Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 32(10): 2387-93, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22904276

ABSTRACT

OBJECTIVE: Protein S is a vitamin K-dependent plasma protein that functions in the feedback regulation of thrombin generation. Our goal was to determine how protein S regulates the intrinsic pathway of blood coagulation. METHODS AND RESULTS: We used plasma, including platelet-rich plasma, and in vitro methods to determine how the intrinsic pathway of blood coagulation is regulated by protein S. We obtained the following results: (1) activated partial thromboplastin time assays with protein S-supplemented plasma confirmed that protein S prolongs clotting time; (2) a modified activated partial thromboplastin time assay with factor IX (fIX)-deficient plasma confirmed that protein S affects fIX-initiated clotting; (3) a fIXa/factor VIIIa (fVIIIa)-mediated thrombin generation assay with either platelet-rich plasma or factor-deficient plasma, initiated with a limiting amount of tissue factor, was regulated by protein S; (4) in the presence of phosphatidylserine vesicles, protein S inhibited fIXa in the absence and presence of fVIIIa; and (5) protein S altered only the K(M) for factor X activation by fIXa in the absence of fVIIIa and both k(cat) and K(M) in the presence of fVIIIa. CONCLUSIONS: From our findings, it can be concluded that protein S inhibits fIXa in the presence or absence of fVIIIa in an activated protein C-independent way.


Subject(s)
Blood Coagulation/physiology , Factor IXa/antagonists & inhibitors , Factor VIIIa/antagonists & inhibitors , Protein C/physiology , Protein S/physiology , Factor IXa/physiology , Factor VIIIa/physiology , Feedback, Physiological/physiology , Humans , In Vitro Techniques , Partial Thromboplastin Time , Signal Transduction/physiology , Thrombin/physiology
2.
J Thromb Thrombolysis ; 27(1): 95-104, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18214639

ABSTRACT

The development of Cardiopulmonary Bypass (CPB) catopulted the field of cardiothoracic surgery into a new dimension--one that changed the lives of individuals with congenital and acquired heart disease worldwide. Despite its contributions, CPB has clear limitations and creates unique challenges for clinicians and patients alike, stemming from profound hemostatic pertubations and accompanying risk for bleeding and possibly thrombotic complications.


Subject(s)
Blood Coagulation , Blood Proteins/analysis , Cardiopulmonary Bypass/adverse effects , Coronary Artery Bypass/adverse effects , Fibrinolysis , Thrombophilia/etiology , Biomarkers , Blood Loss, Surgical , Cardiopulmonary Bypass/instrumentation , Coronary Artery Bypass, Off-Pump/statistics & numerical data , Coronary Disease/blood , Coronary Disease/surgery , Erythrocyte Transfusion/adverse effects , Extracorporeal Circulation/adverse effects , Extracorporeal Circulation/instrumentation , Factor IXa/analysis , Factor IXa/physiology , Graft Occlusion, Vascular/blood , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/physiopathology , Hemodilution/adverse effects , Humans , Intraoperative Period , Postoperative Hemorrhage/blood , Postoperative Hemorrhage/etiology , Postoperative Hemorrhage/physiopathology , Postoperative Period , Thrombin/metabolism , Thrombocytopenia/etiology , Thromboembolism/blood , Thromboembolism/etiology , Thromboembolism/physiopathology , Thrombophilia/blood , Thrombophilia/physiopathology , Transplantation, Autologous/adverse effects , Transplantation, Autologous/instrumentation
3.
Arterioscler Thromb Vasc Biol ; 22(3): 511-6, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11884299

ABSTRACT

Apparently quiescent, nonapoptotic endothelial cells mediate the activation of factor X by activated factor IX in the presence of its cofactor, activated factor VIII. In a previous study, we reported that during the activation of factor X, the interaction of the cofactor with the endothelial cell membrane clearly differs from the interaction of the cofactor with artificial lipid membranes. In the present study, we identified the peptide domain of factor VIII involved in the assembly of the enzyme-cofactor complex on the endothelial cell surface. With the use of monoclonal antibodies against different peptide sequences on the factor VIII light chain, it was observed that the lipid-binding region of the C2 domain on the factor VIII light chain mediates the assembly of the factor X-activating complex on the endothelial cell surface. In addition, a synthetic peptide that constitutes region Ala2318-Tyr2332 of the C2 domain and that is known for its ability to inhibit the binding of factor VIII to artificial lipid membranes also showed inhibition of the cofactor activity of factor VIII on endothelial cells. Thus, the carboxy-terminal part of the factor VIII light chain not only contains sites involved in lipid binding but also contains sites involved in complex assembly on the endothelial cell membrane.


Subject(s)
Endothelium, Vascular/physiology , Factor IXa/physiology , Factor VIII/chemistry , Factor VIII/physiology , Factor X/metabolism , Annexin A5/metabolism , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Endothelium, Vascular/drug effects , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Humans , Peptides/pharmacology , Phosphatidylserines/metabolism , Phospholipids/metabolism , Protein Structure, Tertiary
5.
Blood ; 97(10): 3117-22, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342438

ABSTRACT

Human coagulation factor XI (FXI) is a plasma serine protease composed of 2 identical 80-kd polypeptides connected by a disulfide bond. This dimeric structure is unique among blood coagulation enzymes. The hypothesis was tested that dimeric conformation is required for normal FXI function by generating a monomeric version of FXI (FXI/PKA4) and comparing it to wild-type FXI in assays requiring factor IX activation by activated FXI (FXIa). FXI/PKA4 was made by replacing the FXI A4 domain with the A4 domain from prekallikrein (PK). A dimeric version of FXI/PKA4 (FXI/PKA4-Gly326) was prepared as a control. Activated FXI/PKA4 and FXI/PKA4-Gly326 activate factor IX with kinetic parameters similar to those of FXIa. In kaolin-triggered plasma clotting assays containing purified phospholipid, FXI/PKA4 and FXI/PKA4-Gly326 have coagulant activity similar to FXI. The surface of activated platelets is likely to be a physiologic site for reactions involving FXI/FXIa. In competition binding assays FXI/PKA4, FXI/PKA4-Gly326, and FXI have similar affinities for activated platelets (K(i) = 12-16 nM). In clotting assays in which phospholipid is replaced by activated platelets, the dimeric proteins FXI and FXI/PKA4-Gly326 promote coagulation similarly; however, monomeric FXI/PKA4 has greatly reduced activity. Western immunoblot analysis confirmed that activated monomeric FXI/PKA4 activates factor IX poorly in the presence of activated platelets. These findings demonstrate the importance of the dimeric state to FXI activity and suggest a novel model for factor IX activation in which FXIa binds to activated platelets by one chain of the dimer, while binding to factor IX through the other.


Subject(s)
Blood Platelets/chemistry , Dimerization , Factor IX/physiology , Factor XIa/chemistry , Factor XIa/physiology , Blood Coagulation , Blood Platelets/physiology , Blotting, Western , Cell Membrane/chemistry , Enzyme Activation , Factor IXa/physiology , Factor XI/physiology , Factor XIa/genetics , Humans , Kinetics , Platelet Activation , Prekallikrein/chemistry , Prekallikrein/genetics , Protein Conformation , Recombinant Fusion Proteins
6.
Blood Coagul Fibrinolysis ; 11(1): 27-42, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10691097

ABSTRACT

The use of libraries of phage-displayed human single-chain antibody fragments (scFv) has become a new, powerful tool in rapidly obtaining therapeutically useful antibodies. Here, we describe the generation of human scFv and F(ab')2 directed against the gamma-carboxyglutamic acid (Gla) domain of coagulation factor IX. A large library of human scFv, displayed either on M13 phage or expressed as soluble proteins, was screened for binding to human Gla-domain peptide (Tyr1-Lys43). Among a panel of scFv that bound to the factor IX-Gla domain, six scFv clones recognized full-length factor IX and exhibited strong inhibitory activity of factor IX in vitro. After reformatting as F(ab')2, the affinity for factor IX of three selected clones was determined: 10C12 Kd = 1.6 nmol/l, 13D1 Kd = 2.9 nmol/l, and 13H6 Kd = 0.46 nmol/l. The antibodies specifically bound to factor IX and not to other coagulation factors, as assessed by enzyme-linked immunosorbent-type and human plasma clotting assays. The complementarity determining region amino acid sequences of clones 10C12 and 13D1 only differed at a single residue, whereas 13H6 showed little homology, suggesting that 13H6 binds to a different epitope within the factor IX-Gla domain. Despite the slightly lower affinity of 10C12 F(ab')2 versus 13H6 F(ab')2, 10C12 was consistently more potent than 13H6 in prolonging the activated partial thromboplastin time (APTT), in inhibiting platelet-mediated plasma clotting, and in inhibiting factor X activation by the intrinsic Xase complex. Finally, 10C12 F(ab')2 also recognized and neutralized factor IX/factor IXa of different species, as demonstrated by the specific APTT prolongation of dog, mouse, baboon and rabbit plasma. In summary, the results validate the usefulness of scFv phage-displayed libraries to rapidly generate fully human antibodies as potential new therapeutics for thrombotic disorders.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Factor IXa/immunology , Peptide Library , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antigen-Antibody Reactions/drug effects , Binding Sites, Antibody , Binding, Competitive , Blood Coagulation/drug effects , Blood Coagulation Factors/metabolism , Blood Platelets/physiology , Calcium/pharmacology , Cattle , Cloning, Molecular , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Epitopes/metabolism , Factor IXa/antagonists & inhibitors , Factor IXa/physiology , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Variable Region/isolation & purification , Immunoglobulin Variable Region/metabolism , Immunoglobulin Variable Region/pharmacology , Magnesium/pharmacology , Molecular Sequence Data , Sequence Alignment , Species Specificity
7.
Trends Cardiovasc Med ; 10(5): 198-204, 2000 Jul.
Article in English | MEDLINE | ID: mdl-11282295

ABSTRACT

Blood coagulation factor IX is activated during hemostasis by two distinct mechanisms. Activation through factor VIIa/tissue factor occurs early in the course of fibrin clot formation. Activation by factor XIa appears to be important for maintaining the integrity of the clot over time. In general, coagulation proteases are activated on a phospholipid surface in the presence of a protein cofactor. Until recently, activation of factor IX by factor XIa was thought to be the exception to this rule, as phospholipid has no effect on the reaction and no cofactor had been identified. These curious observations suggest that factor IX is activated by factor XIa in the fluid phase. A large amount of new evidence now indicates that factor IX activation by factor XIa occurs on the surface of activated platelets. The data suggest, however, that this reaction differs significantly from other protease-substrate interactions on the platelet surface. This is likely to be due, in part, to the unusual structure of the factor XI molecule.


Subject(s)
Enzyme Activation , Factor IX/physiology , Factor IXa/physiology , Amino Acid Sequence , Blood Platelets/physiology , Factor IX/genetics , Factor IXa/genetics , Humans , Molecular Sequence Data
10.
Thromb Haemost ; 82(2): 218-25, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10605707

ABSTRACT

Essential to hemostasis is the interaction of factor IXa with factor VIIIa. Recent studies indicate that helix-330 in the protease domain of factor IXa provides a critical binding site for factor VIIIa. Although weaker interactions cannot be ruled out, a primary role of the EGF1 domain of factor IXa in this context may be to serve as a spacer in properly positioning the factor IXa protease domain for optimal interaction with factor VIIIa. The role of the Gla domain, as well as of the EGF2 domain of factor IXa, in binding to factor VIIIa is not clear. The region of factor VIIIa that interacts with the protease domain of factor IXa is quite possibly located in the A2 domain. Furthermore, it should be noted (Table 1) that the corresponding helix residues in factor VIIa bind to tissue factor, and, in factor Xa, they are involved in binding to factor Va. Thus, a common function of this helix (162 in chymotrypsin numbering) in several blood coagulation proteases may be to serve as an anchoring point for the respective cofactor.


Subject(s)
Factor IXa/chemistry , Factor VIIIa/chemistry , Animals , Epidermal Growth Factor , Factor IX/genetics , Factor IXa/genetics , Factor IXa/physiology , Factor VIIIa/physiology , Hemophilia B/genetics , Humans , Models, Molecular , Mutation , Protein Binding , Protein Structure, Tertiary
11.
J Thorac Cardiovasc Surg ; 116(5): 860-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9806393

ABSTRACT

BACKGROUND: Multiple stimuli converge in cardiopulmonary bypass to create a tremendous prothrombotic stimulus. The ideal anticoagulant for cardiopulmonary bypass should selectively target only the intravascular stimuli, thereby eliminating pathologic clotting in the bypass circuit while preserving hemostasis in the thoracic cavity. We propose the inhibition of factor IX as such a targeted anticoagulant strategy. METHODS: We prepared an inhibitor of activated factor IX and applied it to a primate model of cardiopulmonary bypass to confirm the anticoagulant efficacy of activated factor IX in this setting and to assess more subtle markers of thrombin generation, macrophage procoagulant activity, and cellular tissue factor expression. Seven baboons that received activated factor IX (460 microg/kg) and 7 that received heparin (300 IU/kg) and protamine underwent cardiopulmonary bypass for 90 minutes and were followed after the operation for 3 hours. RESULTS: Analysis of plasma factor IX activity demonstrated adequate inhibition (<20%) of factor IX throughout cardiopulmonary bypass. Activated factor IX-treated baboons demonstrated similar circuit patency to heparin-treated baboons but had significantly diminished intraoperative blood loss. Preservation of extravascular hemostasis was further demonstrated in activated factor IX-treated animals by (1) significantly increased levels of thrombin-antithrombin III complex and prothrombin activation peptide (F1+2) without intravascular thrombosis, (2) significantly greater macrophage procoagulant activity in pericardial-derived monocytes, and (3) immunohistochemical evidence of tissue factor expression in pericardial mesothelial cells and macrophages. CONCLUSIONS: Anticoagulation with activated factor IX allows for intravascular anticoagulation with maintenance of extravascular hemostasis. These findings suggest activated factor IX as an agent that not only exemplifies a targeted approach to selective anticoagulation in cardiac surgery but also further characterizes the procoagulant milieu during cardiopulmonary bypass.


Subject(s)
Anticoagulants/pharmacology , Cardiopulmonary Bypass , Factor IX/antagonists & inhibitors , Factor IXa/antagonists & inhibitors , Animals , Blood Coagulation Tests , Blood Loss, Surgical/physiopathology , Cardiopulmonary Bypass/instrumentation , Factor IX/physiology , Factor IXa/physiology , Heparin/pharmacology , Humans , Macrophages/metabolism , Microscopy, Electron, Scanning , Papio , Surface Properties , Thrombin/metabolism , Thrombophilia/blood , Thromboplastin/metabolism
12.
Thromb Haemost ; 79(2): 296-301, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9493579

ABSTRACT

We have made use of a novel flow reactor to study the initiation and propagation of the ex vivo blood coagulation processes at artificial surfaces. The flow reactor consisted of a primary glass or polymer capillary that is connected to a secondary glass capillary, which inner wall was coated with a phospholipid bilayer of 25 mol% dioleoylphosphatidylserine/75 mol% dioleoylphosphatidylcholine (DOPS/DOPC). Citrated platelet free plasma and a CaCl2 solution were delivered by syringe pumps and mixed just before the entrance of the flow reactor. The outflowing plasma was assayed for factor XIa, factor IXa, factor Xa and thrombin activity. Perfusion of recalcified plasma through a bare glass capillary resulted in a transient generation of fluid phase factor XIa. In contrast, factor IXa production increased slowly to attain a stable steady-state level. We established that surface-bound factor XIa was responsible for a continuous production of factor IXa. Factor IXa-induced generation of factor Xa and thrombin was only observed when contact activated plasma was subsequently perfused through a DOPS/DOPC-coated capillary, showing that propagation of the factor IXa trigger requires a procoagulant, phosphatidylserine-containing, phospholipid membrane. The negatively charged inner surface of a heparin-coated polyurethane capillary, generated like the glass capillary significant amounts of factor XIa and factor IXa when perfused with recalcified plasma. No differences were found between unfractionated heparin and heparin devoid of anticoagulant activity. Thus, it is concluded that contact activation and factor IXa generation in flowing plasma is not inhibited by immobilised anticoagulant active heparin. Consequently, factor IXa-dependent thrombin generation at a downstream located phospholipid membrane was similar, regardless the specific anticoagulant activity of immobilised heparin.


Subject(s)
Blood Coagulation , Hemorheology/instrumentation , Factor IXa/physiology , Factor XIa/physiology , Factor Xa/physiology , Hemorheology/methods , Humans
13.
Thromb Res ; 84(4): 225-36, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8948047

ABSTRACT

We developed and analyzed the mathematical model of the intrinsic pathway based on the current biochemical data on the kinetics of blood coagulation individual stages. The model includes eight differential equations describing the spatio-temporal dynamics of activation of factors XI, IX, X, II, I, VIII, V, and protein C. The assembly of tenase and prothrombinase complexes is considered as a function of calcium concentration. The spatial dynamics of coagulation was analyzed for the one-dimensional case. We examined the formation of active factors, their spreading, and growth of the clot from the site of injury in the direction perpendicular to the vessel wall, into the blood thickness. We assumed that the site of injury (in the model one boundary of the space segment under examination) becomes a source of the continuous influx of factor XIa. In the first part, we described the model, selected the parameters, etc. In the second part, we compared the model with experimental data obtained in the homogeneous system and analyzed the spatial dynamics of the clot growth.


Subject(s)
Blood Coagulation/physiology , Calcium/metabolism , Factor IXa/physiology , Factor VIIIa/physiology , Factor Va/physiology , Factor Xa/metabolism , Humans , Kinetics , Models, Theoretical , Protein C/physiology , Thrombin/metabolism
14.
Blood ; 86(8): 3021-5, 1995 Oct 15.
Article in English | MEDLINE | ID: mdl-7579395

ABSTRACT

Several enzymes can activate factor VII in vitro, but the protease responsible for generating factor VIIa in vivo has not been determined. Using recombinant tissue factor that has undergone a COOH-terminal truncation, a sensitive functional assay has been established for measuring plasma factor VIIa levels. To evaluate the mechanism responsible for the generation of factor VIIa in vivo, we measured the levels of this enzyme after administering purified concentrates of factor IX and factor VIII to patients with severe deficiencies of these clotting factors. In patients with hemophilia B, factor VIIa levels were initially reduced to 0.5 +/- 0.1 ng/mL and gradually increased to normal after infusing 100 U/kg of body weight (BW) of factor IX. Despite these increases, there were no significant changes in the generation of factor Xa or thrombin. In patients with hemophilia A, only a slight reduction in factor VIIa levels (2.5 +/- 1.3 ng/mL) was observed as compared with controls (3.3 +/- 1.1 ng/mL) and no significant changes were observed after factor VIII levels were normalized. The administration of recombinant factor VIIa (10 micrograms/kg BW) to patients with factor VII deficiency increased the mean circulating level of the enzyme to 118 ng/mL, but this only resulted in normalization of the levels of the activation peptides of factor IX and factor X. The above data indicate that factor IXa is primarily responsible for the basal levels of free factor VIIa generated in vivo (ie, in the absence of thrombosis or provocative stimuli) and that changes in the plasma concentrations of free factor VIIa in the blood do not necessarily lead to alterations in the extent of factor X activation.


Subject(s)
Factor IXa/physiology , Factor VIIa/biosynthesis , Adolescent , Adult , Enzyme Activation , Factor IX/pharmacology , Factor IX/therapeutic use , Factor VIII/pharmacology , Factor VIII/therapeutic use , Factor VIIa/analysis , Factor VIIa/therapeutic use , Factor Xa/metabolism , Hemophilia A/metabolism , Hemophilia A/therapy , Hemophilia B/metabolism , Hemophilia B/therapy , Humans , Middle Aged , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombin/metabolism , Thromboplastin/pharmacology
15.
Blood ; 86(5): 1794-801, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7655009

ABSTRACT

Tissue factor is the major initiator of coagulation. Both factor IX and factor X are activated by the complex of factor VIIa and tissue factor (VIIa/TF). The goal of this study was to determine the specific roles of factors IXa and Xa in initiating coagulation. We used a model system of in vitro coagulation initiated by VIIa/TF and that included unactivated platelets and plasma concentrations of factors II, V, VIII, IX, and X, tissue factor pathway inhibitor, and antithrombin III. In some cases, factor IX and/or factor X were activated by tissue factor-bearing monocytes, but in some experiments, picomolar concentrations of preactivated factor IX or factor X were used to initiate the reactions. Timed samples were assayed for both platelet activation and thrombin activity. Factor Xa was 10 times more potent than factor IXa in initiating platelet activation, but factor IXa was much more effective in promoting thrombin generation than was factor Xa. In the presence of VIIa/TF, factor X was required for both platelet activation and thrombin generation, while factor IX was only required for thrombin generation. We conclude that VIIa/TF-activated factors IXa and Xa have distinct physiologic roles. The main role of factor Xa that is initially activated by VIIa/TF is to activate platelets by generating an initial, small amount of thrombin in the vicinity of platelets. Factor IXa, on the other hand, enhances thrombin generation by providing factor Xa on the platelet surface, leading to prothrombinase formation. Only tiny amounts of factors IX and X need to be activated by VIIa/TF to perform these distinct functions. Our experiments show that initiation of coagulation is highly dependent on activation of small amounts of factors IXa and Xa in proximity to platelet surfaces and that these factors play distinct roles in subsequent events, leading to an explosion of thrombin generation. Furthermore, the specific roles of factors IXa and Xa generated by VIIa/TF are not necessarily reflected by the kinetics of factor IXa and Xa generation.


Subject(s)
Blood Coagulation , Blood Platelets/physiology , Factor IXa/physiology , Factor Xa/physiology , Monocytes/physiology , Platelet Activation/physiology , Thromboplastin/physiology , Blood Platelets/drug effects , Blotting, Western , Cell Separation , Cells, Cultured , Factor IXa/biosynthesis , Factor IXa/pharmacology , Factor X/analysis , Factor X/biosynthesis , Factor Xa/pharmacology , Humans , Kinetics , Monocytes/cytology , Platelet Activation/drug effects , Thrombin/biosynthesis , Thromboplastin/pharmacology , Time Factors
17.
Blood ; 85(6): 1527-34, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7888673

ABSTRACT

The inhibition mechanism of a polysaccharide anticoagulant, depolymerized holothurian glycosaminoglycan (DHG), was examined by analyzing its effects on the clotting time of human plasma depleted of antithrombin III (ATIII), of heparin cofactor II (HCII), or of both heparin cofactors. The effect exerted by this agent on the activation of prothrombin and factor X in purified human components were also examined and all effects were compared with those of other glycosaminoglycans (GAGs). The capacity of DHG to prolong activated partial thromboplastin time was not reduced in ATIII-depleted, HCII-depleted, HCII-depleted, or ATIII- and HCII-depleted plasma, whereas its capacity to prolong prothrombin time and thrombin clotting time was reduced in HCII-depleted plasma. DHG inhibited the amidolytic activity of thrombin in the presence of HCII with a second order rate constant of 1.2 x 10(8) (mol/L)-1 min-1. These results indicated that DHG has two different inhibitory activities, one being an HCII-dependent thrombin inhibition and the other an ATIII- and HCII-independent inhibition of the coagulation cascade. The heparin cofactors-independent inhibitory activity of DHG was investigated in the activation of prothrombin by factor Xa and in the activation of factor X by tissue factor-factor VIIa complex or by factor IXa. DHG significantly inhibited the activation of factor X by factor IXa in the presence of factor VIIIa, but not in the absence of factor VIIIa. The interaction between DHG and factors IXa, VIIIa, and X was investigated with a DHG-cellulofine column, on which DHG had strong affinity for factors IXa and VIIIa. These findings show that the heparin cofactors-independent inhibition exhibited by DHG was caused by inhibition of the interaction of factor X with the intrinsic factor Xase complex, probably by binding to the factor IXa-factor VIIIa complex.


Subject(s)
Anticoagulants/pharmacology , Antithrombin III/physiology , Factor IXa/physiology , Factor VIIIa/physiology , Factor X/antagonists & inhibitors , Glycosaminoglycans/pharmacology , Heparin Cofactor II/physiology , Thrombin/antagonists & inhibitors , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Sea Cucumbers
18.
J Biochem ; 116(2): 335-40, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7822251

ABSTRACT

We studied the interaction of factor X activation peptide (XAP) with factor IXa and factor Xa and the effect of XAP on factor IXa-catalyzed activation of factor X. XAP associated with factor Xa in the presence of 5 mM Ca2+ was dissociated from factor Xa by gel chromatography using Ultrogel AcA54 in 5 mM EDTA, or in 8 M urea-0.1% SDS. An exogenous isolated XAP inhibited the factor IXa-catalyzed factor X activation both in the presence and absence of factor VIIIa. 4-Amidinophenylmethylsulfonyl (aPMS)-factor Xa independent of XAP also inhibited the factor X activation more effectively than XAP alone in the presence of factor VIIIa. However, aPMS-factor Xa independent of XAP hardly inhibited the factor X activation in the absence of factor VIIIa. The binding of 125I-labeled factor X to the aPMS-factor IXa fixed to a microwell plate was inhibited by unlabeled factor X or XAP, but not by aPMS-factor Xa with or without XAP. Factor IXa directly bound to XAP and aPMS-factor Xa with XAP, but did not bind to aPMS-factor Xa without XAP. These findings suggest that the region of XAP in factor X directly interacts with factor IXa, and factor Xa region other than XAP interacts with factor VIIIa. Desialation or deletion of N-linked carbohydrates of XAP reduced the inhibitory activity of XAP for the factor X activation by factor IXa to approximately 50% of that of the intact XAP. This suggests that the sialic acids in the carbohydrate chains of the XAP region partly contribute to the interaction with factor IXa during its activation.


Subject(s)
Factor IXa/physiology , Factor X/physiology , Factor Xa/physiology , Amino Acid Sequence , Animals , Cattle , Chromatography , Factor IXa/metabolism , Factor VIIIa/pharmacology , Factor X/metabolism , Factor Xa/metabolism , Factor Xa/pharmacology , Humans , Iodine Radioisotopes , Metalloendopeptidases/pharmacology , Molecular Sequence Data , Rabbits
19.
J Lab Clin Med ; 123(4): 585-93, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8145007

ABSTRACT

Factor VIII is an important cofactor in the intrinsic activation of factor X. To function effectively as a cofactor, factor VIII must be activated. In plasma, factor VIII circulates in a complex with von Willebrand factor, and although thrombin can activate complexed factor VIII, the activation by activated factor X is inhibited by von Willebrand factor. In this study, the effect of von Willebrand factor on the generation of factor Xa by the factor IXa-VIII complex was investigated. Purified human factors VIII, IXa, and X were incubated on human umbilical vein endothelial cells or phospholipid vesicles in the presence of calcium ions, and the generation of factor Xa was followed. In the presence of von Willebrand factor, a prolonged lag-phase and a dose-dependent inhibition of factor X activation was observed. These effects were not observed when von Willebrand factor was preincubated with a monoclonal antibody directed against von Willebrand factor that blocks factor VIII binding. When factor VIII was activated with thrombin before the incubation, neither the monoclonal antibody nor von Willebrand factor had an effect on the rate of factor X activation. Preincubation of endothelial cells with the monoclonal antibody resulted in a somewhat higher rate of factor X activation. When endothelial cells from a patient with von Willebrand's disease type I were used, preincubation of the monoclonal antibody had no effect on the rate of factor X activation. We conclude that von Willebrand factor on the surface of endothelial cells can modulate the intrinsic factor X activation. This effect is greatly enhanced, however, by the addition of exogenous von Willebrand factor.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Factor VIII/physiology , Factor Xa/metabolism , von Willebrand Factor/physiology , Cells, Cultured , Endothelium, Vascular/metabolism , Factor IXa/physiology , Humans , Time Factors , von Willebrand Diseases/blood
20.
J Exp Med ; 176(1): 27-35, 1992 Jul 01.
Article in English | MEDLINE | ID: mdl-1613461

ABSTRACT

Generation of coagulation factor Xa by the intrinsic pathway protease complex is essential for normal activation of the coagulation cascade in vivo. Monocytes and platelets provide membrane sites for assembly of components of this protease complex, factors IXa and VIII. Under biologically relevant conditions, expression of functional activity by this complex is associated with activation of factor VIII to VIIIa. In the present studies, autocatalytic regulatory pathways operating on monocyte and platelet membranes were investigated by comparing the cofactor function of thrombin-activated factor VIII to that of factor Xa-activated factor VIII. Reciprocal functional titrations with purified human factor VIII and factor IXa were performed at fixed concentrations of human monocytes, CaCl2, factor X, and either factor IXa or factor VIII. Factor VIII was preactivated with either thrombin or factor Xa, and reactions were initiated by addition of factor X. Rates of factor X activation were measured using chromogenic substrate specific for factor Xa. The K1/2 values, i.e., concentration of factor VIIIa at which rates were half maximal, were 0.96 nM with thrombin-activated factor VIII and 1.1 nM with factor Xa-activated factor VIII. These values are close to factor VIII concentration in plasma. The Vsat, i.e., rates at saturating concentrations of factor VIII, were 33.3 and 13.6 nM factor Xa/min, respectively. The K1/2 and Vsat values obtained in titrations with factor IXa were not significantly different from those obtained with factor VIII. In titrations with factor X, the values of Michaelis-Menten coefficients (Km) were 31.7 nM with thrombin-activated factor VIII, and 14.2 nM with factor Xa-activated factor VIII. Maximal rates were 23.4 and 4.9 nM factor Xa/min, respectively. The apparent catalytic efficiency was similar with either form of factor VIIIa. Kinetic profiles obtained with platelets as a source of membrane were comparable to those obtained with monocytes. These kinetic profiles are consistent with a 1:1 stoichiometry for the functional interaction between cofactor and enzyme on the surface of monocytes and platelets. Taken together, these results indicate that autocatalytic pathways connecting the extrinsic, intrinsic, and common coagulation pathways can operate efficiently on the monocyte membrane.


Subject(s)
Blood Platelets/metabolism , Factor X/metabolism , Factor Xa/physiology , Monocytes/metabolism , Thrombin/pharmacology , Factor IXa/physiology , Factor VIIIa/physiology , Humans , In Vitro Techniques , Kinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...