Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters








Publication year range
1.
FASEB J ; 22(12): 4146-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18708591

ABSTRACT

Adipose tissue is an active endocrine organ that produces a variety of secretory factors involved in the initiation of angiogenic processes. The bioactive peptide apelin is the endogenous ligand of the G protein-coupled receptor, APJ. Here we investigated the potential role of apelin and its receptor, APJ, in the angiogenic responses of human endothelial cells and the development of a functional vascular network in a model of adipose tissue development in mice. Treatment of human umbilical vein endothelial cells with apelin dose-dependently increased angiogenic responses, including endothelial cell migration, proliferation, and Matrigel(R) capillary tubelike structure formation. These endothelial effects of apelin were due to activation of APJ, because siRNA directed against APJ, which led to long-lasting down-regulation of APJ mRNA, abolished cell migration induced by apelin in contrast to control nonsilencing siRNA. Hypoxia up-regulated the expression of apelin in 3T3F442A adipocytes, and we therefore determined whether apelin could play a role in adipose tissue angiogenesis in vivo. Epididymal white adipose tissue (EWAT) transplantation was performed as a model of adipose tissue angiogenesis. Transplantation led to increased apelin mRNA levels 2 and 5 days after transplantation associated with tissue hypoxia, as evidenced by hydroxyprobe staining on tissue sections. Graft revascularization evolved in parallel, as the first functional vessels in EWAT grafts were observed 2 days after transplantation and a strong angiogenic response was apparent on day 14. This was confirmed by determination of graft hemoglobin levels, which are indicative of functional vascularization and were strongly increased 5 and 14 days after transplantation. The role of apelin in the graft neovascularization was then assessed by local delivery of stable complex apelin-targeting siRNA leading to dramatically reduced apelin mRNA levels and vascularization (quantified by hemogloblin content) in grafted EWAT on day 5 when compared with control siRNA. Taken together, our data provide the first evidence that apelin/APJ signaling pathways play a critical role in the development of the functional vascular network in adipose tissue. In addition, we have shown that adipocyte-derived apelin can be up-regulated by hypoxia. These findings provide novel insights into the complex relationship between adipose tissue and endothelial vascular function and may lead to new therapeutic strategies to modulate angiogenesis.


Subject(s)
Adipose Tissue, White/physiology , Carrier Proteins/physiology , Endothelial Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Neovascularization, Physiologic/physiology , Receptors, G-Protein-Coupled/physiology , 3T3 Cells , Adipokines , Adipose Tissue, White/transplantation , Animals , Apelin , Apelin Receptors , Cell Movement , Down-Regulation , Humans , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/pharmacology
2.
J Thromb Haemost ; 6(10): 1697-706, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18647228

ABSTRACT

OBJECTIVE: In order to obtain a neutralizable antithrombotic, a chimeric molecule (SSR126517E) containing the sequence of a long-lasting antithrombin (AT)-dependent anti-factor Xa pentasaccharide, idraparinux, linked to a biotin molecule was synthesized and tested for anticoagulant and antithrombotic activity. METHODS: SSR126517E was tested in several models in vitro and in vivo for its pharmacological properties as well as its ability to be neutralized by avidin. RESULTS: SSR126517E displayed exactly the same properties as idraparinux. In vitro, SSR126517E had a very high affinity for AT (K(d) < 1 nm) and showed a potent anti-FXa effect and inhibition of thrombin generation with IC(50) values similar to those of idraparinux. Ex vivo, after intravenous administration to rats, SSR126517E produced a potent and long-lasting anti-FXa effect comparable to that obtained with idraparinux; as with idraparinux, the subcutaneous bioavailability was 100%. In vivo, SSR126517E was a potent antithrombotic in rat and mouse venous and arterial thrombosis models. Direct comparison in rats showed that SSR126517E was as active as idraparinux, when administered at the same molar dose. Furthermore, injection of avidin triggered the immediate elimination of SSR126517E from the bloodstream, resulting in complete neutralization of the antithrombotic activity of SSR126517E. CONCLUSIONS: These results show for the first time that coupling an oligosaccharide with biotin has no effect on the former's pharmacokinetic and pharmacologic properties and renders neutralization easy by injection of avidin.


Subject(s)
Anticoagulants/pharmacokinetics , Biotinylation , Fibrinolytic Agents/pharmacokinetics , Oligosaccharides/chemistry , Oligosaccharides/pharmacokinetics , Thrombosis/drug therapy , Animals , Antithrombin III/metabolism , Avidin/administration & dosage , Avidin/pharmacology , Factor Xa Inhibitors , Mice , Oligosaccharides/therapeutic use , Rats , Thrombin/biosynthesis
3.
J Thromb Haemost ; 2(6): 925-30, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140128

ABSTRACT

The activity of SanOrg123781A, a new synthetic antithrombotic drug inhibiting both factor Xa and thrombin through antithrombin (AT), was compared to that of unfractionated heparin (UFH) and of the synthetic pentasaccharide (fondaparinux, SP) in an ex vivo arterial thrombosis model in the pig. Six groups of four pigs were administered intravenously with SanOrg123781A (1, 3, 10 and 30 nmol kg(-1)), UFH (30 nmol kg(-1)) or SP (30 nmol kg(-1)). In this arterial model in which platelet thrombus was formed on a thrombogenic surface under a constant high shear rate, UFH and SP had moderate antithrombotic effects while SanOrg123781A exhibited a strong, dose-dependent inhibitory activity on platelet adhesion and platelet thrombus formation. In contrast to UFH, SanOrg123781A did not modify the activated partial thromboplastin time (aPTT) even at 30 nmol kg(-1), but strongly inhibited thrombin generation. At the same dose, despite a lower antithrombotic activity than SanOrg123781A, UFH significantly affected all the coagulation parameters. Taken together, these results show that SanOrg123781A, due to its potent and selective antifactor Xa and antifactor IIa activities is a promising new antithrombotic agent even in arterial setting.


Subject(s)
Polysaccharides/pharmacokinetics , Thrombosis/drug therapy , Animals , Blood Platelets/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Factor Xa Inhibitors , Fondaparinux , Heparin/administration & dosage , Heparin/pharmacokinetics , Heparin/pharmacology , Partial Thromboplastin Time , Polysaccharides/administration & dosage , Polysaccharides/pharmacology , Prothrombin/antagonists & inhibitors , Swine , Thrombosis/prevention & control
4.
J Thromb Haemost ; 1(9): 1959-65, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941037

ABSTRACT

Factor (F)Xa and thrombin bound to the clot during its formation contribute to the propensity of thrombi to activate the coagulation system. The aim of this work was to study the inhibition of clot-bound FXa and clot-bound thrombin by SanOrg123781A, a synthetic hexadecasaccharide that enhances the inhibition of thrombin and FXa by antithrombin (AT). SanOrg123781A, designed to exhibit low non-specific binding to proteins other than AT, was compared with heparin. In buffer, heparin and SanOrg123781A inhibited FXa and thrombin at similar concentrations [concentration inhibiting 50% (IC50) of Xa and IIa activity were, respectively: heparin 120 +/- 7 and 3 +/- 1 ng mL-1; SanOrg123781A 77 +/- 5 and 4 +/- 1 ng mL-1]. In human plasma, the activity of both compounds was reduced, although the activity of heparin was much more affected than that of SanOrg123781A (IC50 values for inhibition of FXa and FIIa activity were, respectively: heparin 100 +/- 5 and 800 +/- 40 ng mL-1; SanOrg123781A 10 +/- 5 and 30 +/- 3 ng mL-1). We demonstrated, in agreement with our previous results, that the procoagulant activity of the clot is essentially due to clot-bound FXa and to some extent to clot-bound thrombin. We showed that heparin and SanOrg123781A were able to inhibit fragment F1+2 generation induced by clot-bound FXa with IC50 values of 2 +/- 0.5 micro g mL-1 and 0.6 +/- 0.2 micro g mL-1, respectively. Both compounds also inhibited clot-bound thrombin activity, the IC50 values of heparin and SanOrg123781A being 1 +/- 0.01 micro g mL-1 and 0.1 +/- 0.1 micro g mL-1, respectively. Moreover, both heparin and SanOrg123781A significantly inhibited fibrinopeptide A generated by the action of clot-bound thrombin on fibrinogen but also by free thrombin generated from prothrombin by clot-bound FXa with IC50 values of 4 +/- 0.6 and 1 +/- 0.1 micro g mL-1, respectively. As with clot-bound enzymatic activities, SanOrg123781A was three times more active than heparin in vivo on fibrinogen accretion onto a pre-existing thrombus and as activators of recombinant tissue-type plasminogen activator-induced thrombolysis. In conclusion, due to the specific activities of SanOrg123781A, this compound is much more active than heparin in the presence of plasma proteins, on clot-bound enzymes and in in vivo models of thrombosis/thrombolysis.


Subject(s)
Blood Coagulation , Factor Xa/metabolism , Polysaccharides/pharmacology , Thrombin/metabolism , Animals , Antithrombin III/physiology , Factor Xa Inhibitors , Fibrinopeptide A/antagonists & inhibitors , Heparin/pharmacology , Humans , Inhibitory Concentration 50 , Male , Peptide Fragments/antagonists & inhibitors , Prothrombin/antagonists & inhibitors , Rabbits , Thrombin/antagonists & inhibitors , Thrombolytic Therapy , Thrombosis/drug therapy
5.
J Thromb Haemost ; 1(9): 2008-13, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941044

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a serious secondary event encountered in the clinical use of heparin. HIT results from the consumption of platelets that are immunologically activated by antibodies directed against complexes formed by platelet factor 4 (PF4) and sulfated polysaccharides that activate platelet aggregation, leading to paradoxical, life-threatening thrombosis. There is strong evidence that the ability of heparin and related compounds to induce HIT is closely linked to the structure of the polysaccharide, and particularly to its negative charge and to the length of the molecule. To test this hypothesis, we synthesized two sulfated oligosaccharides: SanOrg123781, a 16-mer, presenting two terminal charged domains separated by a 7-mer neutral linker, and SR121903, a highly sulfated 17-mer. Both of them displayed strong anti-factor (F) Xa and anti-FIIa activities but their affinities for PF4 were markedly different. SR121903 displaced PF4-bound heparin, whereas SanOrg123781 did not, underlining the importance of the charge of the molecule for the interaction with PF4. Platelet studies, in the presence of HIT serum, showed that SR121903 induced the secretion of platelet-dense granules (measured by the release of serotonin) whereas SanOrg123781 did not, a result in accordance with an absence of affinity of this molecule for PF4. These results were confirmed by measurements of platelet activation by flow cytometry (measured by annexin V binding, CD62 detection and activation of the GpIIb-IIIa complexes). In conclusion, we have demonstrated the importance of the charge of the polysaccharides in the HIT-induced platelet reactions measured by diverse methods, of which some are described for this purpose for the first time.


Subject(s)
Heparin/adverse effects , Oligosaccharides/pharmacology , Platelet Activation/drug effects , Thrombocytopenia/etiology , Cell Degranulation/drug effects , Dose-Response Relationship, Drug , Heparin/chemistry , Humans , Platelet Factor 4/metabolism , Polysaccharides/pharmacology , Static Electricity , Structure-Activity Relationship , Thrombocytopenia/blood , Thrombocytopenia/chemically induced
7.
Thromb Haemost ; 88(3): 432-5, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12353072

ABSTRACT

We investigated the effect of various oligosaccharides (OS) on the inhibition of factor IXa by antithrombin (AT) in a purified system. The OS comprised the AT-binding pentasaccharide sequence prolonged by saccharide chains with various lengths and charges. We show that factor IXa inhibition depended on the molecular weight of the OS. Factor IXa was not inhibited by the AT-binding pentasaccharide alone, but was inhibited if it was prolonged by a sulphated dodecasaccharide at the non-reducing end. The overall charge was also important since factor IXa inhibition was negligible if the pentasaccharide was prolonged by a non-sulphated dodecasaccharide. Using compounds containing a non-sulphated spacer, we showed that the central part of the OS was not critical. This study therefore demonstrates that the minimal OS structure necessary for catalysing factor IXa inhibition by AT is close to that required for catalysing thrombin inhibition.


Subject(s)
Factor IXa/antagonists & inhibitors , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Antithrombin III/pharmacology , Carbohydrate Sequence , Drug Design , Drug Interactions , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heparin/chemistry , Heparin/pharmacology , Humans , Structure-Activity Relationship
8.
Thromb Haemost ; 87(6): 985-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12083506

ABSTRACT

Pharmacokinetics of oligosaccharides displaying various affinities for antithrombin (AT) allowed us to show that there was a close relationship between the plasma half-life of these antithrombotic oligosaccharides and their affinity for AT. Recently, we have described new heparin mimetics comprising an AT binding domain and a thrombin binding domain separated by a neutral methylated saccharide sequence. These compounds displayed strong anti-Xa and antii-IIA activities and, in contrast to heparin, escaped neutralisation by platelet factor 4. The aim of this work was to compare the pharmacokinetics of several of these heparin mimetics in rats. These compounds differed by their length, charge and affinity for AT (AT-binding domain). The results obtained indicate that the prolongation of the AT binding domain did not modify significantly their affinities for AT. However, an increase in the number of charges leads to a decrease in the half-life. When a methylated spacer was added in order to minimise the non-specific interactions to other proteins, half-lives of the heparin mimetic were in the same range than that of the pentasaccharide used as an AT binding domain. In conclusion, this study shows the influence of the charge of the oligosaccharides on their pharmacokinetics and underlinesthe importance of minimising their non-specific binding to plasma proteins in order to obtain compounds with predictive pharmacokinetics.


Subject(s)
Anticoagulants/pharmacokinetics , Drug Design , Heparin/pharmacokinetics , Animals , Anticoagulants/pharmacology , Antithrombin III/metabolism , Binding Sites , Factor Xa Inhibitors , Half-Life , Heparin/analogs & derivatives , Heparin/pharmacology , Humans , Molecular Mimicry , Oligosaccharides/chemistry , Oligosaccharides/pharmacokinetics , Oligosaccharides/pharmacology , Platelet Factor 4/metabolism , Protein Binding , Prothrombin/antagonists & inhibitors , Structure-Activity Relationship , Thrombin/metabolism
9.
Thromb Haemost ; 87(2): 238-44, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11858483

ABSTRACT

The effect of new heparin mimetics (synthetic oligosaccharides) was studied in vitro with regard to thrombin generation (TG) in rat platelet rich plasma (PRP) and whole blood (WB) and in vivo on stasis-induced venous thrombosis in the rat. TG in PRP and in WB was highly dependent on platelet count and strongly influenced by the haematocrit. The peak of TG appeared to be significantly higher in WB than in PRP whereas the endogenous thrombin potential (ETP) was not significantly different under either condition. The effect of hirudin, the synthetic pentasaccharide SR90107/Org31540 (SP) and heparin were measured on TG in PRP and WB. We then compared the effect of two new synthetic heparin mimetics (SR121903A and SanOrg123781) with potent and comparable antithrombin (AT) mediated activity against factor Xa and thrombin. These two compounds were made of a pentasaccharide with a high affinity to AT, prolonged at the non-reducing end by an oligosaccharide chain recognised by thrombin. In SR121903A, the charge density and charge distribution was analogous to that of heparin whereas in SanOrg123781 the charges were only located on the last 5 saccharides of the non-reducing end of the molecule. In PRP and in WB, SR121903A acted on the lag time and on the AUC whereas SanOrg123781 inhibited thrombin formation with no effect on the lag time. SanOrg123781 was more potent in inhibiting TG than SR121903A. This difference was due to the structures of the compounds that differed in their ability to be neutralised by platelet factor 4. The antithrombotic effect of the two compounds was examined in a venous thrombosis model in rats. We observed that SanOrg123781 was more active than SR121903A and heparin. Taken together, these results indicate that the activity of oligosaccharides is greatly influenced by the global charge density of the molecule and show that SanOrg123781 is a potent and promising antithrombotic drug candidate.


Subject(s)
Anticoagulants/pharmacology , Factor Xa Inhibitors , Oligosaccharides/pharmacology , Polysaccharides/pharmacology , Thrombin/antagonists & inhibitors , Venous Thrombosis/prevention & control , Animals , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Anticoagulants/therapeutic use , Antithrombin III/metabolism , Binding Sites , Blood , Blood Platelets/physiology , Carbohydrate Conformation , Carbohydrate Sequence , Drug Evaluation, Preclinical , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Hematocrit , Heparin/chemistry , Heparin/pharmacology , Hirudins/pharmacology , Male , Models, Animal , Molecular Sequence Data , Molecular Structure , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry , Oligosaccharides/therapeutic use , Plasma , Platelet Factor 4/pharmacology , Polysaccharides/chemical synthesis , Polysaccharides/chemistry , Polysaccharides/therapeutic use , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Static Electricity , Structure-Activity Relationship , Thrombin/metabolism , Thromboplastin/pharmacology , Venous Thrombosis/etiology
10.
Thromb Haemost ; 85(5): 852-60, 2001 May.
Article in English | MEDLINE | ID: mdl-11372679

ABSTRACT

SR123781A, a synthetic hexadecasaccharide comprising an antithrombin (AT) binding domain, a thrombin binding domain, and a neutral methylated hexasaccharide sequence, was obtained from glucose through a convergent synthesis. SR123781A showed high affinity for human AT (Kd = 58 +/- 22 nM) and was a potent catalyst of its inhibitory effect with regard to factor Xa (IC50) = 77 +/- 5 ng/ml - 297 +/- 13 U/mg) and thrombin (IC50 = 4.0 +/- 0.5 ng/ml - 150 +/- 30 U/mg). SR123781A which acted exclusively via AT (no effect via heparin cofactor II at a concentration of 6 microg/ml) inhibited thrombin generation occurring via both the extrinsic and intrinsic pathways in vitro in human plasma. SR123781A did not compete for 3H-heparin binding to PF4 and did not activate platelets in the presence of plasma from patients with heparin-induced thrombocytopenia. After intravenous or subcutaneous administration to rats, rabbits or baboons, SR123781A displayed prolonged anti-factor Xa and anti-factor IIa activity ex vivo. After intravenous injection to baboons, decreases of the anti-factor Xa and anti-thrombin activities were parallel and disappeared with the same pharmacodynamics. Intravenous administrations of SR123781A strongly inhibited thrombus formation in an experimental model of thromboplastin-induced venous thrombosis in rats with an ED50 value of 18 +/- 0.1 microg/kg (vs 77 +/- 3 microg/kg for heparin). SR123781A inhibited arterial thrombus formation induced on a silk thread in an arterio-venous shunt and in the vena cava (ED50 values of 225 +/- 10 and 27 +/- 8 microg/kg, respectively). Compared to standard and low molecular weight heparin and to presently used drugs, SR123781A exhibited a highly favourable antithrombotic/bleeding ratio therefore showing that it might be considered as a promising compound in the treatment and prevention of various thrombotic diseases.


Subject(s)
Anticoagulants/chemical synthesis , Heparin/chemical synthesis , Polysaccharides/chemical synthesis , Animals , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Antithrombins/metabolism , Binding Sites , Carbohydrate Conformation , Carbohydrate Sequence , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Factor Xa/metabolism , Factor Xa Inhibitors , Heparin/administration & dosage , Heparin/pharmacokinetics , Humans , Molecular Mimicry , Molecular Sequence Data , Papio , Platelet Aggregation/drug effects , Polysaccharides/administration & dosage , Polysaccharides/pharmacokinetics , Rabbits , Rats , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Venous Thrombosis/drug therapy
11.
Chemistry ; 7(4): 858-73, 2001.
Article in English | MEDLINE | ID: mdl-11288878

ABSTRACT

Kinetic studies of thrombin inhibition by antithrombin in the presence of heparin have shown that thrombin binds to heparin in a preformed heparin-antithrombin complex. To study the relative position of the thrombin binding domain and the antithrombin binding domain on a heparin molecule we have designed and synthesized heparin mimetics, which structurally are very similar to the genuine polysaccharide. Their inhibitory properties with respect to factor Xa and thrombin provide experimental evidence that in heparin the thrombin binding domain must be located at the nonreducing end of the antithrombin binding domain to observe thrombin inhibition. As expected, factor Xa inhibition is not affected by elongation of the antithrombin binding pentasaccharide sequence, regardless of the position in which this elongation takes place.


Subject(s)
Antithrombins/chemistry , Heparin/chemistry , Antithrombins/chemical synthesis , Carbohydrate Conformation , Carbohydrate Sequence , Heparin/chemical synthesis , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data
12.
Chemistry ; 7(22): 4821-34, 2001 Nov 19.
Article in English | MEDLINE | ID: mdl-11763451

ABSTRACT

We have used organic synthesis to understand the role of L-iduronic acid conformational flexibility in the activation of antithrombin by heparin. Among known synthetic analogues of the genuine pentasaccharidic sequence representing the antithrombin binding site of heparin, we have selected as a reference compound the methylated anti-factor Xa pentasaccharide 1. As in the genuine original fragment, the single L-iduronic acid moiety of this molecule exists in water solution as an equilibrium between three conformers 1C4, 4C1 and 2S0. We have thus synthesized three analogues of 1, in which the L-iduronic acid unit is locked in one of these three fixed conformations. A covalent two atom bridge between carbon atoms two and five of L-iduronic acid was first introduced to lock the pseudorotational itinerary of the pyranoid ring around the 2S0 form. A key compound to achieve this connection was the D-glucose derivative 5 in which the H-5 hydrogen atom has been replaced by a vinyl group, which is a progenitor of the carboxylic acid. Selective manipulations of this molecule resulted in the 2S0-type pentasaccharide 23. Starting from the D-glucose derivative 28, a covalent two atom bridge was now built up between carbon atoms three and five to lock the L-iduronic acid moiety around the 1C4 chair form conformation, and the 1C4-type pentasaccharide 43 was synthesized. Finally the L-iduronic acid containing disaccharide 58 which, due to the presence of the methoxymethyl substituent at position five adopts a 4C1 conformation, was directly used to synthesize the 4C1-type pentasaccharide 61. The locked pentasaccharide 23 showed about the same activity as the reference compound 1 in an antithrombin-mediated anti-Xa assay, whereas the two pentasaccharides 43 and 61 displayed very low activity. These results clearly establish the critical importance of the 2S0 conformation of L-iduronic acid in the activation of antithrombin by heparin.


Subject(s)
Antithrombins/chemistry , Heparin/chemistry , Iduronic Acid/chemistry , Carbohydrate Sequence , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Sequence Data , Oligosaccharides/chemistry
13.
Blood ; 96(10): 3439-46, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071639

ABSTRACT

FcgammaRIIA, the only Fcgamma receptor present in platelets, is involved in heparin-associated thrombocytopenia (HIT). Recently, adenosine diphosphate (ADP) has been shown to play a major role in platelet activation and aggregation induced by FcgammaRIIA cross-linking or by sera from HIT patients. Herein, we investigated the mechanism of action of ADP as a cofactor in FcgammaRIIA-dependent platelet activation, which is classically known to involve tyrosine kinases. We first got pharmacologic evidence that the ADP receptor coupled to Gi was required for HIT sera or FcgammaRIIA clustering-induced platelet secretion and aggregation. Interestingly, the signaling from this ADP receptor could be replaced by triggering another Gi-coupled receptor, the alpha(2A)-adrenergic receptor. ADP scavengers did not significantly affect the tyrosine phosphorylation cascade initiated by FcgammaRIIA cross-linking. Conversely, the Gi-dependent signaling pathway, initiated either by ADP or epinephrine, was required for FcgammaRIIA-mediated phospholipase C activation and calcium mobilization. Indeed, concomitant signaling from Gi and FcgammaRIIA itself was necessary for an efficient synthesis of phosphatidylinositol 3,4,5-trisphosphate, a second messenger playing a critical role in the process of phospholipase Cgamma2 activation. Altogether, our data demonstrate that converging signaling pathways from Gi and tyrosine kinases are required for platelet secretion and aggregation induced by FcgammaRIIA.


Subject(s)
Antigens, CD/physiology , Calcium Signaling/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Receptors, IgG/physiology , Adenosine Diphosphate/pharmacology , Adrenergic alpha-Agonists/pharmacology , Anticoagulants/adverse effects , Antigens, CD/pharmacology , Blood Platelets/enzymology , Blood Platelets/metabolism , Blood Platelets/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Epinephrine/pharmacology , GTP-Binding Proteins/pharmacology , GTP-Binding Proteins/physiology , Heparin/adverse effects , Humans , Isoenzymes/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Phospholipase C gamma , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, alpha-2/physiology , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2/physiology , Signal Transduction/drug effects , Thrombocytopenia/chemically induced , Thrombocytopenia/pathology , Type C Phospholipases/drug effects , Type C Phospholipases/metabolism , Tyrosine/metabolism
14.
Arterioscler Thromb Vasc Biol ; 20(11): E107-12, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073863

ABSTRACT

In addition to its pivotal role in hemostasis, factor Xa binds to human umbilical vein endothelial cells through the recognition of a protein called effector cell protease receptor (EPR-1). This interaction is associated with signal transduction, generation of intracellular second messengers, and modulation of cytokine gene expression. Inhibitors of factor Xa catalytic activity block these responses, thus indicating that the factor Xa-dependent event of local proteolysis is absolutely required for cell activation. Because EPR-1 does not contain proteolysis-sensitive sites, we investigated the possibility that signal transduction by factor Xa requires proteolytic activation of a member of the protease-activated receptor (PAR) gene family. Catalytic inactivation of factor Xa by DX9065 suppressed factor Xa-induced increase in cytosolic free Ca(2+) in endothelial cells (IC(50)=0.23 micromol/L) but failed to reduce ligand binding to EPR-1. In desensitization experiments, trypsin or the PAR-2-specific activator peptide, SLIGKV, ablated the Ca(2+) signaling response induced by factor Xa. Conversely, pretreatment of endothelial cells with factor Xa blocked the PAR-2-dependent increase in cytosolic Ca(2+) signaling, whereas PAR-1-dependent responses were unaffected. Direct cleavage of PAR-2 by factor Xa on endothelial cells was demonstrated by cleavage of a synthetic peptide duplicating the PAR-2 cleavage site and by immunofluorescence with an antibody to a peptide containing the 40-amino acid PAR-2 extracellular extension. These data suggest that factor Xa induces endothelial cell activation via a novel cascade of receptor activation involving docking to EPR-1 and local proteolytic cleavage of PAR-2.


Subject(s)
Endothelium, Vascular/metabolism , Factor Xa/metabolism , Receptors, Cell Surface/metabolism , Receptors, Thrombin/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Calcium/metabolism , Cell Division/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Enzyme Activation/physiology , Humans , Inhibitor of Apoptosis Proteins , Kidney , Molecular Sequence Data , Receptor, PAR-2 , Serine Endopeptidases/metabolism , Survivin , Umbilical Veins
15.
Thromb Haemost ; 84(4): 668-74, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11057868

ABSTRACT

The aim of this study was to investigate the effect of factor Xa inhibitors on the prothrombinase activity of platelet-derived microparticles in vitro and in vivo. The factor Xa inhibitors studied were DX9065A (a direct factor Xa inhibitor) and Sanorg34006 (an antithrombin (AT)-mediated factor Xa inhibitor). Microparticles formed from the platelet surface following activation were isolated by size exclusion gel chromatography. After purification, their presence was detected by their procoagulant activity and by flow cytometry. Our results show that factor Xa and/or factor Va were present at the surface of the platelet-derived microparticles. Prothrombinase formed on the microparticles was inhibited by factor Xa inhibitors at IC50 values of 0.45+/-0.05 and 0.045+/-0.005 microM for DX9065A and AT-Sanorg34006 respectively. In an experiment aimed at determining the kinetics of microparticles formation we demonstrated that thrombin traces were sufficient to induce the formation of a significant quantity of microparticles. Both factor Xa inhibitors delayed the formation of microparticles by delaying thrombin generation. The thrombogenic effect of the microparticles were studied in vivo in a modified arterio-venous shunt model in the rat. In this model, the increase in the thrombus weigh due to microparticles or phospholipids did not differ significantly (33% and 23% respectively). In these conditions, prothrombinase activity seemed to play a lesser role in the thrombogenic effect than phospholipids. Nevertheless, factor Xa inhibitors were efficient and inhibited thrombus formation in a dose-dependent manner. These results demonstrate that platelet-derived microparticles display a potent prothrombotic effect in vivo and show that factor Xa inhibitors are potent antithrombotic compounds when thrombosis was induced by microparticles.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Platelets/physiology , Factor Xa Inhibitors , Fibrinolytic Agents/pharmacology , Naphthalenes/pharmacology , Oligosaccharides/pharmacology , Propionates/pharmacology , Animals , Cytoplasmic Granules/physiology , Male , Rats , Rats, Sprague-Dawley
16.
Ann Pharm Fr ; 57(3): 232-9, 1999 May.
Article in French | MEDLINE | ID: mdl-10427858

ABSTRACT

In the early eighties, following breakthroughs in oligosaccharide chemistry, the total chemical synthesis of pentasaccharides has been achieved, representing the antithrombin binding domain of heparin (the active site). The selective inhibitors of coagulation factor Xa thus obtained represent a new type of antithrombotic drugs. In a further step, based on the knowledge of the mechanism of antithrombin activation by heparin, oligosaccharides (pentadeca- to eicosasaccharides), comprising an antithrombin binding domain prolonged by a thrombin binding domain, were designed and synthesised in the Sanofi group. These compounds inhibit both factor Xa and thrombin, in the presence of antithrombin. Endowed with the full anticoagulant activity of heparin but devoid of undesired non-specific interactions, particularly with platelet factor 4 (PF4), they might represent "the ideal heparin-like antithrombotic".


Subject(s)
Factor Xa Inhibitors , Fibrinolytic Agents/chemical synthesis , Oligosaccharides/chemistry , Thrombin/antagonists & inhibitors , Animals , Carbohydrate Sequence , Humans , Molecular Sequence Data
17.
Thromb Haemost ; 81(6): 957-60, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10404775

ABSTRACT

Clopidogrel (25 mg/kg, p.o.), a potent and selective inhibitor of ADP-induced platelet aggregation, significantly inhibited, in the presence of platelets, ex vivo thrombin generation triggered by low concentrations of tissue factor. Clopidogrel reduced the area under the curve (23%, p <0.05) and the thrombin peak concentration (35%, p <0.05) but did not affect the lag phase of thrombin generation. Under the same experimental conditions, heparin (100 microg/ml) inhibited thrombin generation mostly by delaying and by reducing the burst of thrombin. In a stasis-induced venous thrombosis model in rats under low thrombogenic challenge, clopidogrel inhibited thrombus formation (ED50 = 7.9+/-1.5 mg/kg, p.o. - n = 10), confirming the existence of a close relationship between platelet activation and thrombin generation leading to blood coagulation and venous thrombosis.


Subject(s)
Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thrombin/biosynthesis , Ticlopidine/analogs & derivatives , Animals , Blood Platelets/pathology , Blood Platelets/physiology , Clopidogrel , Flow Cytometry , Male , Platelet Aggregation Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , Thrombosis/blood , Thrombosis/prevention & control , Ticlopidine/pharmacology , Ticlopidine/therapeutic use
18.
Bioorg Med Chem Lett ; 9(8): 1155-60, 1999 Apr 19.
Article in English | MEDLINE | ID: mdl-10328304

ABSTRACT

Synthetic pentadeca-, heptadeca- and nonadecasaccharides, comprising an antithrombin III (AT III) binding pentasaccharide prolonged at the non-reducing end by a thrombin binding domain have been obtained. The pentadecasaccharide is the shortest oligosaccharide able to catalyse thrombin inhibition by AT III. The nonadecasaccharide is a more potent thrombin inhibitor than standard heparin.


Subject(s)
Factor Xa Inhibitors , Heparin/analogs & derivatives , Heparin/chemical synthesis , Heparin/pharmacokinetics , Thrombin/antagonists & inhibitors , Carbohydrate Sequence , Models, Chemical , Molecular Sequence Data
19.
Bioorg Med Chem Lett ; 9(8): 1161-6, 1999 Apr 19.
Article in English | MEDLINE | ID: mdl-10328305

ABSTRACT

A synthetic heptadecasaccharide, comprising an antithrombin III binding domain, a thrombin binding domain, and a neutral methylated hexasaccharide sequence, was obtained through a convergent synthesis. This compound displayed in vitro anticoagulant properties similar to that of standard heparin but, in contrast with heparin, escaped neutralization by platelet factor 4, a protein released by activated platelets.


Subject(s)
Heparin/analogs & derivatives , Oligosaccharides/chemical synthesis , Oligosaccharides/pharmacokinetics , Antithrombin III/antagonists & inhibitors , Carbohydrate Sequence , Factor Xa Inhibitors , Molecular Sequence Data , Thrombin/antagonists & inhibitors
20.
Thromb Haemost ; 81(3): 400-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10102469

ABSTRACT

Thrombin generation (TG) initiated by diluted tissue-factor was investigated in whole human blood, in platelet-rich plasma (PRP), in platelet-poor plasma (PPP), and in PPP supplemented with red blood cells (RBCs). TG was characterized by the lag time preceding the thrombin burst and by the endogenous thrombin potential (ETP). RBCs at normal haematocrit were found to influence the lag time to the same extent as platelets. When TG was carried out in PRP or in PPP + RBCs, both the ETP and lag time were dependent on the platelet count or on the haematocrit, but the shapes of the dose-response curves were different. The inhibition of TG in PPP+ RBCs by two direct thrombin and factor Xa inhibitors: hirudin and DX 9065A, and two antithrombin III (AT)-dependent anticoagulants: heparin and SR 90107A was found to be similar to that previously described in PPP and in PRP: hirudin and DX 9065A only delayed TG whereas heparin and SR 90107A both delayed and decreased TG. FACscan analysis following labelling with FITC-annexin V or with phycoerythrin-labelled antiglycophorin A of samples taken in the course of TG initiated in PPP + RBCs showed that no significant haemolysis occurred and revealed that 0.51+/-0.075% (mean +/- sem, n = 3) of RBCs steadily exposed procoagulant phospholipids on their outer surface throughout the TG course. Furthermore, incubation of factors Xa and Va with washed RBCs sampled during TG in PPP +RBCs resulted in a significant and constant prothrombinase activity. Taken together, these data show for the first time that normal RBCs may participate in the haemostatic process through exposure of procoagulant phospholipids.


Subject(s)
Erythrocytes/metabolism , Hemostasis , Thrombin/biosynthesis , Blood Platelets/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL