Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
2.
J Thromb Haemost ; 3(7): 1437-45, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15978101

ABSTRACT

OBJECTIVES AND PATIENTS: We conducted a multicenter double-blind pharmacokinetic/pharmacodynamic (PK/PD) study of the new oral thromboxane receptor antagonist S18886 in 30 patients with peripheral artery disease, who were randomized to receive five different oral dosages of S18886 (1, 2.5, 5, 10 or 30 mg) for 12 weeks (83 days). Primary objective was to determine the effect of S18886 on platelet aggregation ex vivo. RESULTS: Pharmacokinetics of S18886 was linear, with peak plasma levels being reached between 30 min and 2 h and a terminal half-life of 5.8-10 h. No significant accumulation of S18886 in plasma was observed after repeated dosing. The relationship between the S18886 concentration and platelet inhibition was examined in terms of U46619-induced platelet aggregation. Over the range of doses studied, there was a predictable relation between the plasma drug concentration and the degree of platelet inhibition at each dose. Maximal inhibition of U46619-induced platelet aggregation was achieved within 1 h with all oral doses of S18886, and this effect was maintained for at least 12 h. The PK/PD relationship was direct, and U46619-induced platelet aggregation was strongly inhibited by S18886 plasma concentrations above 10 ng mL(-1). This concentration was thus the minimal effective antiplatelet level in this population, and was maintained only by the dosages of 10 and 30 mg. The safety profile of S18886 was excellent, whatever the unit dose, with no attributable adverse events. CONCLUSION: The results of this study, which included modeling and simulation, help identify the minimal effective plasma concentration of S18886 required for potent antiplatelet efficacy in patients with stable peripheral arterial disease.


Subject(s)
Naphthalenes/pharmacology , Naphthalenes/pharmacokinetics , Propionates/pharmacology , Propionates/pharmacokinetics , Receptors, Thromboxane/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/metabolism , Administration, Oral , Aged , Arachidonic Acid/metabolism , Area Under Curve , Collagen/metabolism , Female , Humans , Kinetics , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Time Factors
5.
J Thromb Haemost ; 2(7): 1163-6, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219200

ABSTRACT

BACKGROUND: Microbubbles used for echo-contrast agents accelerate enzymatic fibrinolysis of clots exposed to low-frequency ultrasound (US). It is not known whether microbubbles are also effective in enhancing high-frequency US-driven enzymatic fibrinolysis. METHODS AND RESULTS: Calibrated whole blood clots were exposed to US, or US and galactose-based microbubbles (Levovist), with or without recombinant tissue plasminogen activator (rt-PA) in an in-vitro flow system. We used low-intensity, 2-MHz, pulsed wave US. Relative weight reduction of clot +/- SD was 30.7 +/- 9.5% after exposure to microbubbles, rt-PA and US, 13.1 +/- 2.6% after exposure to rt-PA and US, 10.9 +/- 3.6% after exposure to microbubbles and US, and 6.1 +/- 1.9% after exposure to US alone. anova demonstrated a significant effect of rt-PA (P =0.001), microbubbles (P = 0.012), and interaction of both (P = 0.022). CONCLUSIONS: The application of galactose-based microbubbles (Levovist) strongly accelerates lysis of clots exposed to 2 MHz, low-intensity US in vitro both with and without rt-PA. The findings suggest a synergy between microbubbles and rt-PA. These methods routinely used for transcranial diagnostic applications have the potential to improve the efficacy of intravenous rt-PA in acute ischemic stroke.


Subject(s)
Fibrinolysis/radiation effects , Microbubbles , Tissue Plasminogen Activator/pharmacology , Ultrasonic Therapy/methods , Blood Coagulation/drug effects , Blood Coagulation/radiation effects , Contrast Media , Enzyme Therapy , Galactose , Humans , Models, Cardiovascular , Stroke/therapy , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/therapeutic use
6.
Ann Cardiol Angeiol (Paris) ; 51(3): 164-8, 2002 Jun.
Article in French | MEDLINE | ID: mdl-12471648

ABSTRACT

Vitamin K antagonists (VKA) decrease the synthesis of the active forms of four coagulation factors (factors II, VII, IX, X) and three inhibitors (proteins C, S, Z). There are VKA having a short half life (Sintrom, Pindione) and VKA having a long half life (Apegmone, Previscan, Coumadine). The treatment is monitored by the INR which in the majority of the indications must range between two and three. The first INR is usually performed 36 to 72 h after starting the treatment. There are a number of drug interactions. The rate of major bleedings range from 1.1 to 4.9 for 100 patient-year according to the published studies. Since around 600,000 patients are treated by VKA in our country, the absolute number of serious bleeding is high (> or = 17,000 per year). Anticoagulant clinics are structures aimed to instruct the patient and to advise the general practitioner to monitor the treatment, using computer assisted methods. It has been reported that these structures reduce the incidence of bleeding and of thrombotic events by 3 to 4 times.


Subject(s)
Anticoagulants/administration & dosage , Phenindione/analogs & derivatives , Acenocoumarol/administration & dosage , Administration, Oral , Drug Interactions , Family Practice , Food , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Humans , Patient Education as Topic , Phenindione/administration & dosage , Thrombosis/prevention & control , Time Factors , Vitamin K/antagonists & inhibitors , Warfarin/administration & dosage
9.
Blood ; 98(10): 3159-61, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698306

ABSTRACT

This study investigates whether the polymorphisms of 3 important platelet receptors affected experimental thrombus formation in men. Forty healthy male volunteers randomly recruited were genotyped for the variable number of tandem repeat (VNTR) of GPIbalpha, the -5T/C polymorphism in the Kozak sequence of GPIbalpha, the 807C/T polymorphism of GPIa, and the PI(A1)/PI(A2) polymorphism of GPIIb/IIIa. Platelet thrombus formation was induced ex vivo by exposing a collagen-coated coverslip in a parallel plate perfusion chamber to native blood for 4 minutes. The shear rates at the collagen surface were 650 and 2600 x s(-1). At 2600 x s(-1) platelet thrombus formation was significantly related only to the 807C/T polymorphism. In contrast, at 650 x s(-1) thrombus formation was significantly altered only by the Kozak sequence polymorphism. The VNTR and the PI(A1)/PI(A2) polymorphisms did not influence thrombus formation. Thus, platelet thrombus formation is significantly influenced by genetic variations of the GPIbalpha and GPIa receptors. The effect of these polymorphisms was dependent on the blood flow rate.


Subject(s)
Antigens, Human Platelet/genetics , Arterial Occlusive Diseases/genetics , Platelet Adhesiveness/genetics , Polymorphism, Genetic , Thrombosis/genetics , Adult , Amino Acid Substitution , Antigens, CD/genetics , Antigens, Human Platelet/physiology , Arterial Occlusive Diseases/blood , Genetic Predisposition to Disease , Genotype , Hemorheology , Humans , Integrin alpha2 , Male , Minisatellite Repeats , Perfusion , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Thrombosis/blood
10.
Semin Thromb Hemost ; 27(5): 519-22, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11668422

ABSTRACT

Low molecular weight heparins (LMWHs) are as efficient as unfractionated heparin (UFH) for prevention and treatment of thromboembolism. There is no evidence that monitoring the dose improves the clinical efficacy. In contrast, any overdosage increases the risk of hemorrhage. Because renal function plays a significant role in the elimination of LMWH, curative treatment should be monitored with an anti-factor Xa assay in patients presenting renal insufficiency, in the elderly, and in patients presenting an increased hemorrhagic risk. It is advisable to sample the patient at peak activity (3 to 5 hours after the subcutaneous [sc] administration) and to target the mean anti-factor Xa activity that was found efficient and safe in the clinical trial. This target is different for each LMWH and each dose regimen.


Subject(s)
Anticoagulants/pharmacokinetics , Drug Monitoring , Heparin, Low-Molecular-Weight/pharmacokinetics , Anticoagulants/administration & dosage , Anticoagulants/blood , Factor Xa/metabolism , Factor Xa Inhibitors , Heparin, Low-Molecular-Weight/administration & dosage , Heparin, Low-Molecular-Weight/blood , Humans , Thromboembolism/drug therapy
11.
Thromb Haemost ; 85(6): 1097-103, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11434691

ABSTRACT

A number of studies have reported conflicting data on the association of the PlA1/PlA2 polymorphism of the GPIIIa gene and coronary syndromes. We have investigated the effect of this polymorphism on experimental platelet thrombus formation in man. Forty healthy male volunteers were genotyped for the PlA1/PlA2 polymorphism. Thrombus formation was induced ex vivo by exposing a tissue factor (TF) or a collagen-coated coverslip in a parallel plate perfusion chamber to native blood for 2 and 4 min. The shear rates at these surfaces were 650 and 2,600 s(-1). Platelet and fibrin deposition was quantified by immunoenzymatic methods. The frequencies of PlA1/PlA1 and PlA1/PlA2 genotypes were 52.5% and 47.5%, respectively. Ex vivo deposition of fibrin on TF was not affected by the PlA1/PlA2 polymorphism. However, the ex vivo platelet deposition at 650 s(-1) was higher in blood from PlA1/PlA1 individuals than in PlA1/PlA2 individuals (P= 0.008 at 4 min). On collagen, neither fibrin nor platelet deposition was significantly affected by the PlA1/PlA2 polymorphism. Platelet thrombus formation is significantly influenced by genetic variations in the GPIIIa platelet receptor. This effect depends on the blood flow properties and the nature of the thrombogenic stimulus.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Thrombosis/etiology , Adult , Blood Flow Velocity , Collagen Type I/metabolism , Collagen Type I/pharmacology , Fibrin/drug effects , Fibrin/metabolism , Genotype , Humans , Male , Perfusion , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Polymorphism, Genetic/physiology , Protein Binding/drug effects , Protein Binding/genetics , Stress, Mechanical , Thromboplastin/metabolism , Thromboplastin/pharmacology , Thrombosis/genetics , Thrombosis/physiopathology
13.
Eur J Immunol ; 31(4): 1132-40, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11298338

ABSTRACT

Experimental allergic encephalomyelitis (EAE) is a T cell-mediated autoimmune disease induced in susceptible rat strains by a single immunization with myelin basic protein (MBP). The Lewis (LEW) strain is susceptible to disease induction while the Brown Norway (BN) strain is resistant. This resistance involves non-MHC genes since congenic BN-1L rats, with LEW MHC on a BN-derived background, are also resistant. In the present study we show that, upon immunization with MBP, the non-MHC-encoded resistance to develop clinical EAE in BN-1L rats is associated with a decreased production of IFN-gamma. This may be due to a difference between LEW and BN-1L rats in their ability to produce regulatory cytokines such as IL-4, IL-10 and TGF-beta. In comparison to LEW rats, immune lymph node cells from BN-1L rats express an increased amount of IL-4 mRNA but produce less IL-10. Furthermore, the sera from BN-1L rats contain higher amounts of active TGF-beta1. Therefore, we have investigated the involvement of IL-4 and TGF-beta in the resistance of BN-1L rats to develop EAE using neutralizing mAb. Neutralization of TGF-beta, but not IL-4, renders BN-1L rats susceptible to clinical EAE without affecting the proliferation or the cytokine repertoire of immune lymph node cells. With respect to the origin of the endogenous TGF-beta production, we excluded the involvement of CD8 T cells and discuss a possible role of platelets and of CD4 T cells exhibiting the CD45RC(low) phenotype.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Th1 Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/immunology , Blood Platelets/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Division , Disease Susceptibility/immunology , Female , Flow Cytometry , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-4/antagonists & inhibitors , Interleukin-4/genetics , Interleukin-4/immunology , Leukocyte Common Antigens/immunology , Lymph Nodes/immunology , Myelin Basic Protein/immunology , Neutralization Tests , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta1
14.
Br J Haematol ; 112(2): 506-12, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11167855

ABSTRACT

Active site-inhibited factor VIIa (FFR-rFVIIa) competes with factor VIIa (FVIIa) for binding to tissue factor (TF) and exerts an anti-thrombotic effect. We report an evaluation of the anti-thrombotic properties of FFR-rFVIIa in a model of thrombosis involving two thrombogenic surfaces. Uncoated glass capillaries or glass capillaries coated with TF were incorporated into an arterioarterial shunt in the rat and the occlusion time (OT) of the shunt was determined. An anti-thrombotic activity of FFR-rFVIIa was shown only on the TF-coated surface: the OT of the shunt was significantly prolonged, from 167 +/- 34 s in control animals to 312 +/- 42 s after i.v. bolus administration of 4 mg/kg FFR-rFVIIa. This OT was similar to those observed with the uncoated shunts in untreated animals (353 +/- 84 s). In vitro preincubation of the TF-coated shunt with FFR-rFVIIa significantly prolonged the OT to 245 +/- 45 s in the absence of detectable amounts of FFR-rFVIIa in the plasma. rFFR-rFVIIa weakly prolonged the tail template bleeding time by a factor of 1.5. This effect was more pronounced in animals pretreated with heparin. The anti-thrombotic and prohaemorrhagic effects of FFR-rFVIIa were totally reversed by administration of an equidose of rFVIIa. These results provide new information on the pharmacological properties of FFR-rFVIIa that will be useful for its clinical development.


Subject(s)
Factor VIIa/pharmacology , Fibrinolytic Agents/pharmacology , Thromboplastin/metabolism , Thrombosis/drug therapy , Animals , Bleeding Time , Blood Platelets , Fibrin/analysis , Hemorrhage/chemically induced , Male , Models, Animal , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Thrombosis/blood
15.
Arch Mal Coeur Vaiss ; 94(11 Suppl): 1220-4, 2001 Nov.
Article in French | MEDLINE | ID: mdl-11794962

ABSTRACT

Thrombosis is a multifactorial pathology. In addition to the environmental factors, there are genetic components which either predispose or protect against its occurrence. Some are common to only a small number of subjects: these are the high risk families to thrombotic disease. Others are present in a larger percentage of the general population: this is called polymorphism and its clinical expression is usually modest. The role of these genetic factors in the development of thrombosis is not easy to demonstrate, especially as there are many gene-gene and gene-environment interactions. Nevertheless, the genetic basis of venous thrombosis is well established and the value of this knowledge in the management of these patients is becoming recognised. On the other hand, there are many genetic factors in arterial pathology and the role of each taken in isolation is small. Well-targeted large scale trials are required to determine their effects.


Subject(s)
Genetic Predisposition to Disease , Thrombosis/genetics , Anticoagulants , Binding Sites , Blood Platelets , Factor V/genetics , Humans , Polymorphism, Genetic , Prothrombin/genetics , Risk Factors , Thrombosis/physiopathology
16.
Arzneimittelforschung ; 50(9): 821-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11050699

ABSTRACT

The pharmacodynamic pattern of low molecular weight dermatan sulphate (CAS 24967-94-0, Desmin-LMWDS) was studied in patients presenting chronic renal insufficiency. Three groups of six patients were defined according to their creatinine clearance: group 1, more than 50 ml/min, group 2 between 10 and 50 ml/min and group 3 lower than 10 ml/min (haemodialized patients). Desmin-LMWDS concentrations were determined with the Heptest assay and the chromogenic specific heparin cofactor II dependent anti IIa assay. In patients of group 1 affected by moderate renal insufficiency, the pharmacodynamic profiles were roughly comparable to those obtained in normal subjects. In the two other groups, the profiles were markedly modified by the renal insufficiency. The maximal concentrations were doubled and the areas under the time-activity curve were 4-fold higher in haemodialyzed (group 3) and severe renal insufficient patients (group 2) than in patients of group 1. The clearance of the anti IIa activity were 13.98 +/- 6.25 l/h; 4.12 +/- 2.64 l/h and 2.94 +/- 1.53 l/h and the half-lives were 2.79 +/- 2.60 h, 6.15 +/- 4.02 h and 11.51 +/- 6.54 h in groups 1 to 3, respectively (p < 0.05). The Desmin-LMWDS clearance was directly correlated to the creatinine clearance (r = 0.8244, n = 18, p < 0.001). Thus, as for low molecular weight heparin, renal function plays a major role in the elimination of low molecular weight dermatan sulphate.


Subject(s)
Dermatan Sulfate/pharmacokinetics , Kidney Failure, Chronic/metabolism , Adult , Aged , Area Under Curve , Dermatan Sulfate/administration & dosage , Factor Xa/metabolism , Female , Humans , Injections, Subcutaneous , Male , Middle Aged , Partial Thromboplastin Time , Prothrombin/metabolism , Thrombin Time
17.
Thromb Res ; 100(2): V113-20, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11053624

ABSTRACT

In many countries, low molecular weight heparins (LMWHs) have replaced unfractionated heparin (UH) for prevention and treatment of venous thromboembolism. The present paper reviews the possible advantages of LMWHs over UH. In spite of their lower molecular weight distribution, LMWHs are functionally more heterogeneous than UH. Their anti-Xa/anti-IIa ratio varies significantly, and the injection of the same dose generates different anti-Xa activities and activated partial thromboplastin time (APTT) prolongations. Their pharmacodynamic properties account for their more convenient use in comparison with UH; however, there is a risk of accumulation in case of renal insufficiency. Even if they are less anticoagulant on the basis of the APTT prolongation, they are not less prohemorrhagic than UH. LMWHs are probably less immunogenic and probably induce less osteoporosis. Several meta-analyses published between 1992 and 1999 indicate that LMWHs are as efficient as UH in preventing postoperative deep vein thrombosis (DVT) in general surgery and more efficient than UH in preventing DVT in orthopedic surgery and treating established DVT.


Subject(s)
Heparin, Low-Molecular-Weight/therapeutic use , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Anticoagulants/therapeutic use , Heparin/adverse effects , Heparin/pharmacokinetics , Heparin/therapeutic use , Heparin, Low-Molecular-Weight/adverse effects , Heparin, Low-Molecular-Weight/pharmacokinetics , Humans , Venous Thrombosis/complications , Venous Thrombosis/drug therapy , Venous Thrombosis/prevention & control
18.
Thromb Haemost ; 84(1): 129-36, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10928483

ABSTRACT

Tissue Factor (TF), the receptor for plasma VII/VIIa and the initiator of blood coagulation, is inducible in vascular smooth muscle cells (SMCs) by growth factors and bacterial lysopolysaccharides (LPS) and is expressed in vivo after vascular injury. As TF expression is a determinant of the thrombogenicity of vascular lesions, we investigated the signal pathways involved in this process. Human vascular SMCs were obtained from normal arteries and made quiescent by serum deprivation. Baseline TF antigen and activity were up-regulated by various agonists: fetal calf serum (FCS), LPS, and platelet derived growth factor (PDGF) being the most effective but with different kinetics. TF expression induced by LPS was transient with a maximum 6 h after stimulation and returned to baseline levels after 24 h whereas TF expression induced by serum or PDGF was sustained for at least 24 h. Rapid and transient activation of Extracellular signal-Regulated Kinase (ERK) was observed after stimulation by PDGF and FCS, but not by LPS. The role of ERK, Ras and protein kinase C activities were investigated using specific inhibitors, PD 98059, manumycin A and calphostin C respectively. For TF induction by LPS, PKC activity was required and the ERK/Ras pathway was not involved. In contrast, the effect of PDGF was strictly ERK and Ras dependent, but partially prevented by PKC inhibitors. TF induction by FCS was ERK dependent but partially Ras and PKC dependent. In conclusion, TF expression appears to be a non-specific response of SMCs to numerous stimuli through multiple signal pathways which differ according to the inducing agent.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinases/physiology , Muscle, Smooth, Vascular/drug effects , Protein Kinase C/physiology , Thromboplastin/biosynthesis , Adult , Animals , Cattle/blood , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media/pharmacology , Culture Media, Serum-Free , Enzyme Activation/drug effects , Fetal Blood/physiology , Fibroblast Growth Factor 2/pharmacology , Flavonoids/pharmacology , Genes, Immediate-Early , Humans , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/physiology , Mammary Arteries/cytology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Naphthalenes/pharmacology , Platelet-Derived Growth Factor/pharmacology , Polyenes/pharmacology , Polyunsaturated Alkamides , Protein Kinase C/antagonists & inhibitors , Thrombin/pharmacology
19.
Circulation ; 101(24): 2823-8, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10859288

ABSTRACT

BACKGROUND: We conducted a double-blind, randomized, crossover study to assess the antithrombotic effects of the combination of aspirin (acetylsalicylic acid, ASA) and clopidogrel, with or without a loading dose, versus ASA alone in a model of arterial thrombosis in humans. METHODS AND RESULTS: Eighteen male volunteers received the following 3 regimens for 10 days separated by a 1-month period: (1) 325 mg ASA daily, (2) 325 mg ASA+75 mg clopidogrel daily, (3) 325 mg ASA daily+300-mg clopidogrel loading dose on day 1 and +75 mg clopidogrel per day on days 2 to 10. The antithrombotic effect was measured 1.5, 6, and 24 hours after drug intake on day 1 and 6 hours after drug intake on day 10. Arterial thrombus formation was induced ex vivo by exposing a collagen-coated coverslip in a parallel-plate perfusion chamber to native blood for 3 minutes at an arterial wall shear rate. Without a loading dose, clopidogrel+ASA developed an antithrombotic effect within 6 hours after the first intake. It was superior to that produced by ASA, but it was moderate (P

Subject(s)
Aspirin/therapeutic use , Fibrinolytic Agents/therapeutic use , Platelet Aggregation Inhibitors/administration & dosage , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Adult , Arteries/drug effects , Clopidogrel , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Humans , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Ticlopidine/administration & dosage , Ticlopidine/therapeutic use , Time Factors
20.
J Immunol ; 164(7): 3822-8, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10725743

ABSTRACT

To determine whether polymorphonuclear leukocytes (PMN) modulate the production of tissue factor (TF) by monocytes, PBMC were incubated with increasing concentrations of PMN. PMN did not express any procoagulant activity. After 20-h cocultures, PMN enhanced or inhibited the TF production of PBMC, and this effect depended on the PMN/PBMC ratio. When the ratio increased from 1/1000 to 1/5, without or with LPS, the TF activity of PBMC increased to peak at 2.5-fold the baseline value (p < 0.01). The TF Ag and TF mRNA also increased. This potentiating effect was mediated by reactive oxygen species (ROS) released by PMN during the coculture; it did not require direct cell contact between PMN and PBMC, it was enhanced when PMN were stimulated by fMLP (a chemotactic peptide), and it was inhibited by two antioxidants, N-acetyl cysteine and pyrrolidine dithiocarbamate. In contrast, when the PMN/PBMC ratio was further increased from 1/2 to 2/1, the PBMC TF activity, Ag, and mRNA decreased and were inhibited compared with those of PBMC cultured alone (p < 0.01). This inhibitory effect required direct cell contact between PMN and PBMC, and it was not due to a PMN-mediated cytotoxicity. To confirm the role of ROS, H2O2 enhanced then inhibited the TF activity of PBMC in a dose-dependent manner, similarly to PMN. Thus, PMN may play an important role in the pathogenesis of thrombosis and atherosclerosis by exerting concentration-dependent regulatory effects on the TF production by PBMC via the release of ROS.


Subject(s)
Cell Communication/physiology , Leukocytes, Mononuclear/metabolism , Neutrophils/physiology , Reactive Oxygen Species/physiology , Thromboplastin/biosynthesis , Adult , Blood Coagulation/physiology , Cells, Cultured , Female , Humans , Leukocyte Count , Leukocytes, Mononuclear/physiology , Lymphocyte Activation , Male , Middle Aged , Neutrophil Activation/physiology , Neutrophils/metabolism , RNA, Messenger/blood , Thromboplastin/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...