Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Vasa ; 41(6): 410-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23129036

ABSTRACT

The introduction of drug-eluting stents (DES) to interventional cardiology has been a breakthrough in the treatment of in-stent restenosis. However, the downside of reduced restenosis is a significantly prolonged and practically incalculable time to reendothelialization of thrombogenic stent-surfaces with an increased risk for coronary thrombosis. As the use of DES in non-coronary arteries (e.g. carotid, renal, infrainguinal and even cerebral arteries) is increasing, new vascular beds might be put at risk of ischemia. The practice of stopping antiplatelet drugs in a perioperative setting is highly problematic and contemporary guidelines released by scientific societies from different medical specialties have recently addressed this problem. While many case reports have reported alarming incidents of stent thrombosis, prospective clinical data are scarcely available to guide anticoagulation during the perioperative phase. This review summarizes information on the vascular biology of DES and associated adverse events based on a systematic search of the available literature in public data bases. An emphasis is put on the growing use of DES in non-coronary vessels and the associated danger of putting new vascular beds at risk of thrombotic complications.


Subject(s)
Coronary Artery Disease/therapy , Drug-Eluting Stents , Endovascular Procedures/instrumentation , Percutaneous Coronary Intervention/instrumentation , Peripheral Arterial Disease/therapy , Anticoagulants/administration & dosage , Coronary Restenosis/etiology , Coronary Restenosis/prevention & control , Drug Administration Schedule , Endovascular Procedures/adverse effects , Humans , Percutaneous Coronary Intervention/adverse effects , Platelet Aggregation Inhibitors/administration & dosage , Practice Guidelines as Topic , Prosthesis Design , Risk Assessment , Risk Factors , Thrombosis/etiology , Thrombosis/prevention & control , Time Factors , Treatment Outcome
2.
Perfusion ; 24(1): 19-25, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19567544

ABSTRACT

Pediatric cardiopulmonary bypass is still a challenge because of electrolyte disturbances and inflammation. Many investigations deal with different types of hemofiltration to reduce these potentially harmful side effects. We tested the hypothesis of whether bicarbonate-buffered hemofiltration of the priming solution minimizes electrolyte and acid-base disturbances during the initiation of cardiopulmonary bypass and whether bicarbonate-buffered hemofiltration performed during cardiopulmonary bypass could reduce cytokine levels. Twenty children younger than 2 years of age (mean age 166 +/- 191 days; mean weight 6.42 +/- 3.22 kg) scheduled for pediatric cardiac surgery with cardiopulmonary bypass were enrolled in this prospective clinical study. Cardiopulmonary bypass circuits were primed with a bicarbonate-buffered hemofiltration solution, gelatin and 1 unit of packed red blood cells. The priming was hemofiltered using an ultrahemofilter until approximately 1000 mL of ultrafiltrate was restored with the buffered solution. Further hemofiltration was performed throughout the whole bypass time, especially during rewarming. Blood gas analyses and inflammatory mediators were monitored during the operation. Blood gas analysis results after initiation of cardiopulmonary bypass and throughout the entire study remained within the physiologic ranges. Even potassium decreased from 4.0 +/- 0.3 to 3.4 +/- 0.4 mmol l(-1) after initiation of cardiopulmonary bypass. Plasma levels of tumor necrosis factor alpha decreased significantly (47 +/- 44 vs. 24 +/- 21 pg mL(-1)) whereas complement factor C3a (5.0 +/- 2.9 vs. 16.8 +/- 6.6 ng mL(-1)) and interleukin-6 (7.3 +/- 15.2 vs. 110 +/- 173 pg mL(-1)) increased despite hemofiltration. In conclusion, this study shows that bicarbonate-buffered ultrafiltration is an efficient, simple and safe method for performing hemofiltration, both of the priming solution and during the entire bypass time. The use of a physiological restitution solution prevents electrolyte and acid-base balance disturbances. The elimination of inflammatory mediators seems to be as effective as other ultrafiltration methods.


Subject(s)
Acid-Base Equilibrium/drug effects , Bicarbonates/pharmacology , Cardiopulmonary Bypass/methods , Hemofiltration/methods , Water-Electrolyte Balance/drug effects , Blood Glucose , Cytokines/blood , Hematocrit , Hemoglobins/analysis , Humans , Infant , Lactic Acid/blood , Prospective Studies
3.
Br J Anaesth ; 96(1): 77-87, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16299047

ABSTRACT

BACKGROUND: N-methyl-D-aspartate (NMDA)-receptor activation contributes to postoperative hyperalgesia. Studies in volunteers have shown that intravenous local anaesthetics (LAs) prevent the development of hyperalgesic pain states. One potential explanation for this beneficial effect is the inhibition of NMDA receptor activation. Therefore, we studied the effects of LA on NMDA receptor function. METHODS: The human NR1A/NR2A NMDA receptor was expressed recombinantly in Xenopus laevis oocytes. Peak currents were measured by voltage clamp in Mg- and Ca2+-free, Ba2+-containing Tyrode's solution. Holding potential was -70 mV. Oocytes were stimulated with glutamate/glycine (at EC50) with or without 10 min prior incubation in bupivacaine, levobupivacaine, S-(-)-ropivacaine, or lidocaine (all at 10(-9)-10(-4) M), procaine (10(-4) M), R-(+)-ropivacaine (10(-4) M), QX314 (permanently charged, 5 x 10(-4) M) extracellularly or intracellularly or benzocaine (permanently uncharged, 5 x 10(-3) M). We also determined the effect of the protein kinase C (PKC) inhibitors chelerythrine (5 x 10(-5) M), calphostin C (3 x 10(-6) M) and Ro 31-8220 (10(-7) M), and the effect of PKC activation with phorbolester (10(-6) M). RESULTS: Non-injected oocytes were unresponsive to agonist application, but oocytes expressing NMDA receptors responded with inward currents (1.1+/-0.08 microA). All LA concentration-dependently inhibited agonist responses. The inhibition was reversible and stereoselective. Intracellular QX314 reduced responses to 59% of control, but extracellular QX314 was without effect. Benzocaine reduced responses to 33% of control. PKC inhibitors had no additional inhibitory effect beyond that of bupivacaine. The effect of PKC activation was abolished in the presence of bupivacaine. CONCLUSION: All LA tested inhibited the activation of human NMDA receptors in a concentration dependent fashion. This effect may contribute to reduced hyperalgesia and opiate tolerance observed after systemic administration of LA. The effect is independent of the charge of LA; site of action is intracellular. The mechanism of action may be mediated by inhibition of PKC.


Subject(s)
Anesthetics, Local/pharmacology , Protein Kinase C/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Dose-Response Relationship, Drug , Electrophysiology , Enzyme Inhibitors/pharmacology , Female , In Vitro Techniques , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques/methods , Protein Kinase C/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Xenopus laevis
4.
Br J Anaesth ; 95(5): 603-10, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16155037

ABSTRACT

BACKGROUND: Impedance cardiography (ICG) has been used extensively to estimate stroke volume (SV) and cardiac output (CO) from changes of thoracic electrical bioimpedance (TEB). However, studies comparing ICG with reference methods have questioned the reliability of this approach. Electrical velocimetry (EV) provides a new algorithm to calculate CO from variations in TEB. As the transoesophageal Doppler echocardiographic quantification of CO (TOE-CO) has emerged as a reliable method, the purpose of this study was to determine the limits of agreement between CO estimations using EV (EV-CO) and TOE-CO. METHODS: Standard ECG electrodes were used for non-invasive EV-CO measurements. These were placed on 37 patients scheduled for coronary artery surgery necessitating transoesophageal echocardiography monitoring. Simultaneous EV-CO and TOE-CO measurements were recorded after induction of anaesthesia. EV-CO was calculated using the Bernstein-Osypka equation. TOE-CO was measured across the aortic valve using continuous-wave Doppler echocardiography and a triangular orifice model. RESULTS: A significant high correlation was found between the TOE-CO and the EV-CO measurements (r2=0.86). Data were related linearly. The slope of the line (1.10 (se 0.07)) was not significantly different from unity, and the point at which it intersected the ordinate (-0.46 (0.32) litre min(-1)) was not significantly different from zero. Bland-Altman analysis revealed a bias of 0.18 litre min(-1) with narrow limits of agreement (-0.99 to 1.36 litre min(-1)). CONCLUSIONS: The agreement between EV-CO and TOE-CO is clinically acceptable, and these two techniques can be used interchangeably.


Subject(s)
Cardiac Output , Coronary Artery Bypass , Monitoring, Intraoperative/methods , Stroke Volume , Adult , Aged , Aged, 80 and over , Algorithms , Aortic Valve/diagnostic imaging , Echocardiography, Transesophageal , Female , Hemodynamics , Humans , Male , Middle Aged , Reproducibility of Results , Rheology
5.
Acta Anaesthesiol Scand ; 49(8): 1135-41, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16095455

ABSTRACT

BACKGROUND: Substantial research using echocardiography has established that stroke volume (SV) or cardiac output (CO) can be measured non-invasively at the level of the aortic valve (AV) with high accuracy. Stroke volume is the product of the velocity time integral occurring at the sampling site and the effective systolic AV orifice area (AVOAeff). Nevertheless, a generally accepted method for the determination of AVOAeff is still lacking. METHODS: Aortic valve OAeff was measured in 228 consecutive patients scheduled for coronary artery surgery. Two widely adopted methods were applied to approximate the constantly changing orifice area of the AV: (1) the circular orifice model (AVOA-CM), and (2) the triangular orifice model (AVOA-TM). Aortic valve OA-CM assumes the shape of a circle as an appropriately time averaged geometrical model, and AVOA-TM takes the shape of an equilateral triangle for granted. RESULTS: The AV was easily imaged by echocardiography in both short- and long-axis views in all patients. Relying on AVOA-CM, AVOAeff was 3.49+/-0.77 cm2. AVOA-TM estimates were 2.80+/-0.55 cm2 (mean+/-SD). The results did not agree (bias analysis). CONCLUSIONS: The echocardiographic measurement of SV or CO at the level of the AV has to be reconsidered.


Subject(s)
Aortic Valve/diagnostic imaging , Cardiac Output/physiology , Echocardiography, Doppler/methods , Female , Humans , Male , Middle Aged , Reproducibility of Results , Stroke Volume/physiology , Systole/physiology
6.
Arterioscler Thromb Vasc Biol ; 21(12): 1977-83, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11742873

ABSTRACT

Atherosclerosis was studied in apolipoprotein E (apoE) knockout mice expressing human apolipoprotein A-I (apoA-I) or an apoA-I/apolipoprotein A-II (apoA-II) chimera in which the Arg123-Tyr166 central domain of apoA-I was substituted with the Ser12-Ala75 segment of apoA-II. High density lipoprotein (HDL) cholesterol levels were identical in apoA-I and apoA-I/apoA-II mice, but at 4 months, plaques were 2.7-fold larger in the aortic root of the apoA-I/apoA-II mice (P<0.01). The macrophage-to-smooth muscle cell ratio of lesions was 2.1-fold higher in apo-I/apoA-II mice than in apoA-I mice (P<0.01). This was due to a 2.7-fold higher (P<0.001) in vivo macrophage homing in the aortic root of apoA-I/apoA-II mice. Plasma platelet-activating factor acetyl hydrolase activity was lower (P<0.01) in apoA-I/apoA-II mice, resulting in increased oxidative stress, as evidenced by the higher titer of antibodies against oxidized low density lipoprotein (P<0.01). Increased oxidative stress resulted in increased stimulation of ex vivo macrophage adhesion by apoA-I/apoA-II beta-very low density lipoprotein and decreased inhibition of beta-very low density lipoprotein-induced adhesion by HDL from apoA-I/apoA-II mice. The cellular cholesterol efflux capacity of HDL from apoA-I/apoA-II mice was very similar to that of apoA-I mice. Thus, the Arg123-Tyr166 central domain of apoA-I is critical for reducing oxidative stress, macrophage homing, and early atherosclerosis in apoE knockout mice independent of its role in HDL production and cholesterol efflux.


Subject(s)
Apolipoprotein A-I/genetics , Arteriosclerosis/physiopathology , Cholesterol, HDL/metabolism , Macrophages/metabolism , Animals , Autoantibodies/analysis , Base Sequence , Cell Adhesion , Chimera , Disease Progression , Female , Lipoproteins, HDL/blood , Lipoproteins, LDL/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidative Stress/genetics
7.
Nat Genet ; 28(2): 131-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11381259

ABSTRACT

Hypoxia stimulates angiogenesis through the binding of hypoxia-inducible factors to the hypoxia-response element in the vascular endothelial growth factor (Vegf) promotor. Here, we report that deletion of the hypoxia-response element in the Vegf promotor reduced hypoxic Vegf expression in the spinal cord and caused adult-onset progressive motor neuron degeneration, reminiscent of amyotrophic lateral sclerosis. The neurodegeneration seemed to be due to reduced neural vascular perfusion. In addition, Vegf165 promoted survival of motor neurons during hypoxia through binding to Vegf receptor 2 and neuropilin 1. Acute ischemia is known to cause nonselective neuronal death. Our results indicate that chronic vascular insufficiency and, possibly, insufficient Vegf-dependent neuroprotection lead to the select degeneration of motor neurons.


Subject(s)
Cell Hypoxia/genetics , Endothelial Growth Factors/genetics , Lymphokines/genetics , Motor Neurons/pathology , Nerve Degeneration/genetics , Response Elements/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/physiology , Binding Sites , Electrophysiology , Endothelial Growth Factors/metabolism , Humans , Lymphokines/metabolism , Mice , Mice, Knockout , Motor Neurons/physiology , Muscle Contraction , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1 , Peripheral Nerves/pathology , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Sequence Deletion , Spinal Cord/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Circulation ; 103(20): 2495-500, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11369691

ABSTRACT

BACKGROUND: Atherosclerosis is characterized by an early inflammatory response involving proinflammatory mediators such as platelet-activating factor (PAF)-like phospholipids, which are inactivated by PAF-acetylhydrolase (PAF-AH). The effect of adenovirus-mediated expression of PAF-AH on injury-induced neointima formation and spontaneous atherosclerosis was studied in apolipoprotein E-deficient mice. METHODS AND RESULTS: Intravenous administration of an adenovirus (5 x 10(8) plaque-forming units) directing liver-specific expression of human PAF-AH resulted in a 3.5-fold increase of plasma PAF-AH activity at day 7 (P<0.001); this was associated with a 2.4- and 2.3-fold decrease in malondialdehyde-modified LDL autoantibodies and the lysophosphatidylcholine/phosphatidylcholine ratio, respectively (P<0.001 for both). Non-HDL and HDL cholesterol levels in PAF-AH-treated mice were similar to those of control virus-treated mice. Seven days after virus injection, endothelial denudation of the common left carotid artery was induced with a guidewire. Neointima formation was assessed 18 days later. PAF-AH gene transfer reduced oxidized lipoproteins by 82% (P<0.001), macrophages by 69% (P=0.006), and smooth muscle cells by 84% (P=0.002) in the arterial wall. This resulted in a 77% reduction (P<0.001) of neointimal area. Six weeks after adenovirus-mediated gene transfer, spontaneous atherosclerotic lesions in the aortic root were analyzed. PAF-AH gene transfer reduced atherosclerotic lesions by 42% (P=0.02) in male mice, whereas a nonsignificant 14% reduction was observed in female mice. Basal and PAF-AH activity after gene transfer were higher in male mice than in female mice (P=0.01 and P=0.04, respectively). CONCLUSIONS: Gene transfer of PAF-AH inhibited injury-induced neointima formation and spontaneous atherosclerosis in apolipoprotein E-deficient mice. Our data indicate that PAF-AH, by reducing oxidized lipoprotein accumulation, is a potent protective enzyme against atherosclerosis.


Subject(s)
Adenoviridae/genetics , Apolipoproteins E/deficiency , Arteriosclerosis/prevention & control , Phospholipases A/genetics , Tunica Intima/pathology , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Animals , Apolipoproteins E/genetics , Arteriosclerosis/genetics , Cholesterol, HDL/blood , Cholesterol, VLDL/blood , Female , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oxidative Stress/genetics , Phospholipases A/blood , RNA/genetics , RNA/metabolism , Time Factors , Tunica Intima/metabolism
9.
Curr Opin Anaesthesiol ; 14(1): 17-25, 2001 Feb.
Article in English | MEDLINE | ID: mdl-17016379

ABSTRACT

This review presents a brief overview about the role of regional anaesthesia in patients at risk for myocardial ischemia and/or infarction after cardiac and noncardiac surgical procedures. It includes pathophysiological insights in the problems of plaque rupture and the possible interactions by the use of regional anaesthesia. Special emphasis is put on the subject of thoracic epidural anaesthesia with newer studies showing improvement in relief of angina and improvement of global systolic and diastolic function in patients with coronary artery disease.

10.
FASEB J ; 14(13): 2032-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023987

ABSTRACT

Macrophage infiltration into the subendothelial space at lesion prone sites is the primary event in atherogenesis. Inhibition of macrophage homing might therefore prevent atherosclerosis. Since HDL levels are inversely correlated with cardiovascular risk, their effect on macrophage homing was assessed in apoE-deficient (apoE-/-) mice. Overexpression of human apolipoprotein AI in apoE-/- mice increased HDL levels 3-fold and reduced macrophage accumulation in an established assay of leukocyte homing to aortic root endothelium 3.2-fold (P<0.005). This was due to reduced in vivo betaVLDL oxidation, reduced betaVLDL triggered endothelial cytosolic Ca2+ signaling through PAF-like bioactivity, lower ICAM-1 and VCAM-1 expression, and diminished ex vivo leukocyte adhesion. Adenoviral gene transfer of human PAF-acetylhydrolase (PAF-AH) in apoE-/- mice increased PAF-AH activity 1.5-fold (P<0.001), reduced betaVLDL-induced ex vivo macrophage adhesion 3.5-fold (P<0.01), and reduced in vivo macrophage homing 2.6-fold (P<0.02). These inhibitory effects were observed in the absence of increased HDL cholesterol levels. In conclusion, HDL reduces macrophage homing to endothelium by reducing oxidative stress via its associated PAF-AH activity. This protective mechanism is independent of the function of HDL as cholesterol acceptor. Modulation of lipoprotein oxidation by PAF-AH may prevent leukocyte recruitment to the vessel wall, a key feature in atherogenesis.


Subject(s)
Apolipoproteins E/genetics , Endothelium, Vascular/physiology , Leukocytes/physiology , Lipoproteins, HDL/metabolism , Macrophages, Peritoneal/physiology , Phospholipases A/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Animals , Apolipoprotein A-I/biosynthesis , Arteriosclerosis , Calcium Signaling , Cell Adhesion , Cholesterol/blood , Cytosol/metabolism , Humans , Lipoproteins, VLDL/metabolism , Mice , Mice, Mutant Strains , Models, Biological , Oxidative Stress/physiology , Phospholipases A/genetics
11.
Anesthesiology ; 91(5): 1195-208, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10551568

ABSTRACT

BACKGROUND: Fluorocarbon emulsions have been proposed as temporary artificial oxygen carriers. The aim of the present study is to compare the effectiveness of perflubron emulsion with the effectiveness of autologous blood or colloid infusion for reversal of physiologic transfusion triggers. METHODS: A multinational, multicenter, randomized, controlled, single-blind, parallel group study was performed in 147 orthopedic patients. Patients underwent acute normovolemic hemodilution with colloid to a target hemoglobin of 9 g/dl with an inspiratory oxygen fraction (FIO2) of 0.40. Patients were then randomized into one of four treatment groups after having reached any of the protocol-defined transfusion triggers including tachycardia (heart rate > 125% of posthemodilution rate or > 110 bpm), hypotension (mean arterial pressure < 75% of posthemodilution level or < or = 60 mmHg), elevated cardiac output (> 150% of posthemodilution level) or decreased mixed venous oxygen partial pressure (PVO2; < 38 mmHg). Treatments in the four groups were 450 ml autologous blood harvested during acute normovolemic hemodilution given at FO2 = 0.40; 450 ml colloid at FIO2 = 1.0; 0.9 g/kg perflubron emulsion with colloid (total = 450 ml) at FIO2 = 1.0; and 1.8 g/kg perflubron emulsion with colloid (total = 450 ml) at FIO2 = 1.0. The primary endpoint was duration of transfusion-trigger reversal. A secondary end-point was percentage of transfusion-trigger reversal. RESULTS: Perflubron emulsion was well tolerated with no serious adverse event attributed to drug treatment. Duration of reversal was longest in the 1.8 g/kg perflubron group (median, 80 min; 95% confidence interval, 60-100 min; P = 0.014 vs. autologous blood, P < 0.001 vs. colloid) followed by the 0.9 g/kg perflubron group (median, 59 min; 95% confidence interval, 40-90 min), the autologous blood group (median, 55 min; 95% confidence interval, 30-70 min) and the colloid group (median, 30 min; 95% confidence interval, 27-60 min). Percentage of reversal was also highest in the 1.8 g/kg perflubron group (97%; P < 0.001 vs. autologous blood; P = 0.014 vs. colloid), followed by 0.9 g/kg perflubron (82%), colloid (76%), and autologous blood (60%). CONCLUSIONS: Perflubron emulsion (1.8 g/kg) combined with 100% oxygen ventilation is more effective than autologous blood or colloid infusion in reversing physiologic transfusion triggers.


Subject(s)
Blood Substitutes/therapeutic use , Blood Transfusion, Autologous , Fluorocarbons/therapeutic use , Orthopedic Procedures , Aged , Algorithms , Blood Loss, Surgical , Blood Substitutes/adverse effects , Colloids , Emulsions , Female , Fluorocarbons/adverse effects , Hemodilution , Humans , Hydrocarbons, Brominated , Hypovolemia/prevention & control , Male , Middle Aged , Platelet Count , Single-Blind Method
12.
Blood ; 94(8): 2725-34, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10515876

ABSTRACT

Circulating complexes of leukocytes and activated platelets are markers for atherosclerosis, but their interaction with the arterial endothelial lining has not been studied. Therefore, the effect of activated platelets on rolling and adhesion of labeled human THP-1 monocytoid cells to human umbilical vein endothelial cell (HUVEC) monolayers was studied by epifluorescence microscopy in a parallel plate flow chamber. In the absence of activated platelets, THP-1 rolling on resting HUVEC was negligible at shear rates greater than 300 s(-1). Activation of HUVEC with 100 nmol/L phorbol myristate acetate (PMA) increased THP-1 cell adhesion at shear rates less than 400 s(-1). Therefore, a shear rate of 400 s(-1) was identified as a threshold for THP-1 adhesion. THP-1 rolling on activated HUVEC was reduced by 64% after L-selectin inhibition but was not affected by P-selectin inhibition. The addition of 1 to 50 thrombin receptor-activating peptide (TRAP)-activated platelets per THP-1 cell enhanced interactions between THP-1 cells and HUVEC, resulting in a steep bell-shaped dose-response curve, with a peak of 10 +/- 3 rolling cells/50 seconds at 3 platelets per THP-1 cell (P <.01 v control) with a concomitant 2- to 3-fold increase of firmly adhering cells (P <.01 v control). In reconstituted blood, low numbers of activated platelets had the same effect on THP-1 rolling and adhesion. P-selectin inhibition reduced platelet/THP-1 cell interaction in suspension and deposition of the complexes on the endothelial monolayer. Inhibition of both P- and L-selectin reduced THP-1/HUVEC interactions to 14% (P <.01, n = 4). Sialidase digestion and removal of terminal sialic acid residues from HUVEC or THP-1 cells but not from platelets abolished the platelet mediated augmentation of THP-1 cell adhesion. Thus, THP-1 rolling on HUVEC is shear-dependent and largely mediated by L-selectin. P-selectin expressed on activated platelets increases monocytoid cell adhesion to endothelial cells at shear rates found in coronary arteries through interactions with both endothelial and monocytoid cells and may facilitate macrophage accumulation in the vessel wall.


Subject(s)
Endothelium, Vascular/cytology , Monocytes/cytology , P-Selectin/physiology , Platelet Activation , Stress, Mechanical , Antibodies, Monoclonal/pharmacology , Antigens, CD34/immunology , Cell Adhesion , Coronary Artery Disease/etiology , Dose-Response Relationship, Drug , Fibrinogen , Hemorheology , Humans , Hypercholesterolemia/blood , L-Selectin/physiology , Microscopy, Fluorescence , Microspheres , Neuraminidase/pharmacology , P-Selectin/immunology , Peptide Fragments/pharmacology , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/immunology , Tumor Cells, Cultured , Umbilical Veins
13.
Nat Med ; 5(10): 1135-42, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10502816

ABSTRACT

Cardiac rupture is a fatal complication of acute myocardial infarction lacking treatment. Here, acute myocardial infarction resulted in rupture in wild-type mice and in mice lacking tissue-type plasminogen activator, urokinase receptor, matrix metalloproteinase stromelysin-1 or metalloelastase. Instead, deficiency of urokinase-type plasminogen activator (u-PA-/-) completely protected against rupture, whereas lack of gelatinase-B partially protected against rupture. However, u-PA-/- mice showed impaired scar formation and infarct revascularization, even after treatment with vascular endothelial growth factor, and died of cardiac failure due to depressed contractility, arrhythmias and ischemia. Temporary administration of PA inhibitor-1 or the matrix metalloproteinase-inhibitor TIMP-1 completely protected wild-type mice against rupture but did not abort infarct healing, thus constituting a new approach to prevent cardiac rupture after acute myocardial infarction.


Subject(s)
Cardiac Output, Low/etiology , Heart Rupture/etiology , Metalloendopeptidases/antagonists & inhibitors , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Plasminogen Inactivators/therapeutic use , Protease Inhibitors/therapeutic use , Animals , Arrhythmias, Cardiac , Bone Marrow Transplantation , Cell Movement , Collagenases/metabolism , Gene Transfer Techniques , Leukocytes/cytology , Leukocytes/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9 , Mice , Mice, Mutant Strains , Neovascularization, Physiologic/drug effects , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activators/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
15.
Anesth Analg ; 86(4): 753-8, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9539597

ABSTRACT

UNLABELLED: Acute lung injury is characterized by hypoxemia due to pulmonary ventilation/perfusion-mismatching. I.v. administered prostaglandin E1 (PGE1), a vasodilator with a high pulmonary clearance, has been studied in acute lung injury. Inhalation of the vasodilators nitric oxide and prostacyclin improved oxygenation by selective dilation of the pulmonary vasculature in ventilated lung areas. In the present study, PGE1 inhalation was used for treatment of acute lung injury. Fifteen patients with acute lung injury defined as PaO2/fraction of inspired oxygen (FIO2) <160 mm Hg were treated with PGE1 inhalation in addition to standard intensive care. The drug was continuously delivered via a pneumatic nebulizer. Acute physiology and chronic health evaluation system II and multiple organ failure scores were (mean +/- SEM) 33 +/- 2 and 10 +/- 0.3, respectively. Inhaled PGE1 was administered for 103 +/- 17 h at a dose of 41 +/- 2 microg/h. The PaO2/FIO2 ratio increased from 105 +/- 9 to 160 +/- 17 mm Hg (P < 0.05) and to 189 +/- 25 mm Hg (P < 0.05) after 4 h and 24 h, respectively. PGE1 inhalation decreases in mean pulmonary artery pressure and central venous pressure were not statistically significant. Mean arterial pressure, pulmonary capillary wedge pressure, cardiac output, and heart rate remained unchanged. Intensive care unit mortality was 40%. The present data suggest that inhaled PGE1 is an effective therapeutic option for improving oxygenation in patients with acute lung injury. Whether inhaled PGE1 will increase survival in acute lung injury should be investigated in a controlled prospective trial. IMPLICATIONS: In patients with severe acute lung injury and multiple organ failure, inhaled prostaglandin E1 improved oxygenation and decreased venous admixture without affecting systemic hemodynamic variables. Controlled clinical trials are warranted.


Subject(s)
Alprostadil/therapeutic use , Multiple Organ Failure/complications , Respiratory Distress Syndrome/drug therapy , Vasodilator Agents/therapeutic use , APACHE , Administration, Inhalation , Adult , Aged , Alprostadil/administration & dosage , Blood Pressure/drug effects , Cardiac Output/drug effects , Central Venous Pressure/drug effects , Controlled Clinical Trials as Topic , Critical Care , Epoprostenol/administration & dosage , Epoprostenol/therapeutic use , Female , Heart Rate/drug effects , Humans , Hypoxia/drug therapy , Lung/blood supply , Lung/drug effects , Male , Middle Aged , Nebulizers and Vaporizers , Nitric Oxide/administration & dosage , Nitric Oxide/therapeutic use , Oxygen/administration & dosage , Oxygen/blood , Oxygen Consumption/drug effects , Prospective Studies , Pulmonary Artery/drug effects , Pulmonary Wedge Pressure/drug effects , Respiration, Artificial , Survival Rate , Vasodilator Agents/administration & dosage , Ventilation-Perfusion Ratio/drug effects
17.
Arterioscler Thromb Vasc Biol ; 18(3): 415-22, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9514410

ABSTRACT

The association between accumulation of oxidized low density lipoprotein (LDL) and (1) progression of atherosclerotic plaques and (2) compensatory enlargement was assessed in the coronary arteries of LDL-hypercholesterolemic miniature pigs. In miniature pigs fed a 4% cholesterol diet, LDL cholesterol levels increased from 27+/-3.5 mg/dL (mean+/-SEM, n=36) to 250+/-28 mg/dL (n=10), 260+/-15 mg/dL (n=6), and 260+/-17 mg/dL (n=10) at 6, 14, and 24 weeks, respectively. Mean intimal areas of lesions in the left anterior descending coronary artery of hypercholesterolemic pigs were 0.16+/-0.046 mm2 at 6 weeks (n=10) and increased 5.4-fold (n=6, P<.05) and 10.6-fold (n=10, P<.001) at 14 and 24 weeks, respectively. Plaque growth was associated with an increase in mean internal elastic lamina area, from 1.44+/-0.17 to 4.38+/-0.52 mm2 (P=.007) and in mean luminal area from 1.42+/-0.15 mm2 in control pigs to 4.38+/-0.52 mm2 in pigs fed a cholesterol diet for 24 weeks (P=.007 vs control). Levels of total LDL in the intima, measured immunocytochemically, were 0.031+/-0.0098, 0.11+/-0.057 (P< or =.05), and 0.43+/-0.082 U (P<.001) at 6, 14, and 24 weeks, respectively. Corresponding levels of oxidized LDL were 0.034+/-0.023, 0.11+/-0.050 (P<.05), and 0.44+/-0.065 U (P<.001), respectively, suggesting that virtually all LDL in the intima is oxidized. Levels of oxidized LDL in the lesions were correlated with the intimal areas (r=.85, P<.0001) but were independent of plasma levels of LDL cholesterol and of oxidized LDL. Plaque levels of oxidized LDL were also correlated with internal elastic lamina areas (r=.72, P<.0001) and with luminal areas (r=.50, P=.0098). Plaque growth in the coronary arteries of LDL-hypercholesterolemic miniature pigs is associated with (1) an increase in plaque levels of oxidized LDL at constant plasma levels of LDL cholesterol and of oxidized LDL and (2) compensatory vessel enlargement proportional to plaque levels of oxidized LDL.


Subject(s)
Adaptation, Physiological , Arteriosclerosis/pathology , Cholesterol, LDL/blood , Coronary Vessels/physiopathology , Hypercholesterolemia/pathology , Lipoproteins, LDL/blood , Adaptation, Physiological/physiology , Animals , Arteries/physiopathology , Cholesterol/blood , Hypercholesterolemia/blood , Hypercholesterolemia/physiopathology , Immunohistochemistry , Swine , Swine, Miniature
18.
Biochem Biophys Res Commun ; 244(1): 230-2, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9514911

ABSTRACT

Carbon monoxide is hypothesized to be produced by the enzyme heme oxygenase predominantly in liver and spleen, bound to hemoglobin, and excreted by the lungs. Thus, venous carboxyhemoglobin is expected to be higher or equal to arterial carboxyhemoglobin. Unspecific inflammatory stimuli have been shown to induce heme oxygenase in lung tissue possibly leading to pulmonary carbon monoxide production. Arterial and central venous carboxyhemoglobin levels were measured in critically ill patients on the third day of ICU stay (n = 59) as well as in otherwise healthy humans prior to orthopedic surgery (n = 29). Arterial and central venous carboxyhemoglobin were higher in ICU patients than in healthy humans, respectively. In both groups, arterial carboxyhemoglobin was significantly higher than central venous carboxyhemoglobin. The arteriovenous carboxyhemoglobin differences were similar in both groups. The data suggest (a) increased CO-generation in critical illness and (b) pulmonary CO-production in healthy and critically ill humans.


Subject(s)
Arteries/metabolism , Carbon Monoxide/metabolism , Carboxyhemoglobin/metabolism , Lung/metabolism , Veins/metabolism , Carbon Monoxide/blood , Critical Care , Heart Atria/metabolism , Heme Oxygenase (Decyclizing)/biosynthesis , Humans , Lung/enzymology , Oximetry
19.
J Am Coll Cardiol ; 29(3): 479-85, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9060881

ABSTRACT

OBJECTIVES: The present study was designed to test the hypothesis that long-term dietary supplementation with the nitric oxide precursor L-arginine would enhance vascular or platelet-derived nitric oxide activity, or both, and thereby inhibit platelet reactivity in hypercholesterolemic humans. BACKGROUND: We have shown that reduced vascular activity of nitric oxide in hypercholesterolemic rabbits can be restored by L-arginine supplementation. The improvement in nitric oxide activity is associated with an inhibition of platelet aggregation ex vivo. This effect is most likely due to increased elaboration of endothelium- or platelet-derived nitric oxide, or both, because the inhibition of platelet reactivity was associated with elevation of intraplatelet cyclic guanosine monophosphate and was reversed by the nitric oxide synthase antagonist N-methyl-arginine. METHODS: In a double-blinded, randomized, placebo-controlled trial, hypercholesterolemic patients were assigned to L-arginine hydrochloride, 8.4 g/day orally, or placebo for 2 weeks. Platelet-rich plasma was obtained for aggregometry induced by collagen (1 to 10 micrograms/ml) at four points: baseline, after 2 weeks of treatment, after a 2-week washout and after a long-term washout of 16 weeks on average. Aggregation was quantified by light transmittance and expressed as a percent transmittance observed with platelet-poor plasma. RESULTS: Compared with normocholesterolemic control subjects, platelets from hypercholesterolemic subjects stimulated with 5 micrograms/ml of collagen showed increased aggregability (68.6% in hypercholesterolemic patients vs. 54.5% in normocholesterolemic control subjects, p < or = 0.02). After 2 weeks of treatment with L-arginine (but not placebo), platelet reactivity was modestly reduced; this effect persisted for 2 weeks after discontinuation of arginine (52.6% in arginine-treated patients vs. 65.1% in normocholesterolemic control subjects, p = 0.07). After 18 weeks (i.e., 16 weeks after discontinuing arginine treatment), the platelets of hypercholesterolemic patients once again became hyperaggregable, and the extent of platelet aggregation was significantly increased compared with the 4-week point (73.6% after vs. 52.6% during arginine treatment, p < 0.01). No significant change in platelet reactivity was seen in placebo-treated hypercholesterolemic patients throughout the study. L-Arginine treatment was well tolerated without side effects. CONCLUSIONS: This double-blinded, placebo-controlled study demonstrates that dietary supplementation with L-arginine can modestly attenuate the increased platelet reactivity seen in hypercholesterolemic patients. The data are consistent with our previous studies in hypercholesterolemic animals, demonstrating that L-arginine restores endogenous nitric oxide activity and inhibits platelet aggregation. Enhancement of endogenous nitric oxide activity is a potential novel therapeutic strategy worthy of further study.


Subject(s)
Arginine/pharmacology , Food, Fortified , Hypercholesterolemia/physiopathology , Platelet Aggregation/drug effects , Double-Blind Method , Female , Humans , Male , Nitric Oxide/biosynthesis , Prospective Studies
20.
Arterioscler Thromb Vasc Biol ; 17(12): 3557-64, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9437205

ABSTRACT

Hypercholesterolemia reduces vascular nitric oxide (NO) activity. This dysfunction may promote endothelial monocyte interaction, as NO is a potent inhibitor of cell adhesion. We have previously shown that in hypercholesterolemic (HC) rabbits, chronic oral supplementation of L-arginine (Arg) restores NO activity and inhibits monocyte-endothelial cell interaction, in association with a reduction in atherogenesis. We hypothesized that enhancement of endothelial NO activity in HC humans would reduce monocyte adhesiveness. We used a functional binding assay to assess the adhesiveness of human mononuclear cells (MNCs) ex vivo to determine the effects of hypercholesterolemia and L-arginine administration. MNCs from HC subjects adhered in greater numbers (50% more cells per high-power field; P < .0001) than cells derived from normocholesterolemic (NC) subjects. To determine whether enhancement of endogenous NO activity could inhibit mononuclear cell adhesiveness, in a double-blinded placebo-controlled study, oral arginine HCl (8.4 g/d) was administered to HC subjects. Over a course of 2 weeks, this treatment abolished the increased adhesiveness of HC MNCs (160 +/- 11% versus 104 +/- 5%; before and after 2 weeks of Arg treatment; results expressed as a percentage of the binding values obtained using cells derived from paired NC individuals). By contrast, MNC adhesion remained significantly elevated in placebo-treated HC subjects. To examine whether endothelium-derived NO could act as a paracrine modulator of monocyte behavior, monocytes were exposed to NO donors or cocultered in the presence of endothelial cells exposed to antagonists of NO synthase in the presence or absence of L-arginine. NO donors inhibited monocyte adhesiveness. Furthermore, the adhesiveness of monocytes cocultured with endothelial cells was increased by antagonists of NO synthase; this effect was reversed by L-arginine. This study shows that the adhesiveness of human MNCs is increased by hypercholesterolemia. The increase in adhesiveness was reversed in vivo by administration of the NO precursor L-arginine. NO donors or endothelium-derived NO inhibits the adhesiveness of monocytes in vitro, supporting the hypothesis that the effects of L-arginine are mediated by NO.


Subject(s)
Arginine/metabolism , Hypercholesterolemia/pathology , Leukocytes, Mononuclear/cytology , Adult , Cell Adhesion , Double-Blind Method , Endothelium, Vascular/cytology , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...