Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Artif Organs ; 36(6): 505-11, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22607158

ABSTRACT

The European ST-elevated myocardial infarction (STEMI) guideline suggested the intra-aortic balloon pump (IABP) with a recommendation level I and a level of evidence C as an effective measure in combination with balloon angioplasty in patients with cardiogenic shock (CS), stent implantation, and inotropic and vasopressor support. Similarly, upon mechanical complication due to myocardial infarction (MI), the guideline suggests that in patients with a ventricular septal defect or in most patients with acute mitral regurgitation, preoperative IABP implantation is indicated for circulatory support. The American College of Cardiology/American Heart Association STEMI guideline recommends the use of the IABP with a recommendation level I and a level of evidence B if CS does not respond rapidly to pharmacological treatment. The guideline notes that the IABP is a stabilizing measure for angiography and early revascularization. Even in MI complications, the use of preoperative IABP is recommended before surgery. Within this overview, we summarize the current evidence on IABP use in patients with CS complicated by MI. From our Cochrane data analysis, we conclude that in CS due to acute MI (AMI) treated with adjuvant systemic fibrinolysis, the IABP should be implanted. In patients with CS following AMI, treated with primary percutaneous coronary intervention (PCI), the IABP can be implanted, although data are not distinctive (i.e., indicating positive and negative effects). In the future, randomized controlled trials are needed to determine the use of IABP in CS patients treated with PCI. When patients with CS are transferred to a PCI center with or without thrombolysis, patients should receive mechanical support with an IABP. To treat mechanical MI complications-in particular ventricular septal defect-patients should be treated with an IABP to stabilize their hemodynamic situation prior to cardiac surgery. Similar recommendations are given in the German Austrian guidelines on treatment of infarction-related CS patients (http://www.awmf.org/leitlinien/detail/ll/019-013.html).


Subject(s)
Intra-Aortic Balloon Pumping/methods , Myocardial Infarction/complications , Shock, Cardiogenic/complications , Shock, Cardiogenic/surgery , Europe , Humans , Myocardial Infarction/drug therapy , Practice Guidelines as Topic , Thrombolytic Therapy , United States
2.
Shock ; 37(4): 378-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22266974

ABSTRACT

We conducted the IABP Cardiogenic Shock Trial (ClinicalTrials.gov ID NCT00469248) as a prospective, randomized, monocentric clinical trial to determine the hemodynamic effects of additional intra-aortic balloon pump (IABP) treatment and its effects on severity of disease in patients with acute myocardial infarction complicated by cardiogenic shock (CS). Intra-aortic balloon pump counterpulsation is recommended in patients with CS complicating myocardial infarction. However, there are only limited randomized controlled trial data available supporting the efficacy of IABP following percutaneous coronary intervention (PCI) and its impact on hemodynamic parameters in patients with CS. Percutaneous coronary intervention of infarct-related artery was performed in 40 patients with acute myocardial infarction complicated by CS, within 12 h of onset of hemodynamic instability. Serial hemodynamic parameters were determined over the next 4 days and compared in patients receiving medical treatment alone with those treated with additional intra-aortic balloon counterpulsation. There were no significant differences among severity of disease (i.e., Acute Physiology and Chronic Health Evaluation II score) initially and no differences among both groups for disease improvement. We observed significant temporal improvements of cardiac output (4.8 ± 0.5 to 6.0 ± 0.5 L/min), systemic vascular resistance (926 ± 73 to 769 ± 101 dyn · s(-1) · cm(-5)), and the prognosis-validated cardiac power output (0.78 ± 0.06 to 1.01 ± 0.2 W) within the IABP group. However, there were no significant differences between the IABP group and the medical-alone group. Additional IABP treatment did not result in a significant hemodynamic improvement compared with medical therapy alone in a randomized prospective trial in patients with CS following PCI. Therefore, the use and recommendation for IABP treatment in CS remain unclear.


Subject(s)
Hemodynamics/physiology , Intra-Aortic Balloon Pumping/adverse effects , Myocardial Infarction/complications , Myocardial Infarction/surgery , Shock, Cardiogenic/etiology , Aged , Female , Humans , Male , Middle Aged , Myocardial Infarction/therapy
3.
Clin Res Cardiol ; 101(5): 375-84, 2012 May.
Article in English | MEDLINE | ID: mdl-22212516

ABSTRACT

BACKGROUND: The IABP-SHOCK-trial was a morbidity-based randomized controlled trial in patients with infarction-related cardiogenic shock (CS), which used the change of the quantified degree of multiorgan failure as determined by APACHE II score over a 4-day period as primary outcome measure. The prospective hypothesis was that adding IABP therapy to "standard care" would improve CS-triggered multi organ dysfunction syndrome (MODS). The primary endpoint showed no difference between conventionally managed cardiogenic shock patients and those with IABP support. In an inflammatory marker substudy, we analysed the prognostic value of interleukin (IL)-1ß, -6, -7, -8, and -10 in patients with acute myocardial infarction complicated by cardiogenic shock. DESIGN: Inflammatory marker substudy of the prospective, randomized, controlled, open label IABP-SHOCK-trial (ClinicalTrials.gov ID-NCT00469248). SETTING AND METHODS: A single-center study was performed in a 12-bed Intensive-Care-Unit in an university hospital in which 40 consecutive patients were enrolled with an observational period of 96 h. RESULTS: The pro- and anti-inflammatory markers IL-6, -7, -8 and -10 showed a predictive power for mortality of infarct-related CS patients, while IL-1ß did not discriminate. The maximal values during the observational period, in case of IL-7 the minimal value, showed the best power to predict mortality. Both, ROC and multivariate analyses confirmed these suggestions (area under the curve: IL-8, 0.80 ± 0.08; IL-6, 0.79 ± 0.08; IL-10, 0.76 ± 0.08; IL-7, 0.69 ± 0.08). Inflammatory markers were not affected by the presence of IABP support. CONCLUSION: The inflammatory response in patients with myocardial infarction complicated by cardiogenic shock, as reflected by the inflammatory markers IL-6, IL-7, IL-8 and IL-10, demonstrates a clinically relevant prognostic contribution to clinical outcome.


Subject(s)
Interleukins/blood , Myocardial Infarction/blood , Shock, Cardiogenic/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Coronary Angiography , Female , Humans , Intensive Care Units , Interleukin-10/blood , Interleukin-6/blood , Interleukin-7/blood , Interleukin-8/blood , Male , Middle Aged , Multivariate Analysis , Myocardial Infarction/complications , Myocardial Infarction/mortality , Myocardial Reperfusion/adverse effects , Prognosis , Shock, Cardiogenic/etiology , Shock, Cardiogenic/mortality
4.
Crit Care Med ; 38(1): 152-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19770739

ABSTRACT

OBJECTIVE: Patients undergoing percutaneous coronary intervention (PCI) for acute myocardial infarction with cardiogenic shock (CS) are often treated with intra-aortic balloon pump counterpulsation (IABP), even though the evidence to support this is limited. We determined whether IABP as an addition to PCI-centered therapy ameliorates multiorgan dysfunction syndrome (MODS) in patients with acute myocardial infarction complicated by CS. DESIGN: A prospective, randomized, controlled, open-label clinical trial recruiting patients between March 2003 and June 2004 (ClinicalTrials.gov ID NCT00469248). SETTING: Tertiary care university hospital. PATIENTS AND INTERVENTIONS: Forty-five consecutive patients with AMI and CS undergoing PCI were randomized to treatment with or without IABP. MEASUREMENTS AND MAIN RESULTS: Acute Physiology and Chronic Health Evaluation (APACHE) II scores (primary outcome measure), hemodynamic values, inflammatory markers, and plasma brain natriuretic peptide (BNP) levels (secondary outcomes) were collected over 4 days from randomization. The prospective hypothesis was that adding IABP therapy to "standard care" would improve CS-triggered MODS. The addition of IABP to standard therapy did not result in a significant improvement in MODS (measured by serial APACHE II scoring over 4 days). IABP use had no significant effect on cardiac index or systemic inflammatory activation, although BNP levels were significantly lower in IABP-treated patients. Initial and serial APACHE II scoring correlated with mortality better than cardiac index, systemic inflammatory state, and BNP levels in this group of patients. Nonsurvivors had significantly higher initial APACHE II scores (29.9 +/- 2.88) than survivors (18.1 +/- 1.66, p < .05). Nevertheless, discrepancies among patients within the groups cannot be ruled out and might interfere with our results. CONCLUSIONS: In this randomized trial addressing addition of IABP in CS patients, mechanical support was associated only with modest effects on reduction of APACHE II score as a marker of severity of disease, improvement of cardiac index, reduction of inflammatory state, or reduction of BNP biomarker status compared with medical therapy alone. However, the limitations of our present trial preclude any definitive conclusion, but request for a larger prospective, randomized, multicentered trial with mortality as primary end point.


Subject(s)
Intra-Aortic Balloon Pumping/methods , Multiple Organ Failure/prevention & control , Myocardial Infarction/mortality , Myocardial Infarction/surgery , Shock, Cardiogenic/mortality , Shock, Cardiogenic/surgery , APACHE , Adult , Age Factors , Aged , Aged, 80 and over , Combined Modality Therapy , Coronary Angiography , Counterpulsation/methods , Critical Illness/mortality , Critical Illness/therapy , Electrocardiography , Female , Follow-Up Studies , Hospital Mortality , Humans , Intensive Care Units , Male , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/diagnosis , Probability , Prospective Studies , Risk Assessment , Sex Factors , Shock, Cardiogenic/complications , Shock, Cardiogenic/diagnosis , Survival Analysis , Treatment Outcome
5.
J Cardiovasc Pharmacol ; 52(2): 151-60, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18670364

ABSTRACT

Animal data strongly support a role for inflammation in myocardial ischemia reperfusion injury. Attempts at cardioprotection by immunomodulation (such as with the specific C5 antibody pexelizumab) in humans have been disappointing. We hypothesized that a broader spectrum antiinflammatory agent might yield successful cardioprotection. The serine protease inhibitor nafamostat (FUT-175), which is already in clinical use, is a potent antiinflammatory synthetic serine protease inhibitor with anticomplement activity that we tested in a well-established rabbit model of 1 hour of myocardial ischemia followed by 3 hours of reperfusion. Compared to vehicle-treated animals, nafamostat (1 mg/kg of body weight) administered 5 minutes before reperfusion significantly reduced myocardial injury assessed by plasma creatine kinase activity (38.1 +/- 6.0 versus 57.9 +/- 3.7I U/g protein; P < 0.05) and myocardial necrosis (23.6 +/- 3.1% versus 35.7 +/- 1.0%; P < 0.05) as well as myocardial leukocyte accumulation (P < 0.05). In parallel in vitro studies, Nafamostat was a significantly more potent broad spectrum complement suppressor than C1 inhibitor. Nafamostat appears to have capability as an inhibitor of both complement pathways and as a broad-spectrum antiinflammatory agent by virtue of its serine protease inhibition. Administration of nafamostat before myocardial reperfusion after ischemia produced significant, dose-dependent cardioprotection. Reduced leukocyte accumulation and complement activity seem involved in the mechanism of this cardioprotective effect.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Complement Activation/drug effects , Guanidines/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Serine Proteinase Inhibitors/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzamidines , Complement C1 Inhibitor Protein/pharmacology , Complement C1 Inhibitor Protein/therapeutic use , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/therapeutic use , Complement Pathway, Alternative/drug effects , Complement Pathway, Classical/drug effects , Creatine Kinase/metabolism , Guanidines/administration & dosage , Guanidines/pharmacology , Hemodynamics , Humans , Immunohistochemistry , Male , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Necrosis , Neutrophils/drug effects , Neutrophils/pathology , Rabbits , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/pharmacology
6.
Shock ; 29(4): 497-503, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18598004

ABSTRACT

Bacterial toxins cause cardiac dysfunction and death through an inflammatory process, but the mechanism remains unclear. Simvastatin is recognized as having anti-inflammatory properties beyond its lipid-lowering effects. We examined Staphylococcus aureus alpha-toxin in isolated heart and in vivo models and tested simvastatin's effects in sepsis. Isolated Langendorff-perfused rat hearts were exposed to a recirculating perfusate containing alpha-toxin (0.5 microg mL(-1)). Compared with controls, there was a significant increase in coronary perfusion pressure and fall in myocardial performance. Significant increases in p53 expression and apoptosis (1.3 +/- 0.5 to 7.1 +/- 1.4 terminal deoxynucleaotidyl transferase nick end labeling-positive cells; P < 0.05) compared with controls were observed, but markers of necrosis were similar. In parallel experiments, anaesthetized rats receiving alpha-toxin (40 microg kg(-1), i.v.) had in vivo hemodynamic parameters and serum markers of necrosis monitored for 4 h before the hearts were analyzed for histological change, p53 expression, and apoptosis. Over 4 h, alpha-toxin exposure produced substantial hemodynamic effects. In addition, p53 expression (0.2 +/- 0.2 to 7.1 +/- 0.5 p53-positive myocytes; P < 0.05), TNF-alpha levels, the degree of apoptosis, and markers of necrosis were all significantly increased compared with control animals. Pretreatment with simvastatin protected against alpha-toxin-induced sepsis associated with reduced p53, TNF-alpha, apoptosis, and necrosis. We found significant changes in systemic hemodynamics, coronary perfusion pressure, myocardial function, and increased p53 expression with apoptosis due to bacterial exotoxin. In vivo changes were significantly inhibited by pretreatment with simvastatin. We provide novel evidence for the mechanisms by which septicemia causes myocardial depression and hint at a potential role for simvastatin as an inhibitor of apoptosis in sepsis.


Subject(s)
Apoptosis/drug effects , Cardiomyopathies/drug therapy , Sepsis/drug therapy , Simvastatin/therapeutic use , Animals , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Exotoxins/toxicity , Heart/drug effects , Heart/physiopathology , Hemodynamics/drug effects , In Situ Nick-End Labeling , In Vitro Techniques , Myocardium/metabolism , Myocardium/pathology , Necrosis , Rats , Sepsis/chemically induced , Sepsis/complications , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Proteomics ; 7(24): 4579-88, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18072194

ABSTRACT

Myocardial ischemia (MI) and reperfusion (R) results in activation of the p38 MAP kinase pathway. This pathway phosphorylates transcription factors and cytoplasmic proteins leading to expression of adhesion molecules and cytokines, increased neutrophil activation, and finally, myocardial necrosis and apoptosis. We studied the effects of a p38 MAP kinase inhibitor, PD169316, on cardioprotection, protein expression, and tyrosine phosphorylation, in a rabbit model of 1 h of (MI) and 3 h of (R). PD169316 administered just before (R) significantly reduced myocardial neutrophil accumulation, necrosis area (28.4 +/- 7.9% vs. 56.4 +/- 7.9% necrosis/AAR), and CK release compared to a vehicle treated group (p<0.05). We found several proteins altered in expression following MI + R alone or with p38 inhibition including myofilament proteins, energetics proteins, heat shock proteins, and the mitochondrial porin VDAC-1. p38 MAPK inhibition significantly reduced the phosphorylation of VDAC-1 which is a known mitochondrial regulator of cell survival. Thus, p38 MAP kinase inhibition with PD169316 is cardioprotective, reduces neutrophil activation, and controls protein expression and phosphorylation in MI and reperfusion.


Subject(s)
Cardiotonic Agents/pharmacology , Imidazoles/pharmacology , Myocardial Reperfusion Injury/enzymology , Voltage-Dependent Anion Channels/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antibodies, Phospho-Specific , Electrophoresis, Gel, Two-Dimensional , Hemodynamics/drug effects , Male , Models, Biological , Myocardium/enzymology , Myocardium/pathology , Necrosis , Neutrophils/drug effects , Neutrophils/metabolism , Organ Size/drug effects , Phosphorylation/drug effects , Proteome/metabolism , Proteomics , Rabbits
8.
Circulation ; 116(11 Suppl): I121-6, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17846291

ABSTRACT

BACKGROUND: Reperfusion injury of ischemic myocardium has been attributed to neutrophil infiltration, inflammatory activation and cardiac necrosis/apoptosis. Serine protease inhibition with aprotinin is cardioprotective, but the mechanism is unknown. METHODS AND RESULTS: We studied aprotinin in a rat model of myocardial ischemia for 20 minutes and reperfusion for 20 minutes, 8 hours or 24 hours. Aprotinin (20,000 IU/kg) given 5 minutes before reperfusion significantly reduced leukocyte accumulation (P<0.01), myocardial injury (determined by CK depletion, P<0.01) and myocyte apoptosis (P<0.05) compared with vehicle treated rats. Differential gene expression analysis showed myocardial ischemia plus reperfusion increased expression of proinflammatory genes like P-selectin, E-selectin, intercellular adhesion molecule, tumor necrosis factor-alpha, tumor necrosis factor-alpha receptor, interleukin-6, monocyte chemoattractant protein-1, p53, and Fas (CD59). Aprotinin before reperfusion suppressed expression of these inflammatory genes. Finally, differential protein expression analysis demonstrated increased intercellular adhesion molecule-1, tumor necrosis factor-alpha, and p53 after myocardial ischemia plus reperfusion, and this effect was diminished by aprotinin. CONCLUSIONS: We demonstrated myocardial ischemia plus reperfusion induced leukocyte accumulation, inflammation, gene expression, protein expression and finally tissue injury and showed aprotinin limiting reperfusion injury through each of these stages, even after 24 hours of reperfusion. This effect seems partly attributable to suppression of proinflammatory genes and leukocyte accumulation. This work casts further light on the complex signaling of ischemia and reperfusion.


Subject(s)
Aprotinin/therapeutic use , Gene Expression Regulation/physiology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Protein Biosynthesis/physiology , Animals , Aprotinin/pharmacology , Gene Expression Regulation/drug effects , Myocardial Ischemia/drug therapy , Myocardial Reperfusion/methods , Myocardial Reperfusion Injury/prevention & control , Protein Biosynthesis/drug effects , Rats
9.
Crit Care Med ; 35(12): 2732-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17893627

ABSTRACT

OBJECTIVES: Levosimendan, a novel inodilator, has been shown to improve hemodynamic function in patients with acute exacerbation of congestive heart failure. We wanted to determine the hemodynamic effects of levosimendan following ineffective conventional therapy (with catecholamines) in patients with cardiogenic shock following myocardial infarction. DESIGN: Observational hemodynamic study. SETTING: Tertiary care center university hospital. PATIENTS: Fifty-six patients with cardiogenic shock secondary to myocardial infarction were treated with percutaneous revascularization (intra-aortic balloon pump where appropriate) and commenced on conventional inotropic therapy. INTERVENTIONS: Patients with persisting cardiogenic shock 24 hrs after revascularization were additionally treated with levosimendan (rapid bolus of 12 microg/kg for 10 mins, then 0.05-0.2 mug/kg/min for 24 hrs) (n = 25). MEASUREMENTS AND MAIN RESULTS: With conventional catecholamine therapy (norepinephrine and dobutamine), we observed only marginal improvement in mean arterial pressure or cardiac index. In contrast, the addition of levosimendan produced a significant increase in cardiac index (2.1 +/- 0.56 to 3.0 +/- 1.11 L/min/m2, p < .01) and cardiac power index (0.32 +/- 0.08 to 0.44 +/- 0.18 W, p < .01), whereas systemic vascular resistance decreased significantly (1208 +/- 333 to 858 +/- 299 dyne.sec.cm(-5), p < .01). There was no significant change in blood pressure during levosimendan treatment. Hemodynamic improvement was sustained after levosimendan infusion was stopped. CONCLUSIONS: Levosimendan infusion in cardiogenic shock following acute myocardial infarction improved cardiovascular hemodynamics without leading to hypotension.


Subject(s)
Cardiotonic Agents/pharmacology , Hemodynamics/drug effects , Hydrazones/pharmacology , Pyridazines/pharmacology , Shock, Cardiogenic/drug therapy , Angioplasty, Balloon, Coronary , Cardiotonic Agents/adverse effects , Cardiotonic Agents/therapeutic use , Catecholamines/pharmacology , Catecholamines/therapeutic use , Dobutamine/pharmacology , Dobutamine/therapeutic use , Female , Humans , Hydrazones/adverse effects , Hydrazones/therapeutic use , Infusions, Intravenous , Male , Middle Aged , Pyridazines/adverse effects , Pyridazines/therapeutic use , Shock, Cardiogenic/therapy , Simendan
10.
Biochim Biophys Acta ; 1774(1): 5-15, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16920414

ABSTRACT

OBJECTIVE: Several studies have demonstrated that Sirolimus-eluting stents reduce restenosis in patients with coronary artery disease. Here, we tested whether direct delivery of Sirolimus into the vessel wall during balloon angioplasty can modify vascular remodeling over several weeks. METHODS AND RESULTS: During angioplasty of the rabbit iliac artery we administered an intramural infusion of Sirolimus or its vehicle directly through a balloon catheter into the vessel wall. After 3 weeks neointimal formation was decreased (0.71+/-0.1 vs. 1.4+/-0.12 intima/media ratio), and this process was attributed to the inhibitory properties of Sirolimus on ECM deposition and smooth muscle cell proliferation. Sirolimus also significantly reduced the deposition of elastin, collagen III and fibronectin within the vascular wall. In parallel, proteomic profiles of arterial wall segments were obtained and 485 protein spots were consistently matched between non-dilated and dilated vessels. Differential expression of 12 proteins were observed between the groups and direct sequencing of digested peptides was performed. Local delivery of sirolimus during angioplasty attenuated the expression of structural proteins that included lamin A, vimentin, alpha-1-antitrypsin, and alpha-actin. CONCLUSIONS: Local administration of Sirolimus during angioplasty prevents smooth muscle cell proliferation associated with vascular remodeling as well as the expression of extracellular matrix and structural proteins. Therefore, local injection of Sirolimus during balloon inflation may be an alternative therapeutic approach for preventing restenosis in small stenotic vessels (i.e., <2.5 mm).


Subject(s)
Angioplasty, Balloon , Muscle, Smooth, Vascular/drug effects , Sirolimus/administration & dosage , Actins/biosynthesis , Animals , Cell Proliferation/drug effects , Collagen Type III/biosynthesis , Extracellular Matrix/drug effects , Fibronectins/biosynthesis , Graft Occlusion, Vascular/prevention & control , Iliac Artery/metabolism , Laminin/biosynthesis , Male , Muscle, Smooth, Vascular/cytology , Rabbits , Tunica Intima/pathology , Vimentin/biosynthesis , alpha 1-Antitrypsin/biosynthesis
11.
Biochim Biophys Acta ; 1764(10): 1536-45, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17045855

ABSTRACT

Myocardial ischemia-reperfusion injury can be related to complement activation with generation of chemotactic mediators, release of cytokines, leukocyte accumulation, and subsequent severe tissue injury. In this regard, activation of transcription factors (i.e., NFkappaB) and de novo protein synthesis or inflammatory protein degradation seems to play an important role. In the present study, we analyzed the cardiac protein expression following myocardial ischemia (60 min) and reperfusion (180 min) in a rabbit model utilizing two-dimensional electrophoresis and nanoHPLC/ESI-MS/MS for biochemical protein identification. To achieve cardioprotective effects, we used a novel highly selective small molecule C1s inhibitor administered 5 min prior to reperfusion. The reduction of myocardial injury was observed as diminished plasma creatine kinase activity in C1s-INH-248-treated animals (65.2+/-3 vs. 38.5+/-3 U/g protein after 3 h of reperfusion, P<0.05). With proteome analysis we were able to detect 509+/-21 protein spots on the gels of the 3 groups. A pattern of 480 spots with identical positions was found on every gel of myocardial tissue of sham animals, vehicle and C1s-INH-248-treated animals. We analyzed 11 spots, which were identified by mass spectrometry: Superoxide dismutase, alpha-crystallin-chain-B, mitochondrial stress protein, Mn SOD, ATP synthase A chain heart isoform, creatine kinase, and troponin T. All of these proteins were significantly decreased in the vehicle group when we compared to sham-treated animals. Treatment with C1s-INH-248 preserved levels of these proteins. Thus, blocking the classical complement pathway with a highly specific and potent synthetic inhibitor of the activated C1 complex archives cardio-protection by altering and preserving different anti-inflammatory and cytoprotective cascades.


Subject(s)
Complement System Proteins/physiology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Proteome/analysis , Amino Acid Sequence , Animals , Biomarkers/analysis , Complement C1 Inactivator Proteins/pharmacology , Complement System Proteins/drug effects , Creatine Kinase/analysis , Electrocardiography , Male , Microtubule-Associated Proteins/analysis , Molecular Sequence Data , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Necrosis/pathology , Neutrophils/physiology , Rabbits , Superoxide Dismutase/analysis , Superoxide Dismutase/chemistry , alpha-Crystallin B Chain/analysis
12.
Ann Thorac Surg ; 81(4): 1262-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16564255

ABSTRACT

BACKGROUND: Interaction of circulating leukocytes and vascular endothelium plays an important role in vasoconstriction, endothelial dysfunction, and vascular injury. Dilation procedures of grafts before coronary artery bypass graft surgery might lead to vascular injury and subsequent bypass graft disease. METHODS: We analyzed in vitro the adherence of fluorescence-labeled polymorphonuclear neutrophils (PMNs) to endothelium of human saphenous vein grafts or internal mammary artery grafts after stimulation with thrombin (0.5 to 2 U/mL) or dilating procedures. Furthermore, we investigated endothelial function of prepared grafts. RESULTS: Thrombin stimulation resulted in a dose-dependent increase of PMN adherence to the endothelium of saphenous vein and internal mammary artery, which was attenuated by the selectin-blocking carbohydrate fucoidin or anti-P-selectin monoclonal antibody. Mechanical dilation of saphenous vein or internal mammary artery led to a marked increase in PMN adherence (65 +/- 5 versus 5 +/- 3 PMN/mm2; p < 0.01), which was significantly attenuated by fucoidin or anti-P-selectin monoclonal antibodies. Treatment of internal mammary artery with the vasodilator papaverine led to a marked increase of PMN adherence (59 +/- 8 versus 12 +/- 4 PMN/mm2; p < 0.01) when papaverine was administered directly into the vessel. However, external treatment with papaverine did not affect PMN adhesion. Endothelial dysfunction was observed in dilated venous grafts and in arterial grafts internally treated with papaverine; in contrast, external treatment did not affect endothelial function. CONCLUSIONS: This study showed that mechanical or pharmacologic dilation of venous or arterial coronary grafts, usually performed before anastomosis of aortocoronary bypass grafts, led to increased selectin-mediated PMN adhesion on vascular endothelium and subsequent endothelial dysfunction.


Subject(s)
Endothelium, Vascular , Mammary Arteries , Neutrophils/physiology , Saphenous Vein , Cell Adhesion , Coronary Artery Bypass , Dilatation , Humans , In Vitro Techniques , Mammary Arteries/drug effects , Mammary Arteries/physiology , Mammary Arteries/transplantation , Papaverine/pharmacology , Preoperative Care , Saphenous Vein/drug effects , Saphenous Vein/physiology , Saphenous Vein/transplantation , Thrombin/pharmacology , Vasodilator Agents/pharmacology
13.
Circulation ; 106(16): 2104-10, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12379581

ABSTRACT

BACKGROUND: Simvastatin, a 3-hydroxy-methylglutaryl coenzyme A reductase inhibitor, has been shown to lower serum cholesterol levels in clinical use. Moreover, statins exert beneficial effects in vascular diseases by inhibition of leukocyte rolling, adherence, and transmigration. The aim of this study was to determine if pretreatment with simvastatin attenuates Staphylococcus aureus alpha-toxin-induced increase in leukocyte-endothelial interactions during exotoxemia. METHODS AND RESULTS: The effects of simvastatin on leukocyte-endothelial cell interactions were observed by intravital microscopy in the rat mesenteric microcirculation. Simvastatin (50 or 100 microg/kg) was administered 18 hours before the study. Activation of microcirculation was induced by bolus administration of 40 microg/kg S aureus alpha-toxin. Exotoxemia resulted in a significant and time-dependent increase in leukocyte rolling, adherence, and transmigration of leukocytes as well as P-selectin expression on the intestinal vascular endothelium. Pretreatment with simvastatin significantly inhibited exotoxin-induced leukocyte rolling from 71+/-10 to 14+/-4.7 cells/min (P<0.01) and adherence from 14+/-3.5 to 0.4+/-0.2 cells (P<0.01). In addition, simvastatin pretreatment significantly inhibited transmigration of leukocytes from 10.5+/-1.2 to 4.2+/-0.9 (P<0.05) cells. Immunohistochemical detection of endothelial cell adhesion molecule P-selectin showed a 50% decrease in endothelial cell surface expression after simvastatin treatment. Furthermore, simvastatin treatment resulted in enhanced expression of endothelial cell NO synthase III in the intestinal microcirculation. CONCLUSIONS: These results demonstrate that simvastatin interferes with exotoxin-induced leukocyte-endothelial cell interactions, which may be relevant in various infectious diseases. Statin treatment may offer a new therapeutic strategy for these clinical conditions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacterial Toxins/antagonists & inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Toxemia/prevention & control , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Culture Techniques , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Hemodynamics , Hemolysin Proteins , Immunohistochemistry , Leukocytes/drug effects , Leukocytes/immunology , Male , Mesenteric Veins/anatomy & histology , Mesenteric Veins/drug effects , Mesenteric Veins/physiopathology , Microscopy, Video , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase Type III , P-Selectin/analysis , P-Selectin/immunology , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Staphylococcal Infections/prevention & control , Toxemia/immunology , Toxemia/pathology , Venules/drug effects , Venules/physiopathology
14.
J Thorac Cardiovasc Surg ; 124(5): 942-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12407377

ABSTRACT

OBJECTIVE: Early coronary reperfusion of the ischemic myocardium is a desired therapeutic goal to preserve myocardium. However, reperfusion itself contributes to an additional myocardial injury (ie, reperfusion injury), which has been attributed to neutrophil infiltration with subsequent release of proteases and oxygen-derived radicals. We studied the effects of the serine protease inhibitor aprotinin (Trasylol) on myocardial ischemia and reperfusion in a rat model. METHODS: The effects of aprotinin (5000 and 20,000 U/kg) were examined in vivo in a rat model of regional myocardial ischemia (20 minutes) and long-term reperfusion (24 hours). Cardioprotecive effects were determined by means of measurement of creatine kinase and myeloperoxidase activity within the myocardium, as well as histochemical analysis. RESULTS: Aprotinin (20,000 U/kg) administrated 2 minutes before reperfusion significantly attenuated myocardial injury expressed as creatine kinase washout compared with that seen in vehicle-treated rats (65 +/- 25 vs 585 +/- 98 creatine kinase difference in units per 100 mg, P <.01). Administration of 5000 U/kg of the protease inhibitor resulted in partial inhibition of myocardial reperfusion injury. Moreover, cardiac myeloperoxidase activity in the ischemic myocardium, a marker of neutrophil accumulation, was significantly reduced after aprotinin treatment. Histologic analysis of the reperfused myocardium demonstrated reduced polymorphonuclear leukocyte infiltration and reduced tissue injury. Furthermore, aprotinin treatment resulted in decreased induction of cardiac myocyte apoptosis compared with that seen in vehicle-treated rats. CONCLUSIONS: Inhibition of serine proteases with aprotinin appears to be an effective means of preserving ischemic myocardium from reperfusion injury, even after 24 hours of reperfusion. Aprotinin might exert cardioprotection through inhibition of polymorphonuclear leukocyte-induced myocardial injury and inhibition of reperfusion-induced apoptosis of cardiac myocytes.


Subject(s)
Aprotinin/administration & dosage , Cardiotonic Agents/administration & dosage , Myocardial Ischemia/drug therapy , Myocardial Ischemia/surgery , Myocardial Reperfusion , Myocardium/metabolism , Myocardium/pathology , Serine Proteinase Inhibitors/administration & dosage , Animals , Apoptosis/drug effects , Biomarkers/blood , Combined Modality Therapy , Creatine Kinase/drug effects , Creatine Kinase/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Heart Ventricles/metabolism , Leukocyte Count , Male , Models, Cardiovascular , Myocardial Ischemia/blood , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Neutrophils/drug effects , Neutrophils/metabolism , Peroxidase/drug effects , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Treatment Outcome
15.
Shock ; 17(1): 30-5, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11795666

ABSTRACT

The effect of Staphylococcus aureus alpha toxin (alpha-toxin) on selectin-mediated neutrophil adhesion was investigated in polymorphonuclear leukocyte- (PMN) induced vasocontraction and endothelial dysfunction. Adherence of human PMNs to rat aortic endothelium increased significantly following stimulation of the endothelium with alpha-toxin (0.1, 0.5, and 1 microg/mL). This effect could be significantly attenuated by monoclonal antibodies directed against P-selectin or fucoidin, a carbohydrate known to block selectins. Unstimulated human PMNs (10(6)cells/mL) were added to organ chambers containing rat aortic rings stimulated with alpha-toxin (0.5 microg/mL). PMNs elicited a significant vasocontraction in alpha-toxin-stimulated, but not in control aortic, rings (142+/-12 mg versus 12+/-4 mg, P < 0.05). This PMN-induced vasocontraction was virtually blunted by pretreatment with MAb directed against P-selectin or fucoidin (P < 0.05). Endothelial function as assessed by endothelium-dependent vasorelaxation to acetylcholine was substantially inhibited after induction of PMN-induced vasocontraction in alpha-toxin-stimulated aortic rings. This endothelial dysfunction was reduced by P-selectin MAb or fucoidin. In contrast, endothelium-independent relaxation to sodium nitrite was not altered by PMN incubation, indicating that vascular smooth muscle function was unaffected. Thus, PMN-endothelial interaction following S. aureus a-toxin activation of the vascular endothelium is at least, in part, mediated by selectins. As a consequence, PMN-induced vasocontraction and endothelial dysfunction occur. Such mechanisms may be involved in microcirculation abnormalities encountered in sepsis or septic shock due to S. aureus infection.


Subject(s)
Bacterial Toxins/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Hemolysin Proteins/pharmacology , Neutrophils/metabolism , Vasoconstriction/drug effects , Animals , Aorta/drug effects , Aorta/physiopathology , Azepines/pharmacology , Cell Adhesion/drug effects , Fibrinolytic Agents/pharmacology , Humans , In Vitro Techniques , Neutrophils/cytology , Rats , Thrombin/pharmacology , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...