytes and may play a role in altering contractility. To investigate this possibility, rat ventricular cardiomyocytes were activated using TNF-alpha, IL-1beta, or LPS, washed, cultured with quiescent rat polymorphonuclear leukocytes (PMNs) for 4 h, and electrically stimulated to determine fractional shortening. PMNs cultured with activated cardiomyocytes reduced control fractional shortening of 20.5 +/- 0.7% by -2.8 +/- 0.3% per adherent PMN (P < 0.001). Fixing PMNs with paraformaldehyde or glutaraldehyde did not prevent PMN-mediated decreases in cardiomyocyte fractional shortening. However, PMN adherence and decreased fractional shortening were prevented by anti-ICAM-1 and anti-CD18 antibodies. Reduced fractional shortening was reproduced in the absence of PMNs by ICAM-1 binding using cross-linking antibodies (reduced by 36 +/- 3% from control, P < 0.01). Immunofluorescent staining demonstrated increased cortical cytoskeleton-associated focal adhesion kinase expression after ICAM-1 cross-linking, suggesting involvement of the actin cytoskeleton. Indeed, disruption of F-actin filament assembly using cytochalasin D or latrunculin A did not prevent PMN adherence but prevented decreased fractional shortening. Inhibition of the cytoskeleton-associated Rho-kinase pathway with HA-1077 prevented ICAM-1-mediated decreases in cardiomyocyte contractility, further suggesting a central role of the actin cytoskeleton. Importantly, ICAM-1 cross-linking did not alter the total intracellular Ca2+ transient during cardiomyocyte contraction but greatly increased heterogeneity of intracellular Ca2+ release. Thus we have identified a novel regulatory mechanism of cardiomyocyte contractility involving the actin cytoskeleton as a central regulator of the normally highly coordinated pattern of sarcoplasmic Ca2+ release. Cardiomyocyte ICAM-1 binding, by PMNs or other ligands, induces decreased cardiomyocyte contractility via this pathway.


Subject(s)
Cytoskeleton/physiology , Intercellular Adhesion Molecule-1/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Actins/physiology , Animals , Calcium/metabolism , Cell Adhesion/physiology , Cells, Cultured , Coculture Techniques , Intercellular Adhesion Molecule-1/metabolism , Male , Neutrophils/metabolism , Neutrophils/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
4.
Crit Care ; 7(6): R176-83, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14624693

ABSTRACT

INTRODUCTION: Ischemia/reperfusion occurs in myocardial infarction, cardiac dysfunction during sepsis, cardiac transplantation and coronary artery bypass grafting, and results in injury to the myocardium. Although reperfusion injury is related to the nature and duration of ischemia, it is also a separate entity that may jeopardize viable cells and ultimately may impair cardiac performance once ischemia is resolved and the organ heals. METHOD: The present study was conducted in an ex vivo murine model of myocardial ischemia/reperfusion injury. After 20 min of ischemia, isolated hearts were perfused for up to 2 hours with solution (modified Kreb's) only, solution plus insulin-like growth factor (IGF)-1, or solution plus tumor necrosis factor (TNF)-alpha. Cardiac contractility was monitored continuously during this period of reperfusion. RESULTS: On the basis of histologic evidence, IGF-1 prevented reperfusion injury as compared with TNF-alpha; TNF-alpha increased perivascular interstitial edema and disrupted tissue lattice integrity, whereas IGF-1 maintained myocardial cellular integrity and did not increase edema. Also, there was a significant reduction in detectable creatine phosphokinase in the perfusate from IGF-1 treated hearts. By recording transduced pressures generated during the cardiac cycle, reperfusion with IGF-1 was accompanied by markedly improved cardiac performance as compared with reperfusion with TNF-alpha or modified Kreb's solution only. The histologic and functional improvement generated by IGF-1 was characterized by maintenance of the ratio of mitochondrial to nuclear DNA within heart tissue. CONCLUSION: We conclude that IGF-1 protects ischemic myocardium from further reperfusion injury, and that this may involve mitochondria-dependent mechanisms.


Subject(s)
Insulin-Like Growth Factor I/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Animals , Mice , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...