Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Hamostaseologie ; 23(4): 186-98, 2003 Nov.
Article in German | MEDLINE | ID: mdl-14603383

ABSTRACT

Cardiovascular disease, to which venous thrombosis and pulmonary embolism contribute a major part, are the leading cause of death in industrialized countries. Thrombosis is caused by temporary risk factors or by thrombophilia. Increasing knowledge concerning the pathophysiology of thrombosis and new diagnostic tools allow a thrombotic risk assessment. In this paper thrombotic risk factors will be discussed with respect to their significance and diagnosis as well as methods of prophylaxis and treatment for patients at risk for or with diagnosed thrombosis. Furthermore, currently available as well as new anticoagulants (pentasaccharide, melagatran) will be discussed and their potential for patient-oriented and cost-efficient prophylaxis and treatment of thromboembolic complications outlined.


Subject(s)
Thrombophilia/therapy , Venous Thrombosis/therapy , Enoxaparin/therapeutic use , Fibrinolytic Agents/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Risk Factors , Thrombophilia/prevention & control , Tinzaparin , Venous Thrombosis/congenital , Venous Thrombosis/etiology , Venous Thrombosis/prevention & control
2.
J Thromb Thrombolysis ; 12(2): 165-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11729368

ABSTRACT

BACKGROUND: Novel antithrombotic agents such as hirudin have shown promise in the therapy of acute coronary syndromes. PEG-hirudin (polyethyleneglycol conjugated hirudin) has been developed to provide a longer plasma half-life and more stable antithrombotic plasma levels. Privious trials indicated a narrow therapeutic window for hirudin and a number of aPTT (activated partial thromboplastin time)-monitored trials investigating hirudin in acute coronary syndromes had to be stopped because of intracranial bleeding complications. OBJECTIVES: The present study evaluates the ecarin clotting time (ECT), a parameter based on the conversion of prothrombin by the snake venom enzyme ecarin, for the monitoring of PEG-hirudin therapy. METHODS: Plasma from either healthy volunteers (n=20) or from patients (n=10) suffering from unstable angina pectoris (UAP) was spiked with increasing PEG-hirudin concentrations. In a prospective randomized clinical trial patients with UAP were treated with intravenous PEG-hirudin or heparin over 72 hours. Patients were randomized to the following treatment groups: (1) heparin control group, n=15; (2) PEG-hirudin low dose (0.1 mg/kg bolus, 0.01 mg/kg/h infusion), n=19; (3) intermediate dose (0.15 mg/kg and 0.015 mg/kg/h), n=17; 4) high-dose (0.2 mg/kg and 0.02 mg/kg/h), n=16. Spiked plasma samples and plasma from UAP patients treated with i.v. PEG-hirudin were analyzed for aPTT, ECT, and PEG-hirudin levels. RESULTS: A linear correlation up to the highest therapeutic concentrations could be observed between PEG-hirudin plasma concentrations and the ECT. This was true for both plasma samples spiked with PEG-hirudin in vitro as well as for samples taken from patients treated with i.v. PEG-hirudin (correlation coefficient 0.9, respect.) In contrast the aPTT did not show a reliable linear correlation to PEG-hirudin concentrations. CONCLUSION: Monitoring of PEG-hirudin therapy by ECT may help to avoid inadequate anticoagulation or overdosing. Thus, the safety and efficacy profile of PEG-hirudin therapy is likely to be enhanced by ECT monitoring.


Subject(s)
Antithrombins/pharmacokinetics , Drug Monitoring/methods , Endopeptidases , Hirudins/pharmacokinetics , Angina, Unstable/blood , Angina, Unstable/drug therapy , Antithrombins/analysis , Antithrombins/therapeutic use , Blood Coagulation Tests/methods , Blood Coagulation Tests/standards , Drug Monitoring/standards , Fibrinolytic Agents , Hirudin Therapy , Hirudins/analogs & derivatives , Hirudins/blood , Humans , Partial Thromboplastin Time , Regression Analysis
3.
Basic Res Cardiol ; 96(2): 143-50, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11327332

ABSTRACT

Oxidation of lipids is considered a key feature of atherogenesis. Lipid peroxidation products such as oxidized LDL or the bioactive aldehyde 4-hydroxynonenal (HNE) exert mitogenic effects on vascular smooth muscle cells (VSMC). These effects appear to be concentration-dependent since in addition to our previous reports on growth promotion at lower concentrations we here indicate induction of apoptosis in VSMC by 4-hydroxynonenal (HNE) at higher concentrations (100 micromol/L). In a line with HNE's previously documented effects on key mitogenic signaling elements, we also report on activation by this aldehyde of the redox-sensitive transcription factor NF-kappaB, a key regulator of apoptosis: HNE (1.0 micromol/L) induced DNA-binding of NF-kappaB in VSMC. The effect was inhibited by antioxidants, N-acetylcysteine and pyrrolidine dithio-carbamate. HNE caused phosphorylation but not degradation of the inhibitory subunit IkappaB-alpha. HNE itself acts as an oxidant as was investigated with measurements of 8-isoprostane which ranks among the most valuable available biomarkers of lipid peroxidation: HNE (1.0 micromol/L) increased 8-isoprostane levels in VSMC by 4.5-fold (p < 0.05). Compared to the controls, plasma samples from apoEnull mice exhibited elevated levels of 8-isoprostane (40 pg/mL, 3.2-fold increase) and the combined aldehydes HNE and malonaldehyde (1.5 micromol/L, 2.5-fold increase), (p < 0.05, resp). In addition, immunohistochemistry indicated the presence of HNE-protein adducts in atheroscerlotic lesions of apoEnull mice. Thus HNE is present in atherosclerotic tissue at concentrations that are bioactive in vitro. The data further indicate the involvement of the lipid peroxidation product HNE in atherogenesis.


Subject(s)
Aldehydes/pharmacology , Apoptosis/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Dinoprost/biosynthesis , I-kappa B Proteins , Muscle, Smooth, Vascular/cytology , NF-kappa B/metabolism , Animals , Aorta/cytology , Apolipoproteins E/genetics , Apoptosis/physiology , Cells, Cultured , Coronary Artery Disease/metabolism , Culture Media, Conditioned/pharmacology , DNA/metabolism , DNA-Binding Proteins/metabolism , Dinoprost/analogs & derivatives , Dinoprost/blood , Dose-Response Relationship, Drug , F2-Isoprostanes , Humans , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Lipid Peroxides/blood , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , NF-KappaB Inhibitor alpha , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism
4.
Chem Biol Interact ; 130-132(1-3): 627-36, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11306081

ABSTRACT

Aldose reductase (AR) is a broad-specificity aldo-keto reductase with wide species and tissue distribution. The enzyme has been implicated in the development of pleiotropic complications of long-term diabetes. However, the euglycemic function of the enzyme remains unclear. To examine its potential role in cell growth, changes in AR mRNA and protein were measured in human aortic smooth muscle cells exposed in culture to serum or thrombin. Stimulation by these mitogens led to an increase in the abundance of AR mRNA and protein. Furthermore, inhibition of the AR by tolrestat and sorbinil diminished DNA synthesis and cell proliferation in response to serum. Immunohistochemical staining with anti-AR antibodies revealed no significant expression of AR in the smooth muscle cells of rat carotid arteries. However, 10 and 21 days after balloon injury, intense staining was associated with the proliferating cells of the neointima. Treatment of these animals with 40 mg/kg/day sorbinil diminished the ratio of neointima to the media. Together, these observations suggest that, in vascular smooth muscle cells (VSMC), AR is a growth-responsive gene product and that inhibition of AR prevents VSMC growth and decreases intimal hyperplasia and restenosis.


Subject(s)
Aldehyde Reductase/metabolism , Imidazolidines , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/genetics , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Cell Division , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Mitogens/pharmacology , Naphthalenes/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Rats , Up-Regulation
5.
Thromb Haemost ; 86(5): 1305-13, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11816722

ABSTRACT

Human atherosclerotic lesions exhibit increased expression of plasminogen activator inhibitor type-1 (PAI-1) that has been implicated in atherogenesis. Although vascular smooth muscle cells are a predominant source of PAI-1 expression potentially favorable modulation of PAI-1 expression by fibrates has not yet been characterized in these cells. Human aortic smooth muscle cells were exposed to selected growth factors. PAI-1 expression was stimulated most powerfully by TGF-beta (EC50 = 0.2 ng/ml, up to 12-fold increase). Gemfibrozil inhibited basal PAI-1 expression by 23% (p = ns) and TGF-beta-induced PAI-1 expression by 52% (p = 0.017) whereas t-PA and total protein synthesis was not affected. Changes in PAI-1 protein accumulation reflected PAI-1 gene expression attributable to modulation of half-life of PAI-1 mRNA by gemfibrozil. Inhibition by other fibrates was less. Gemfibrozil specifically attenuates TGF-beta-induced PAI-1 expression in human arterial smooth muscle cells. Thus, fibrates are promising agents for normalizing increased PAI-1 expression in arterial walls in patients in whom PAI-1 expression is increased.


Subject(s)
Gemfibrozil/pharmacology , Muscle, Smooth, Vascular/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Aorta/cytology , Cells, Cultured , Drug Interactions , Gene Expression Regulation/drug effects , Humans , Muscle, Smooth, Vascular/cytology , Plasminogen Activator Inhibitor 1/genetics , RNA Stability/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Transforming Growth Factor beta/pharmacology
6.
Cardiovasc Pathol ; 10(6): 311-5, 2001.
Article in English | MEDLINE | ID: mdl-11755377

ABSTRACT

Atherosclerosis is based on endothelial dysfunction leading to impaired vasomotor function. This is partially due to nitric oxide (NO) depletion caused by oxidative stress. Since the vasoconstrictor endothelin-1 (ET-1) might also be involved in endothelial dysfunction, we investigated whether oxidative stress regulates ET-1 expression in vascular smooth muscle cells (VSMC). Human aortic VSMC were treated with H(2)O(2) (200 microM) for up to 8 h. mRNA expression of preproendothelin (prepro-ET) was analyzed by RT-PCR. ET-1 protein and the marker for oxidative stress, 8-isoprostane, were determined by ELISA. Activity of cytosolic phospholipase A2 (cPLA(2)) as an indicator of ET-1 autocrine activity was measured photometrically. Stimulation of VSMC with H(2)O(2) resulted in increased expression of prepro-ET mRNA after 1 h with a maximum after 6 h (fourfold), similar to treatment with angiotensin II. ET-1 protein was significantly increased by H(2)O(2) treatment with a maximum after 8 h (P<.05). This effect was inhibited by the antioxidants resveratrol (100 microM) and quercetin (50 microM). In quiesced VSMC, incubation with H(2)O(2)-conditioned medium resulted in increased cPLA(2) activity compared to the controls (P<.05). This activity was partially inhibited by the ET(A)-receptor antagonist, PD 142893 (10 microM), indicating functional ET-1 in the conditioned medium. The presence of oxidative stress in H(2)O(2)-treated VSMC was associated by significantly increased formation of 8-isoprostane (P<.05). The data indicate for the first time that oxidative stress increases ET-1 generation and autocrine ET-1 activity in VSMC, a mechanism that might contribute to endothelial dysfunction in atherosclerosis.


Subject(s)
Dinoprost/analogs & derivatives , Endothelin-1/biosynthesis , Muscle, Smooth, Vascular/metabolism , Oxidative Stress , Antioxidants/pharmacology , Aorta , Cells, Cultured , Cytosol/enzymology , Dose-Response Relationship, Drug , Endothelin-1/genetics , Endothelin-1/immunology , Enzyme-Linked Immunosorbent Assay , F2-Isoprostanes/analysis , F2-Isoprostanes/metabolism , Humans , Hydrogen Peroxide/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oligopeptides/pharmacology , Phospholipases A/analysis , Phospholipases A/biosynthesis , Phospholipases A2 , Quercetin/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/pharmacology
7.
Ann Hematol ; 79(11): 604-11, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11131919

ABSTRACT

Activation of the protease-activated receptor (PAR)-1, one of four known PARs (PAR-1 to PAR-4), can be mimicked by thrombin receptor activating peptides (TRAPs) based on the PAR-1 tethered ligand. Interestingly, despite being activatable by thrombin, rodent platelets do not express PAR-1 and thus do not respond to PAR-1-derived TRAPs, indicating different activation mechanisms between human and rodent platelets. Using a rat platelet aggregation model, we determined that TRAPs based on the tethered ligand of PAR-1 fail to activate rat platelet aggregation at concentrations up to 1 mmol/l. In addition, TRAPs inhibit thrombin-mediated rat platelet aggregation, indicating the presence of a modified PAR-1 in this species. In order to determine characteristics of this putative receptor, we tested a panel of synthesized TRAPs based on the rat sequence (R) and human sequence (H) of the PAR-1 tethered ligand for their ability to inhibit thrombin-induced rat platelet aggregation. Peptides R1-9, R4-9, R4-10, and H4-10 inhibited rat platelet aggregation in response to alpha-thrombin [inhibitory concentration (IC) 50% 0.25-1.5 mmol/l]. None of these peptides blocked epinephrine-, collagen-, or arachidonic acid-induced platelet aggregation. Alanine substitution mapping of H4-10 indicated that both Leu4 and Arg5 are essential for inhibition. Inhibition of thrombin's catalytic activity required peptide concentrations tenfold higher than inhibition of platelet aggregation (IC50% 3-5 mmol/l). No prolongation of thrombin clotting time in response to TRAPs was detected at peptide concentrations up to 5 mmol/l. Our data suggest that (1) rat platelets express a PAR-1 subtype, (2) residues Leu4 and Arg5 of the tethered ligand peptide are required for binding to this new receptor, and (3) further analysis of peptide sequences might reveal a novel PAR-1 subtype.


Subject(s)
Blood Platelets/chemistry , Receptors, Thrombin/blood , Animals , Humans , Peptide Fragments/pharmacology , Peptides/antagonists & inhibitors , Platelet Aggregation/drug effects , Rats , Receptor, PAR-1 , Receptors, Thrombin/physiology
8.
Circulation ; 102(13): 1490-6, 2000 Sep 26.
Article in English | MEDLINE | ID: mdl-11004138

ABSTRACT

BACKGROUND: Improvement of thrombolysis may be achieved by concomitant strong platelet inhibition. To monitor platelet function in patients with myocardial infarction (n=46) who were treated with the fibrinolytic agent reteplase, the glycoprotein (GP) IIb/IIIa blocker abciximab, and the ADP receptor antagonist ticlopidine, we developed a flow cytometric assay. METHODS AND RESULTS: Binding of abciximab to platelets was directly monitored as the percentage of platelets stained by a goat anti-mouse antibody. Blood drawn 10 minutes and 2 hours after the start of therapy with reteplase and abciximab and during the 12-hour infusion of abciximab demonstrated a maximal blockade of GP IIb/IIIa (10 minutes, 86.2+/-10.3%; 12 hours, 85.8+/-7.1%). Starting at 24 hours, abciximab binding gradually decreased (24 hours, 74.6+/-16.2%; 48 hours, 66.8+/-14.9%; 72 hours, 60.5+/-16.7%; 96 hours, 49.4+/-17.8%; 120 hours, 35.8+/-16. 4%; and 144 hours, 29.9+/-15.3%). Binding of a chicken anti-fibrinogen antibody to platelets, indicating the level of functional blockade of GP IIb/IIIa, was inversely correlated with the binding of abciximab (r=-0.72, P:<0.0001). In blood drawn at 10 minutes, platelet aggregation was maximally inhibited but recovered within 48 hours even if the majority of GP IIb/IIIa receptors were still blocked by abciximab. Reteplase did not influence abciximab binding and did not activate platelets, as measured by P-selectin expression, fibrinogen binding, and platelet aggregation. Platelet inhibition that was achieved during the first 24 hours by abciximab was directly maintained by additional treatment with ticlopidine. CONCLUSIONS: Flow cytometric monitoring of platelet function allows differentiation of the effects of reteplase, abciximab, and ticlopidine. The combination of abciximab and ticlopidine is an attractive therapeutic strategy that provides a fast and continuous platelet inhibition.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood Platelets/drug effects , Fibrinolytic Agents/therapeutic use , Immunoglobulin Fab Fragments/therapeutic use , Myocardial Infarction/blood , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Recombinant Proteins/therapeutic use , Ticlopidine/therapeutic use , Tissue Plasminogen Activator/therapeutic use , Abciximab , Aged , Antibodies, Monoclonal/blood , Blood Platelets/physiology , Female , Fibrinolytic Agents/blood , Flow Cytometry , Humans , Immunoglobulin Fab Fragments/blood , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Recombinant Proteins/blood , Ticlopidine/blood , Time Factors , Tissue Plasminogen Activator/blood
9.
Arterioscler Thromb Vasc Biol ; 20(7): 1745-52, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10894812

ABSTRACT

Abnormal proliferation of vascular smooth muscle cells (VSMCs) is an important feature of atherosclerosis, restenosis, and hypertension. Although multiple mediators of VSMC growth have been identified, few effective pharmacological tools have been developed to limit such growth. Recent evidence indicating an important role for oxidative stress in cell growth led us to investigate the potential role of aldose reductase (AR) in the proliferation of VSMCs. Because AR catalyzes the reduction of mitogenic aldehydes derived from lipid peroxidation, we hypothesized that it might be a potential regulator of redox changes that accompany VSMC growth. Herein we report several lines of evidence suggesting that AR facilitates/mediates VSMC growth. Stimulation of human aortic SMCs in culture with mitogenic concentrations of serum, thrombin, basic fibroblast growth factor, and the lipid peroxidation product 4-hydroxy-trans-2-nonenal (HNE) led to a 2- to 4-fold increase in the steady-state levels of AR mRNA, a 4- to 7-fold increase in AR protein, and a 2- to 3-fold increase in its catalytic activity. Inhibition of the enzyme by sorbinil or tolrestat diminished mitogen-induced DNA synthesis and cell proliferation. In parallel experiments, the extent of reduction of the glutathione conjugate of HNE to glutathionyl-1,4-dihydroxynonene in HNE-exposed VSMCs was decreased by serum starvation or sorbinil. Immunohistochemical staining of cross sections from balloon-injured rat carotid arteries showed increased expression of AR protein associated with the neointima. The media of injured or uninjured arteries demonstrated no significant staining. Compared with untreated animals, rats fed sorbinil (40 mg. kg(-1). d(-1)) displayed a 51% and a 58% reduction in the ratio of neointima to the media at 10 and 21 days, respectively, after balloon injury. Taken together, these findings suggest that AR is upregulated during growth and that this upregulation facilitates growth by enhancing the metabolism of secondary products of reactive oxygen species.


Subject(s)
Aldehyde Reductase/metabolism , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Imidazolidines , Muscle, Smooth, Vascular/enzymology , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/genetics , Aldehydes/pharmacology , Angioplasty, Balloon/adverse effects , Animals , Aorta/cytology , Carotid Stenosis/enzymology , Carotid Stenosis/pathology , Cell Division/physiology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glutathione/metabolism , Hemostatics/pharmacology , Humans , Imidazoles/pharmacology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Muscle, Smooth, Vascular/cytology , Naphthalenes/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Recurrence , Thrombin/pharmacology , Tritium
10.
Diabetes Res Clin Pract ; 45(2-3): 89-99, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10588360

ABSTRACT

Diabetes mellitus is associated with increased frequency, severity and more rapid progression of cardiovascular diseases. Metabolic perturbations from hyperglycemia result in disturbed endothelium-dependent relaxation, activation of coagulation pathways, depressed fibrinolysis, and other abnormalities in vascular homeostasis. Atherosclerosis is localized mainly at areas of geometric irregularity at which blood vessels branch, curve and change diameter, and where blood is subjected to sudden changes in velocity and/or direction of flow. Shear stress resulting from blood flow is a well known modulator of vascular cell function. This paper presents what is currently known regarding the molecular mechanisms responsible for signal transduction and gene regulation in vascular cells exposed to shear stress. Considering the importance of the hemodynamic environment of vascular cells might be vital to increasing our understanding of diabetes.


Subject(s)
Blood Vessels/physiopathology , Diabetes Complications , Diabetes Mellitus/physiopathology , Diabetic Angiopathies/physiopathology , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Animals , Blood Vessels/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Diabetes Mellitus/genetics , Diabetic Angiopathies/genetics , Endothelium, Vascular/metabolism , Humans , Models, Cardiovascular , Stress, Mechanical
11.
Circulation ; 100(18): 1858-64, 1999 Nov 02.
Article in English | MEDLINE | ID: mdl-10545429

ABSTRACT

BACKGROUND: Changes in platelet aggregation (PA) and platelet surface receptor expression induced by thrombolytic therapy for acute myocardial infarction may influence the rate of initial reperfusion and early reocclusion. METHODS AND RESULTS: In the RAPID-1 (Reteplase Angiographic Phase II International Dose-finding study), RAPID-2 (Reteplase vs Alteplase Patency Investigation During myocardial infarction), INJECT (INternational Joint Efficacy Comparison of Thrombolytics), and GUSTO-3 (Global Use of Strategies To Open occluded coronary arteries) trials, 126 patients were enrolled in a single center. Patients were treated with either conventional alteplase (100 mg/180 min; n=15), accelerated alteplase (100 mg/90 min; n=21), reteplase 10+10-U double bolus (n=50), reteplase 10+5-U double bolus (n=15), reteplase 15-U single bolus (n=15), or streptokinase (1.5 MU/60 min; n=10). PA (after stimulation with ADP), P-selectin expression and fibrinogen binding to glycoprotein (GP) IIb/IIIa (determined by flow cytometry with and without stimulation with ADP), and levels of soluble P-selectin, prothrombin fragments F1 and F2, thrombin-antithrombin complexes (TAT), and antithrombin III (ATIII) were determined. PA decreased significantly at 1 and 2 hours in patients treated by 10+10-U reteplase or by streptokinase. Fibrinogen binding to platelet GP IIb/IIIa followed a similar pattern. Significant thrombin generation and significantly elevated thrombin levels during thrombolysis were reflected by increased F1 and F2 fragments and TAT levels in all treatment groups. ATIII levels decreased significantly during thrombolytic therapy. CONCLUSIONS: A decrease in PA in patients treated by reteplase or streptokinase compared with alteplase could be observed in the early phase. Double bolus (10+10 U) reteplase and streptokinase resulted in lower PA at 1 and 2 hours than therapy with accelerated alteplase. Total fibrinogen and fibrinogen binding to GP IIb/IIIa tended to be lower during the first 2 hours after reteplase than after accelerated alteplase.


Subject(s)
Fibrinogen/metabolism , Myocardial Infarction/drug therapy , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombolytic Therapy , Double-Blind Method , Humans , Middle Aged , Myocardial Infarction/blood , P-Selectin/metabolism , Protein Binding , Recombinant Proteins/therapeutic use , Streptokinase/therapeutic use , Tissue Plasminogen Activator/therapeutic use
12.
Circulation ; 100(6): 659-65, 1999 Aug 10.
Article in English | MEDLINE | ID: mdl-10441105

ABSTRACT

BACKGROUND: Smooth muscle cell (SMC) proliferation is a critical component of neointimal formation in many models of vascular injury and in human lesions as well. Cell-cycle inhibition by gene transfer techniques can block SMC proliferation and lesion formation in animal models, although these methods are not yet applicable to the treatment of human disease. Flavopiridol is a recently identified, potent, orally available cyclin-dependent kinase inhibitor. METHODS AND RESULTS: Using human aortic SMCs, we found that flavopiridol in concentrations as low as 75 nmol/L resulted in nearly complete inhibition of basic fibroblast growth factor-induced and thrombin-induced proliferation. At this dose, flavopiridol inhibited cyclin-dependent kinase activity, as measured by histone H1 phosphorylation, but had no effect on mitogen-activated protein kinase activation. Induction of the cell cycle-related proteins cyclin D1, proliferating cell nuclear antigen, and phosphorylated retinoblastoma protein was also blocked by flavopiridol. Flavopiridol had no effect on cellular viability. To test whether flavopiridol had a similar activity in vivo when administered orally, we examined neointimal formation in rat carotid arteries after balloon injury. Flavopiridol 5 mg/kg reduced neointimal area by 35% and 39% at 7 and 14 days, respectively, after injury. CONCLUSIONS: Flavopiridol inhibits SMC growth in vitro and in vivo. Its oral availability and selectivity for cyclin-dependent kinases make it a potential therapeutic tool in the treatment of SMC-rich vascular lesions.


Subject(s)
Carotid Artery Injuries , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Growth Inhibitors/pharmacology , Muscle Proteins/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Piperidines/pharmacology , Wound Healing/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Carotid Arteries/drug effects , Carotid Arteries/enzymology , Carotid Arteries/pathology , Catheterization/adverse effects , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Humans , Hyperplasia , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Thrombin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Tunica Intima/drug effects , Tunica Intima/enzymology , Tunica Intima/injuries , Tunica Intima/pathology
13.
Thromb Haemost ; 82(1): 109-14, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10456463

ABSTRACT

Thrombolysis is well established in the treatment of acute myocardial infarction. However, clinical application of thrombolytic agents has limitations with respect to efficacy and specificity. To achieve highly effective and at the same time clot-selective plasminogen activation urokinase was coupled to a bispecific antibody consisting of the monovalent Fab' from the antifibrin monoclonal antibody 59D8 and the monovalent Fab' from the anti-glycoprotein GPIIb/IIIa monoclonal antibody 7E3. The bispecific antifibrin-antiplatelet urokinase conjugate (BAAUC) was synthesized and characterized. Assays with either immobilized platelets, GPIIb/IIIa or fibrin showed an increase in plasminogen activation compared to uncoupled urokinase by 10-fold, 58-fold and 13-fold, respectivley (p < 0.0001 each). In vitro clot lysis was performed on platelet-rich and fibrin-rich clots and revealed an up to 5-fold higher potency of BAAUC compared to uncoupled urokinase (p < 0.0001). In vitro platelet aggregation was effectively inhibited by the hybrid molecule, whereas urokinase had no effect. BAAUC and two monospecific urokinase-conjugates, UK-59D8-IgG and UK-7E3-(Fab')2 were compared with each other with regard to similar tests. In vitro clot assays with platelet-rich and platelet-poor clots were performed. BAAUC achieved a significantly higher plasminogen activation compared to each of the monospecific conjugates (p < 0.05, respectively). We conclude that BAAUC, a bispecific plasminogen activator with antifibrin and antiplatelet properties has the potency to lyse both fibrin-rich and platelet-rich thrombi with high efficacy and to effectively inhibit platelet aggregation.


Subject(s)
Antibodies, Bispecific/pharmacology , Blood Coagulation/drug effects , Immunoconjugates/pharmacology , Platelet Aggregation/drug effects , Blood Platelets/immunology , Fibrin/immunology , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/pharmacology , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/pharmacology
14.
J Biol Chem ; 274(28): 19814-22, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10391925

ABSTRACT

Thrombin is a potent vascular smooth muscle cell (VSMC) mitogen. Because recent evidence implicates reactive oxygen intermediates (ROI) in VSMC proliferation in general and atherogenesis in particular, we investigated whether ROI generation is necessary for thrombin-induced mitogenesis. Treatment of human aortic smooth muscle cells with thrombin increased DNA synthesis, an effect that was antagonized by diphenyleneiodonium but not by other inhibitors of cellular oxidase systems. This effect of thrombin was accompanied by increased O-2 and H2O2 generation and NADH/NADPH consumption. ROI generation in response to thrombin pretreatment could also be blocked by diphenyleneiodonium, suggesting that the NAD(P)H oxidase was necessary for ROI generation and thrombin-induced mitogenesis. Because of observed differences between the VSMC and neutrophil oxidase, we examined whether the cytosolic components of the phagocytic NAD(P)H oxidase were present in VSMC. p47(phox) and Rac2 were present in VSMC. Furthermore, thrombin increased expression of p47(phox) and Rac2 and stimulated their translocation to the cell membrane. We examined whether p47(phox) might be similarly regulated in vivo in a rat aorta balloon injury model and found that p47(phox) protein was increased after injury. Immunocytochemistry localized expression of p47(phox) to the neointima and media of injured arteries. Our data demonstrate that generation of O-2 and H2O2 is required for thrombin-mediated mitogenesis in VSMC and that p47(phox) is regulated by thrombin in vitro and is associated with vascular lesion formation in vivo.


Subject(s)
Muscle, Smooth, Vascular/enzymology , NADH, NADPH Oxidoreductases/metabolism , Phosphoproteins/metabolism , Reactive Oxygen Species/metabolism , Thrombin/pharmacology , Animals , Aorta , Cell Division/drug effects , Cells, Cultured , Enzyme Activation/drug effects , GTP-Binding Proteins/metabolism , Humans , Hydrogen Peroxide/metabolism , Mitogens/pharmacology , NAD/metabolism , NADP/metabolism , NADPH Oxidases , Rats , Superoxides/metabolism , rac GTP-Binding Proteins
15.
Thromb Haemost ; 82 Suppl 1: 14-8, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10695479

ABSTRACT

Cardiovascular disease is responsible for approximately 50% of total mortality in Europe, the USA and Japan. Established risk factors including smoking, hypercholesterolemia, and hypertension explain about half of the incidence of cardiovascular disease only. Reduced endogenous fibrinolytic activity secondary to increased plasma activity of plasminogen activator inhibitor type-1 (PAI-1) is now considered as a new cardiovascular risk factor. In this review, evidence is gathered for the notion that PAI-1 constitutes a predictor of cardiovascular disease and also contributes to the development of cardiovascular disease as a pathogenetic factor. The review will focus on experimental studies modulating PAI-1 activity and clinical studies addressing coronary heart disease, myocardial infarction, restenosis after coronary angioplasty, and graft occlusion after coronary artery bypass grafting.


Subject(s)
Cardiovascular Diseases , Plasminogen Activator Inhibitor 1/physiology , Animals , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Humans
16.
Thromb Haemost ; 82 Suppl 1: 32-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10695483

ABSTRACT

The initiating event of atherogenesis is thought to be an injury to the vessel wall resulting in endothelial dysfunction. This is followed by key features of atherosclerotic plaque formation such as inflammatory responses, cell proliferation and remodeling of the vasculature, finally leading to vascular lesion formation, plaque rupture, thrombosis and tissue infarction. A causative relationship exists between these events and oxidative stress in the vessel wall. Besides leukocytes, vascular cells are a potent source of oxygen-derived free radicals. Oxidants exert mitogenic effects that are partially mediated through generation of growth factors. Mitogens, on the other hand, are potent stimulators of oxidant generation, indicating a putative self-perpetuating mechanism of atherogenesis. Oxidants influence the balance of the coagulation system towards platelet aggregation and thrombus formation. Therapeutic approaches by means of antioxidants are promising in both experimental and clinical designs. However, additional clinical trials are necessary to assess the role of antioxidants in cardiovascular disease.


Subject(s)
Arteriosclerosis , Animals , Antioxidants/therapeutic use , Arteriosclerosis/complications , Arteriosclerosis/drug therapy , Arteriosclerosis/metabolism , Arteriosclerosis/physiopathology , Growth Substances/physiology , Humans , Oxidative Stress , Stress, Mechanical , Thrombosis/etiology , Thrombosis/physiopathology
17.
Thromb Haemost ; 82 Suppl 1: 38-43, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10695484

ABSTRACT

An increasing number of descriptive reports on soluble adhesion molecules and association with various diseases are published. Throughout these reports soluble adhesion molecules are identified as markers of inflammation. Since atherosclerosis demonstrates features of a chronic inflammatory disease, a potential association of soluble adhesion molecules with atherosclerosis has been postulated. However, conflicting results have been reported. One reason for this might be the differing definitions of atherosclerosis and patient groups. Besides the definition of atherosclerosis based on clinical symptoms, few reports use a direct quantification of atherosclerosis in their search for a marker of atherosclerosis. In those reports that quantify atherosclerosis, sVCAM-1 seems to be more specific for atherosclerosis than other markers. The serum level of sVCAM-1 appears to correlate with the extent of atherosclerosis and might allow for the detection of early stages of atherosclerosis. Large scale prospective studies will have to prove that sVCAM-1 can be used as a diagnostic tool for the detection of early stages of asymptomatic atherosclerosis and whether an early therapeutic intervention based on this approach is able to prevent progression and manifestation of the clinical sequelae of atherosclerosis.


Subject(s)
Arteriosclerosis/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Arteriosclerosis/physiopathology , Biomarkers , Humans
18.
Thromb Haemost ; 80(6): 881-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869154

ABSTRACT

RATIONALE: Reocclusion after thrombolysis diminishes the benefits of early reperfusion after acute myocardial infarction (AMI). No clinical or laboratory variables have been identified as predictors for reocclusion yet. METHODS AND RESULTS: To evaluate hemostatic variables as potential risk determinants platelet aggregation (PA, representing platelet activity), thrombin/antithrombin complexes (TAT, representing thrombin generation), and plasminogen activator inhibitor type 1 (PAI-1, representing endogenous fibrinolysis) were determined in 31 patients with AMI at 0, 1, 2. and 12 h after the start of thrombolysis as well as at hospital discharge. Reocclusion (defined as reinfarction or angiographically confirmed, clinically silent coronary reocclusion) occurred in 5 patients within 5-14 days and in 8 patients within 1 year. TAT plasma concentrations were lower in patients with reocclusion than in those without (9.9+/-5.7 vs. 22.9+/-22.2 ng/ml at 2 h, 6.5+/-3.1 vs. 1 1.2+/-6.4 ng/ml at 12 h, means+/-SD, p <0.05 each). Neither concentration nor activity of PAI-1 in plasma differed between both patient groups. However, both slope and maximum of PA (induced by 2 micromol/l ADP) were augmented in patients with reocclusion (slope: 39.4+/-1.7 vs. 32.5+/-7.4 at 2 h, p <0.001; 42.6+/-2.6 vs. 36.6+/-8.9 at 12 h, p <0.01). Results were independent of the thrombolytic agent used (alteplase or reteplase). A PA slope at 2 h higher than the average slope before thrombolysis (37.2+/-5.7) could be identified as best predictor for early (within 5-14 d, p=0.017, sensitivity 1.00, specificity 0.69) and late reocclusion (within 1 y, p=0.009, 0.88 and 0.74, respectively). CONCLUSIONS: Increased PA following coronary thrombolysis appears to be associated with early and late reocclusion. This marker could be useful in identifying patients who may benefit from more aggressive antiplatelet (such as GP IIb/IIIa receptor antagonists), interventional, or both strategies.


Subject(s)
Myocardial Infarction/blood , Plasminogen Activators/therapeutic use , Platelet Aggregation , Thrombolytic Therapy , Tissue Plasminogen Activator/therapeutic use , Adenosine Diphosphate/pharmacology , Adult , Aged , Aged, 80 and over , Anticoagulants/administration & dosage , Anticoagulants/therapeutic use , Antithrombin III/analysis , Aspirin/administration & dosage , Aspirin/therapeutic use , Coronary Vessels/pathology , Diabetes Mellitus/epidemiology , Drug Therapy, Combination , Female , Fibrinolysis , Heparin/administration & dosage , Heparin/therapeutic use , Humans , Male , Middle Aged , Myocardial Infarction/drug therapy , Myocardial Infarction/epidemiology , Obesity/epidemiology , Peptide Fragments , Peptide Hydrolases/analysis , Plasminogen Activator Inhibitor 1/analysis , Platelet Aggregation/drug effects , Predictive Value of Tests , Prothrombin , Recombinant Proteins/therapeutic use , Recurrence , Risk Factors , Smoking/epidemiology , Vascular Patency
19.
Circ Res ; 83(10): 1027-34, 1998 Nov 16.
Article in English | MEDLINE | ID: mdl-9815150

ABSTRACT

Recent studies have demonstrated that vascular smooth muscle cells are responsive to changes in their local hemodynamic environment. The effects of shear stress on the expression of human protease activated receptor-1 (PAR-1) and tissue plasminogen activator (tPA) mRNA and protein were investigated in human aortic smooth muscle cells (HASMCs). Under conditions of low shear stress (5 dyn/cm2), PAR-1 mRNA expression was increased transiently at 2 hours compared with stationary control values, whereas at high shear stress (25 dyn/cm2), mRNA expression was decreased (to 29% of stationary control; P<0.05) at all examined time points (2 to 24 hours). mRNA half-life studies showed that this response was not due to increased mRNA instability. tPA mRNA expression was decreased (to 10% of stationary control; P<0.05) by low shear stress after 12 hours of exposure and was increased (to 250% of stationary control; P<0.05) after 24 hours at high shear stress. The same trends in PAR-1 mRNA levels were observed in rat smooth muscle cells, indicating that the effects of shear stress on human PAR-1 were not species-specific. Flow cytometry and ELISA techniques using rat smooth muscle cells and HASMCs, respectively, provided evidence that shear stress exerted similar effects on cell surface-associated PAR-1 and tPA protein released into the conditioned media. The decrease in PAR-1 mRNA and protein had functional consequences for HASMCs, such as inhibition of [Ca2+] mobilization in response to thrombin stimulation. These data indicate that human PAR-1 and tPA gene expression are regulated differentially by shear stress, in a pattern consistent with their putative roles in several arterial vascular pathologies.


Subject(s)
Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Receptors, Thrombin/genetics , Tissue Plasminogen Activator/genetics , Aorta, Abdominal/cytology , Calcium/metabolism , Cell Division/drug effects , Cell Division/physiology , Child , Dactinomycin/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Hemostatics/pharmacology , Humans , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptor, PAR-1 , Stress, Mechanical , Thrombin/pharmacology , Thrombosis/physiopathology , Tissue Plasminogen Activator/metabolism
20.
Biochem J ; 330 ( Pt 3): 1469-74, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9494121

ABSTRACT

The mitogenic effects of thrombin are mediated by a G-protein-coupled receptor. Because the effects of thrombin are strongly influenced by the expression of its receptor, an understanding of its regulatory mechanisms is essential. To identify mechanisms of human thrombin receptor (HTR) gene regulation, a series of HTR-promoter-luciferase constructs were made and transfected into human microvascular endothelial cells for analysis. Deletion from bp -303 to -164 abolished reporter gene expression. Dimethyl sulphate treatment in vivo and DNase I footprinting in vitro demonstrated that a cluster of three GC box consensus sites was occupied, and electrophoretic mobility-shift assays established that Sp1 and Sp3 both bind to this 3' GC box cluster. We mutated each of the three GC boxes individually and all three collectively within this 3' cluster. Basal promoter activity was decreased to 46%, 78% and 29% of control for each of the GC boxes mutated individually, and to 6% when the three were mutated collectively. To test the individual abilities of Sp1 and Sp3 to activate or repress HTR transcription, we conducted co-transfection experiments with wild-type or mutated HTR-promoter-luciferase constructs. Co-transfection with Sp1 significantly augmented wild-type HTR promoter activity. Sp3 alone did not affect activity, and inhibited Sp1-mediated activation. Competition for shared binding sites by Sp1 and Sp3 might differentially regulate HTR expression in vascular endothelial cells.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation , Muscle, Smooth, Vascular/cytology , Receptors, Thrombin/biosynthesis , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Aorta , Cells, Cultured , DNA-Binding Proteins/biosynthesis , Endothelium, Vascular/cytology , Humans , Luciferases/biosynthesis , Microcirculation , Polymerase Chain Reaction , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Sequence Deletion , Sp1 Transcription Factor/biosynthesis , Sp3 Transcription Factor , Transcription Factors/biosynthesis , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...