Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 5(3): 512-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17181826

ABSTRACT

BACKGROUND: Inhibitory antibodies (Abs) to factor VIII (FVIII inhibitors) constitute the most significant complication in the management of hemophilia A. The analysis of FVIII inhibitors is confounded by polyclonality and the size of FVIII. OBJECTIVES: The goal of this study was to dissect the polyclonal response to human FVIII in hemophilia A mice undergoing a dosage schedule that mimics human use. METHODS: Splenic B-cell hybridomas were obtained following serial i.v. injections of submicrogram doses of FVIII. Results of a novel, anti-FVIII domain-specific enzyme-linked immunosorbent assay were compared to Ab isotype and anti-FVIII inhibitory activity. RESULTS: The robust immune response resulted in the production of approximately 300 hybridomas per spleen. We characterized Abs from 506 hybridomas, representing the most comprehensive analysis of a protein antigen to date. Similar to the human response to FVIII, anti-A2 and anti-C2 Abs constituted the majority of inhibitors. A novel epitope was identified in the A2 domain by competition ELISA. Anti-A2 and anti-C2 Abs were significantly associated with IgG(1) and IgG(2a) isotypes, respectively. Because the IgG(2a) isotype is associated with enhanced Fc receptor-mediated effector mechanisms, this result suggests that anti-C2 Abs and inflammation may be linked. Additionally, we identified a novel class of Abs with dual specificity for the A1 and A3 domains. Forty per cent of the Abs had no detectable inhibitory activity, indicating that they are prominent and potentially pathologically significant. CONCLUSION: The expanded delineation of the humoral response to FVIII may lead to improved management of hemophilia A through mutagenesis of FVIII B-cell epitopes.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Formation , Coagulants/immunology , Factor VIII/immunology , Hemophilia A/immunology , Immunoglobulin Isotypes/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/blood , Antibody Specificity , Binding Sites, Antibody , Binding, Competitive , Cell Line , Coagulants/administration & dosage , Cricetinae , Disease Models, Animal , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay/methods , Epitope Mapping/methods , Factor VIII/administration & dosage , Factor VIII/genetics , Factor VIII/metabolism , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Hybridomas/metabolism , Immunoglobulin Isotypes/biosynthesis , Immunoglobulin Isotypes/blood , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/immunology , Reproducibility of Results , Spleen/cytology , Spleen/metabolism , Swine , Transfection
2.
Arterioscler Thromb Vasc Biol ; 19(10): 2554-62, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10521387

ABSTRACT

A murine antihuman factor IX monoclonal antibody (BC2) has been generated and evaluated for its capacity to prolong the activated partial thromboplastin time (aPTT) in vitro and ex vivo and to prevent arterial thrombosis in a rat model in vivo. BC2 extended aPTT to a maximum of 60 to 80 seconds at 100 to 1000 nmol/L in vitro (rat and human plasma, respectively) and ex vivo (rat) after dosing of rats up to 6 mg/kg in vivo. BC2, administered as bolus (1 to 6 mg/kg) followed by infusion (0.3 to 2 mg x kg(-1) x h(-1)), dose-dependently prevented thrombosis of an injured rat carotid artery (FeCl(3)-patch model), increased time to artery occlusion, and reduced incidence of vessel occlusion. BC2 efficacy in preventing arterial thrombosis exceeded that of heparin (bolus 15 to 120 U/kg followed by infusion 0.5 to 4.0 U x kg(-1) x min(-1)), whereas the latter rendered the blood incoagulable (aPTT>1000 seconds). BC2 demonstrated complete antithrombotic efficacy also as a single bolus given either as prevessel or postvessel injury as evidenced by reduction of thrombus mass (from 4.18+/-0.49 to 1.80 +/-0.3 mg, P<0.001), increasing vessel patency time (from 14.9+/-0.9 minutes to 58.3+/-1.7 minutes, P<0.001) and decreasing incidence of vessel occlusion from 100% to 0% in vehicle- versus BC2-treated rats, respectively. BC2 (3 mg/kg, IV) administered in a single bolus resulted in 50% reduction in thrombus mass (P<0.01), extended vessel patency time (P<0.001), extended aPTT only 4-fold, and had no effect on blood loss via a tail surgical wound; heparin, at doses that reduced thrombus mass to a similar extent, extended aPTT beyond 1000 seconds (over 500-fold) and increased blood loss from 1.8+/-0.7 to 3.3 +/-0.6 mL (P<0.001). These data suggest that BC2 may provide enhanced therapeutic efficacy in humans at lesser interference with blood hemostasis than heparin.


Subject(s)
Antibodies, Monoclonal/pharmacology , Factor IX/immunology , Thrombosis/immunology , Anesthesia , Animals , Anticoagulants/pharmacology , Arterial Occlusive Diseases/chemically induced , Arterial Occlusive Diseases/drug therapy , Arterial Occlusive Diseases/immunology , Aspirin/pharmacology , Blood Loss, Surgical , Carotid Arteries , Disease Models, Animal , Dose-Response Relationship, Immunologic , Factor IX/metabolism , Fibrinolytic Agents/pharmacology , Heparin/pharmacology , Humans , Immunoglobulin G/pharmacology , Iron , Male , Microscopy, Electron, Scanning , Partial Thromboplastin Time , Prothrombin Time , Rats , Rats, Sprague-Dawley , Thrombosis/chemically induced , Thrombosis/drug therapy
3.
Blood Coagul Fibrinolysis ; 7(1): 5-14, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8845463

ABSTRACT

A unique blood coagulation factor X variant has been identified in a family with a history of bleeding. Plasma from affected family members had prolonged prothrombin times and activated partial thromboplastin times, low to below normal factor X coagulant activity, and normal factor X antigen levels. Sequencing of DNA from the propositus revealed a single G to A substitution in one allele of factor X at base 964 resulting in an amino acid substitution of Asn for Asp at residue 282. This residue corresponds with the active site Asp102 of chymotrypsin. The substitution eliminates a TaqI restriction site and provided the basis for a screening assay to detect the mutation in polymerase chain reaction (PCR) amplified factor X exon VIII DNA. Fourteen additional family members were identified as having the mutation at base 964. Plasma factor X purified from the proposita using an anti-factor X monoclonal antibody immunoadsorbent exhibited an approximately 50% decrease in specific activity compared with factor X purified from a normal individual in a similar manner. Bleeding in family members with the mutation, termed factor X Stockton, appears to be due to disruption of normal hemostasis by the presence in plasma of circulating abnormal factor X. Factor X Stockton is the first naturally occurring substitution at the active site Asp of a serine protease and underscores the importance of this amino acid residue in factor Xa coagulant activity.


Subject(s)
Factor X/genetics , Hemorrhagic Disorders/genetics , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , Female , Humans , Molecular Sequence Data , Mutation , Partial Thromboplastin Time , Pedigree
4.
J Biol Chem ; 269(46): 28606-12, 1994 Nov 18.
Article in English | MEDLINE | ID: mdl-7961808

ABSTRACT

Prior studies using the mutant thrombin, thrombin Quick I, indicate that this protease induces maximum platelet aggregation and intraplatelet [Ca2+] fluxes at agonist concentrations where dissociable, equilibrium platelet binding is undetectable and led to the conclusion that thrombin interaction with its platelet "receptor" is best described kinetically by formation of an enzyme-substrate complex. This conclusion was substantiated further in the present studies by demonstrating that both thrombin Quick I and thrombin mimicked the thrombin receptor agonist peptide in the induction of the platelet activation-dependent events required for functional Prothrombinase assembly and that a rabbit antibody raised against KATNATLDPRSFLLR, a pentadecapeptide which represents amino acids 32-46 in the platelet thrombin receptor/substrate and spans the thrombin cleavage site, inhibited both thrombin- and thrombin Quick I-induced platelet activation responses equivalently. The antipeptide antibody had a more pronounced inhibitory effect on the rate of the thrombin-induced response rather than the magnitude of the response suggesting competition for the cleavage site, consistent with the observation that pretreatment of metabolically-inhibited platelets with thrombin, which was removed by washing, eliminated specific antibody binding due to removal and/or masking of antibody epitopes. Concentrations of the antipeptide antibody that inhibited thrombin- and thrombin Quick I-induced increases in intracellular [Ca2+] flux by as much as 97% did not alter the dissociable equilibrium binding of 125-I-FPR-thrombin to platelets. These combined data indicate that the hydrolytic event initiated by thrombin or thrombin Quick I interaction with the platelet receptor/substrate for thrombin is unrelated to the dissociable equilibrium binding of thrombin to membrane sites described previously by classical receptor-ligand binding analyses.


Subject(s)
Platelet Activation/physiology , Thrombin/physiology , Amino Acid Sequence , Blood Platelets/enzymology , Humans , Molecular Sequence Data , Peptide Fragments/metabolism , Receptors, Thrombin/agonists , Receptors, Thrombin/metabolism , Substrate Specificity , Thrombin/metabolism , Thrombin/pharmacology , Thromboplastin/metabolism
5.
Hybridoma ; 13(5): 423-9, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7860098

ABSTRACT

Kringle domains are found in several plasma proteins of blood coagulation and fibrinolysis. A murine monoclonal antibody, designated alpha HII-5, was produced against a synthetic peptide representing residues 216-231 of human prothrombin kringle 2. The sequence of the hexadecapeptide (Glu-Asn-Phe-Cys-Arg-Asn-Pro-Asp-Gly-Asp-Glu-Glu-Gly-Val-Gly-Cys) is conserved in several kringle-containing proteins, represents a predicted region of high local hydrophilicity in prothrombin kringle 2, and contains the anionic (Asp-223 and Asp-225) residues that contribute to lysine binding by plasminogen kringle 4. In a solution-phase immunoassay, antibody alpha HII-5 bound prothrombin and the kringle 5 light chain fragment of plasminogen (miniplasminogen), but not plasminogen or plasmin. In contrast, using a solid-phase assay with antigen immobilized onto a surface (polystyrene microtiter plates, glass, or nitrocellulose) antibody alpha HII-5 specifically bound prothrombin, plasminogen, recombinant tissue plasminogen activator (tPA), and the apo(a) subunit of lipoprotein(a). By immunoblotting analysis antibody alpha HII-5 bound determinants on prothrombin fragment 2 and plasminogen kringle 5. These observations suggest that a subset of kringle domains on plasma proteins, including prothrombin kringle 2 and plasminogen kringle 5, contains a homologous antigenic determinant in the region of the kringle lysine-binding site. In contrast to prothrombin kringle 2, the homologous peptide site on plasminogen is not available for antibody binding except when plasminogen is adsorbed to a nonphysiological surface, or when kringles 1-4 are removed.


Subject(s)
Antibodies, Monoclonal/immunology , Kringles/immunology , Prothrombin/immunology , Amino Acid Sequence , Animals , Immunoassay/methods , Immunoblotting , Mice , Molecular Sequence Data , Plasminogen/immunology , Sequence Homology, Amino Acid
6.
Thromb Haemost ; 72(1): 21-7, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7974370

ABSTRACT

We determined factor VII antigen (FVIIag) levels in 705 healthy blood donors ranging in age from 17 to 79 years using a two-site solid-phase enzyme immunoassay developed in our laboratory. The mean (+/- SD) FVIIag level for the total population was 102 +/- 31%. FVIIag levels for men (n = 375) and women (n = 330) were 101 +/- 28% and 103 +/- 33%, respectively. A significant increase in FVIIag was observed with age in both men (r = 0.25, p < 0.0001) and women (r = 0.35, p < 0.0001). FVIIag levels were significantly higher in women > 60 years when compared to men (median women: 125%; median men: 111%; p < 0.05). On a subset of the study group (n = 45), FVIIag levels were correlated to total cholesterol (r = 0.27, p = 0.08) and triglyceride (r = 0.41, p < 0.01). Assuming the commonly used reference interval of 60-140% for FVII, the frequencies of FVIIag values for < 60% and > 140% using our assay were 2.1% and 9.2%, respectively. Gender and age-related differences in FVIIag levels must be considered in a reference interval. We further suggest that assay-specific reference ranges be established, which may include values outside the commonly used values of 60-140%.


Subject(s)
Antigens/blood , Blood Donors , Factor VII/immunology , Adolescent , Adult , Age Distribution , Aged , Female , Humans , Immunoenzyme Techniques , Lipids/blood , Male , Middle Aged , Reference Values , Reproducibility of Results , Sex Distribution
8.
Blood Coagul Fibrinolysis ; 3(5): 563-74, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1450323

ABSTRACT

Factor X, a vitamin K-dependent protein, is the plasma zymogen for the active serine protease factor Xa. Factor Xa is the proteolytic enzyme for prothrombinase, the multi-protein membrane complex that catalyses the cleavage of prothrombin to thrombin. A panel of 10 monoclonal antibodies (identified by their corresponding clone numbers: 1, 2, 3, 5, 7, 26, 27, 54, 73, and 79) to factor X were produced by immunizing mice with purified factor X. All of the antibodies bound both human factor X and factor Xa in a solid-phase ELISA and binding of the antibodies was not affected by removal of Ca2+ with EDTA. In immunoblot analysis, antibody alpha HFX-54 bound to the light chain and antibodies alpha HFX-1, -5, -7, and -26 bound to the heavy chain of reduced factor X. Antibodies alpha BFX-2b, alpha HFX-27, -54, and -73 prolonged both the factor X-dependent clotting time and activated partial thromboplastin time (APTT) of normal plasma while antibody alpha HFX-1 only prolonged the APTT. None of the antibodies significantly inhibited factor X activation by purified Russell's viper venom factor X activator. In prothrombin activation assays using purified factor Xa, factor Va, prothrombin, Ca2+ and phospholipid vesicles, seven of the antibodies (alpha HFX-1, -3, -26, -27, -54, -73 and alpha BFX2b) showed some inhibition of thrombin generation ranging from 18 to 60% of the control. The decrease in factor X plasma clotting activity was most likely due to inhibition of factor Xa activity in prothrombinase, although some antibody-dependent inhibition of factor X activation may contribute to the observed inhibition of plasma clotting. Prothrombinase activity on platelets was inhibited in an identical manner by the monoclonal antibodies. When prothrombin was activated in the absence of factor Va, only antibody alpha BFX-2b inhibited activation. Calcium-independent determinants on both the heavy chain (determinants 1 and 26) and light chain (determinant 54) of factor X may play a role in prothrombin activation by prothrombinase. Other epitopes (antibodies alpha HFX-3, -27, -73) appeared to be influenced by association of factor Xa with factor Va. Topographic regions on factor X important for factor X activation and factor Xa function may be identified by the use of these monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Factor X/immunology , Binding Sites, Antibody , Blood Coagulation , Factor X/chemistry , Humans , Neutralization Tests
9.
Hybridoma ; 10(6): 659-72, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1726159

ABSTRACT

Three murine monoclonal antibodies (designated alpha Pg-28, alpha Pg-96, and alpha Pg-247) against human plasminogen were prepared. All three antibodies bound plasminogen and the elastase-digestion product of plasminogen consisting of residues Val442-Asn790 (miniplasminogen). The epitopes recognized by each antibody were distinct. Antibodies alpha Pg-96 and alpha Pg-247 blocked tissue plasminogen activator-dependent lysis in a fibrin plate assay while antibody alpha Pg-28 had no effect. Antibody alpha Pg-96 blocked urokinase- and tissue plasminogen activator-catalyzed, and streptokinase-mediated, plasminogen activation. Antibodies alpha Pg-28 and alpha Pg-247 partially inhibited tissue plasminogen activator-catalyzed plasminogen activation. Antibodies alpha Pg-28 and alpha Pg-247 also inhibited streptokinase-mediated plasminogen activation, but not urokinase-catalyzed activation. Antibody alpha Pg-247 inhibited plasmin catalysis of substrate S-2251 by decreasing the VMAX and increasing the KM of plasmin for the synthetic substrate S-2251 four-fold. The other antibodies had no significant effect on plasmin activity. This differential inhibition of plasminogen activation suggests that activation by urokinase, tissue plasminogen activator, and streptokinase possibly involve distinct regions of miniplasminogen structure.


Subject(s)
Antibodies, Monoclonal/immunology , Peptide Fragments/immunology , Plasminogen/antagonists & inhibitors , Animals , Antibody Specificity , Enzyme Activation , Epitopes/immunology , Humans , Kinetics , Mice , Plasminogen/immunology , Plasminogen/metabolism , Plasminogen Activators/metabolism
10.
J Biol Chem ; 266(28): 18435-8, 1991 Oct 05.
Article in English | MEDLINE | ID: mdl-1655750

ABSTRACT

Using three experimental approaches, we have addressed the questions of whether the presence of saturably bound thrombin plays a role in potentiating the activation of platelet phospholipase C (PLC) and/or accumulation of the 3-phosphorylated phosphoinositides (3-PPI), i.e. phosphatidylinositol 3,4-bisphosphate and phosphatidylinositol 3,4,5-trisphosphate, and whether the generation of tethered ligand (Vu, T-K.H., Hung, D. T., Wheaton, V. I., and Coughlin, S. R. (1991) Cell 64, 1057-1068) by thrombin can account fully for thrombin's proteolytic effects in activating platelets, as gauged by the above parameters. We have 1) measured PLC activation or 3-PPI after we have exposed platelets to thrombin for various periods and either blocked thrombin's proteolytic activity without interrupting its binding or blocked both binding and proteolytic activity of thrombin; 2) attempted to potentiate 3-PPI accumulation, using combinations of protein kinase C stimulation, Ca2+ elevation, and saturating but proteolytically inactive thrombins; and 3) compared the activation of platelets by thrombin with activation by the "thrombin" receptor-directed peptide, SFLLRNPNDKYEPF (SFLL; a portion of the tethered ligand created by thrombin's proteolytic activity), and examined the effect of thrombin on this latter activation. We conclude that the initial and sustained effects of thrombin in stimulating PLC and the accumulation of 3-PPI are completely attributable to thrombin's proteolytic activity. Further, thrombin's effects in promoting these responses can be accounted for by the actions of SFLL peptide, and by implication, formation of tethered ligand.


Subject(s)
Blood Platelets/enzymology , Phosphatidylinositols/metabolism , Receptors, Cell Surface/metabolism , Thrombin/metabolism , Type C Phospholipases/metabolism , Amino Acid Sequence , Arginine/analogs & derivatives , Arginine/pharmacology , Blood Platelets/drug effects , Dansyl Compounds/pharmacology , Enzyme Activation , Hirudins/pharmacology , Humans , Kinetics , Ligands , Molecular Sequence Data , Peptide Fragments/pharmacology , Phosphorylation , Receptors, Thrombin
11.
Mol Cell Biol ; 11(7): 3537-44, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1904541

ABSTRACT

The Saccharomyces cerevisiae CDC42 gene product, a member of the ras superfamily of low-molecular-weight GTP-binding proteins, is involved in the control of cell polarity. We have analyzed the effects of three CDC42 mutations (Gly to Val-12, Gln to Leu-61, and Asp to Ala-118) in the putative GTP-binding and hydrolysis domains and one mutation (Cys to Ser-188) in the putative isoprenylation site. The first three mutations resulted in either a dominant-lethal or dose-dependent dominant-lethal phenotype when present on plasmids in haploid cdc42-1ts or wild-type strains. Both wild-type and cdc42-1ts cells carrying plasmids (pGAL) with either the CDC42Val-12 or CDC42Leu-61 alleles under the control of a GAL promoter were arrested with a novel phenotype of large cells with elongated or multiple buds. Cells carrying pGAL-CDC42Ala-118 were arrested as large, round, unbudded cells reminiscent of cdc42-1ts arrested cells. The different phenotype of the CDC42Ala-118 mutant versus the CDC42Val-12 and CDC42Leu-61 mutants was unexpected since the phenotypes of all three analogous ras mutants were similar to each other. This suggests that aspects of the biochemical properties of the Cdc42 protein differ from those of the Ras protein. The cdc42Ser-188 mutant gene was incapable of complementing the cdc42-1ts mutation and was recessive to both wild-type and cdc42-1ts. In double-mutant alleles, the cdc42Ser-188 mutation was capable of suppressing the dominant lethality associated with the three putative GTP-binding and hydrolysis mutations, suggesting that isoprenylation is necessary for the activity of the wild-type and mutant proteins.


Subject(s)
Fungal Proteins/genetics , GTP-Binding Proteins/genetics , Genes, Fungal , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Base Sequence , GTP Phosphohydrolases/genetics , Genes, Dominant , Genes, Lethal , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Peptides/chemical synthesis , Phenotype , Plasmids , Promoter Regions, Genetic , Saccharomyces cerevisiae/physiology , cdc42 GTP-Binding Protein, Saccharomyces cerevisiae
12.
J Biol Chem ; 266(21): 13726-30, 1991 Jul 25.
Article in English | MEDLINE | ID: mdl-1856206

ABSTRACT

Activation of vitamin K-dependent plasma proteases occurs by specific interaction with components of the blood coagulation cascade. In this report, we describe the direct expression and enzymatic characterization of the human coagulation zymogen factor X and its activated form, factor Xa, from transformed Chinese hamster ovary fibroblast cell lines. Expression was achieved using either a full-length factor X cDNA or a unique mutant factor Xa cDNA. The functional factor Xa precursor contained a novel tripeptide bridge in place of the native 52-amino acid activation peptide. This mutation allowed for intracellular processing and secretion of the activated form of factor X. Secreted recombinant factors X (rX) and Xa (rXa) were purified by sequential anion-exchange and immunoaffinity chromatography. The enzymatic activities of factors rX and rXa were compared with those of plasma factors X and Xa in three independent assay systems. In comparison to human plasma factor X, the amidolytic, prothrombinase complex, and plasma clotting activities of factor rX were 50, 85, and 43%, respectively. The corresponding comparative activities for factor rXa were 32, 64, and 48%, respectively. The ability to directly express mutant forms of biologically active human factor X will facilitate the structure/function analysis of this important blood coagulation protein and may lead to the development of novel coagulation inhibitors.


Subject(s)
Factor X/genetics , Factor Xa/genetics , Animals , Base Sequence , Cell Line , Cricetinae , Cricetulus , DNA Mutational Analysis , Enzyme Activation , Enzyme Precursors/genetics , Factor X/metabolism , Factor Xa/metabolism , Humans , Molecular Sequence Data , Oligonucleotides/chemistry , Recombinant Proteins
13.
J Biol Chem ; 266(13): 8384-91, 1991 May 05.
Article in English | MEDLINE | ID: mdl-2022654

ABSTRACT

Prothrombin contains two kringle domains that are removed during activation to the blood clotting enzyme alpha-thrombin. By analogy with other kringle-containing proteins the prothrombin kringles may play a role in the protein-protein interactions necessary for prothrombin activation. Four monoclonal antibodies to prothrombin kringle 2 have been produced against human prothrombin, and a fifth monoclonal antibody was produced against a synthetic peptide consisting of amino acid residues 216-231 of kringle 2. Each antibody was tested for its ability to block prothrombin activation by factor Xa. In the presence of phosphatidylcholine/phosphatidylserine vesicles and factor Va, two of the antibodies, alpha HII-3 and alpha HII-4, inhibited prothrombin activation at a 90 and 50% level, respectively. Two other monoclonal antibodies (alpha HII-6 and alpha HII-7) and the antipeptide antibody (alpha HII-5) had no effect on prothrombin activation. When factor Xa was the catalyst alone, antibody alpha HII-3 lost the ability to inhibit prothrombin activation whereas antibody alpha HII-4 again partially inhibited the reaction. When human platelets were the reaction surface, the patterns of inhibition by the anti-fragment 2 antibodies were identical to that observed with phospholipid vesicles. These data suggest a role for prothrombin fragment 2 in activation, possibly by mediating the interaction of substrate prothrombin with factor Xa or factor Va on the phospholipid surface.


Subject(s)
Antibodies, Monoclonal/immunology , Blood Platelets/metabolism , Phospholipids/metabolism , Prothrombin/metabolism , Amino Acid Sequence , Animals , Enzyme Activation , Factor Va/metabolism , Factor Xa/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Prothrombin/immunology
14.
Gene ; 99(2): 291-4, 1991 Mar 15.
Article in English | MEDLINE | ID: mdl-1902434

ABSTRACT

A 1.5-kb cDNA (FX) encoding full-length human coagulation factor X was isolated from a human fetal liver cDNA library. The identity of the insert in a selected phage lambda clone was confirmed to be FX by nucleotide (nt) sequence analysis and restriction mapping. This FX cDNA clone contained 1467 bp of coding sequence, no 5'-untranslated sequence, a short 3'-untranslated sequence of 10 nt and a poly(A) tail at the 3'-end. The FX cDNA was inserted into a mammalian expression vector and transfected into COS-1 monkey kidney cells. Media from transfected cells showed evidence of factor X antigen and, following addition of Russel's viper venom factor X activator, enhanced amidolytic activity toward a synthetic peptide rho-nitroanilide substrate. Immunoprecipitation with an anti-factor X monoclonal antibody of [35S]methionine-labeled cell-conditioned media showed evidence of polypeptides of 74, 55, and 17 kDa, as determined by SDS-PAGE followed by autoradiography. Together, these results indicate that an active factor X can be successfully expressed in a recombinant DNA expression system. This approach will allow the systematic structure/function investigation of this important blood-clotting enzyme.


Subject(s)
DNA, Recombinant/analysis , Factor X/genetics , Haplorhini/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Factor X/biosynthesis , Gene Expression/drug effects , Humans , In Vitro Techniques , Kidney/metabolism , Molecular Sequence Data , Radioimmunoassay , Transfection , Vitamin K/pharmacology
15.
Blood ; 76(1): 1-16, 1990 Jul 01.
Article in English | MEDLINE | ID: mdl-2194585

ABSTRACT

During the past 20 years contributions from many laboratories have led to the development of isolation procedures, delineation of primary structures, and more recently, to the expression of recombinant proteins associated with the coagulation cascade. In general, studies of coagulation proteins under defined conditions have demonstrated the prescience of Davie and Ratnoff and MacFarlane in their proposals of the coagulation cascade. The more recent discovery of thrombomodulin by Esmon et al has led to the identification and characterization of components of the vitamin K-dependent anticoagulant pathway. In this review we have attempted to analyze and compare the functional properties of each of the vitamin K-dependent enzyme complexes associated with the procoagulant and anticoagulant phases of blood clotting. Although dissimilarities exist, the vitamin K-dependent complexes have analogous requirements and appear to function with a common general mode of organization. Membrane-bound cofactors serve as anchoring sites for the appropriate membrane-binding enzymes. This process localizes the complex on the membrane surface and increases the catalytic efficiency for substrate utilization. Complex formation provides extraordinary improvements in the catalytic efficiency for the complexes as compared with their soluble enzyme components. Membrane-bound complexes provide a mechanism that can be regulated at a site by membrane presentation, zymogen activation, and cofactor activation or presentation. The kinetic constants obtained for the various coagulation reactions determined in vitro provide some insights into how these pathways may function in vivo. The catalytic efficiency (kcat/Km) for factor X activation by factor VIIIa/factor IXa is far in excess of the catalytic efficiency of activation of factor X by tissue factor/factor VIIa (Table 3). This may provide a rational interpretation for the observation that patients with hemophilia A and B bleed even though they appear to have an alternative pathway to factor X activation. In addition, tissue factor is not ordinarily presented by the vascular tissue that has direct access to blood. However, it appears that extravascular constitutive tissue factor is available once the blood vessel becomes disrupted. The efforts to identify the initiating reactions of the blood coagulation process have not been unambiguously successful. We conclude that factor VII is most likely a zymogen, just as are the other proenzymes of the blood clotting process. In addition, it is difficult to rationalize the importance of the intrinsic pathway of coagulation involving factor XII, prekallikrein, and high molecular weight kininogen since the congenital absence of any one of these factors does not result in abnormal bleeding.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Blood Coagulation/physiology , Multienzyme Complexes/physiology , Vitamin K/physiology , Enzyme Precursors/metabolism , Enzyme Precursors/physiology , Humans , Multienzyme Complexes/metabolism , Serine Endopeptidases/metabolism , Serine Endopeptidases/physiology , Vitamin K/metabolism
16.
J Biol Chem ; 265(15): 8863-8, 1990 May 25.
Article in English | MEDLINE | ID: mdl-2341407

ABSTRACT

Coagulation factor V (fV) is a single-chain glycoprotein (Mr 330,000; domain structure A1-A2-B-A3-C1-C2) that is activated to factor Va (fVa; Mr 174,000) by thrombin, which cleaves away the B domain leaving a heterodimeric structure composed of a heavy chain (A1-A2; Mr 94,000) and a light chain (A3-C1-C2; Mr 74,000). We analyzed the ultrastructure of scanning transmission electron microscope images of bovine and human fV, bovine fVa, and its constituent light chains and heavy chains. Factor V molecules had irregularly globular (10-12 nm) to oblong (8-14 nm) core structures which commonly displayed a peripheral satellite appendage of variable morphology attached to the core by a narrow stalk. Scanning transmission electron microscope mass analyses indicated that monomolecular bovine fV molecules had a mass of 322 +/- 45 kDa and human fV, 315 +/- 31 kDa. Factor Va molecules were irregular, globular (8-12 nm) structures that resembled the fV core structure, lacked the satellite appendage representing B domainal structures, and had a mass of 180 +/- 22 kDa. Our findings permit us to propose a structural model of fV suggesting the relative orientation of its closely associated light chain and heavy chain core components and indicating that these constituents remain associated in the transition from fV to fVa.


Subject(s)
Factor V/ultrastructure , Factor Va/ultrastructure , Animals , Cattle , Humans , Macromolecular Substances , Microscopy, Electron , Microscopy, Electron, Scanning , Models, Molecular , Molecular Weight , Protein Conformation
17.
Biochem J ; 266(2): 393-8, 1990 Mar 01.
Article in English | MEDLINE | ID: mdl-2180399

ABSTRACT

A peptide corresponding to a surface loop in the C-terminal domain of chicken ovotransferrin (residues 570-584) was made by solid-phase synthesis and used to immunize rabbits. A 15-amino acid-residue disulphide-linked loop occurs in both domains of all five transferrins for which the sequence is available and lies on the opposite side of the iron-binding site from the interdomain cleft. Polyclonal antibodies to the peptide were specific for non-reduced holo-ovotransferrin and the C-terminal domain, as shown by e.l.i.s.a. and immunoblotting. The antibody did not inhibit binding of ovotransferrin to receptors on chick-embryo reticulocytes but was able to bind ovotransferrin bound to the cellular receptors at 0 degree C. The loop composed of residues 570-584 appears to be remote from the transferrin receptor-binding site.


Subject(s)
Receptors, Transferrin/metabolism , Transferrin/ultrastructure , Animals , Binding Sites , Blotting, Western , Chick Embryo , Immunologic Techniques , Peptide Fragments/immunology , Reticulocytes/metabolism , Transferrin/metabolism
18.
Blood ; 74(7): 2418-25, 1989 Nov 15.
Article in English | MEDLINE | ID: mdl-2804372

ABSTRACT

Vitamin K deficiency or administration of vitamin K antagonists results in the biosynthesis of abnormal des-gamma-carboxy forms of the vitamin K-dependent proteins. Monoclonal antibody H-11 binds several vitamin K-dependent proteins at a determinant that includes the first two residues of gamma-carboxyglutamic acid. Antibody H-11 binds fully carboxylated prothrombin and protein C in the presence of EDTA but binding is inhibited by the divalent metal ions, calcium, magnesium, and manganese. By contrast, des-gamma-carboxy prothrombin and protein C bind antibody H-11 the same in the presence of EDTA or calcium ion. Antibody H-11 thus appears to bind a conserved antigenic site containing gamma-carboxyglutamic acid that in the presence of divalent metal ion undergoes a conformational transition. This ability of antibody H-11 to bind des-gamma-carboxy prothrombin and protein C in the presence of calcium ion allowed the development of an immunoassay for these proteins in plasma. Prothrombin and protein C from stably anticoagulated individuals receiving warfarin were characterized by their ability to bind antibody H-11 in the presence of calcium ion. Binding of prothrombin and protein C to antibody H-11 in the presence of calcium correlated temporally with warfarin administration. The inability of calcium ion to inhibit binding of antibody H-11 to abnormal prothrombin and protein C in plasma suggests that the circulating forms of both proteins following warfarin administration cannot undergo the metal ion-dependent conformational transition that includes sequence residues 1 through 12.


Subject(s)
Antibodies, Monoclonal/immunology , Blood Coagulation Factors/immunology , Blood Proteins/immunology , Protein C/immunology , Prothrombin/immunology , Cations, Divalent/pharmacology , Glutamates/immunology , Humans , In Vitro Techniques , Vitamin K , Warfarin/therapeutic use
19.
J Biol Chem ; 264(30): 17882-7, 1989 Oct 25.
Article in English | MEDLINE | ID: mdl-2478538

ABSTRACT

The murine monoclonal antibody H-11 binds a conserved epitope found at the amino terminal of the vitamin K-dependent blood proteins prothrombin, factors VII and X, and protein C. The sequence of polypeptide recognized by antibody H-11 contains 2 residues of gamma-carboxyglutamic acid, and binding of the antibody is inhibited by divalent metal ions. By using a solid-phase immunoassay with 125I-labeled antibody and immobilized vitamin K-dependent protein, binding of the antibody to the vitamin K-dependent proteins was inhibited by increasing concentrations of calcium, manganese, and magnesium ion. The transition midpoints for antibody binding were in the millimolar concentration range and were different for each metal ion. In general, the transition midpoints were lowest for manganese ion, intermediate for calcium ion, and highest for magnesium ion. Antibody H-11 bound specifically to a synthetic peptide corresponding to residues 1-12 of human prothrombin that was synthesized as the gamma-carboxyglutamic acid-containing derivative. Binding of the antibody to the peptide was not inhibited by calcium ion. These data suggest that inhibition of antibody H-11 binding by divalent metal ions is not due simply to neutralization of negative charge by Ca2+. This transition which is conserved in vitamin K-dependent proteins containing the H-11 antigenic site is likely due to a structural transition of the amino-terminal polypeptide possibly from a random (accessible) to ordered (inaccessible) structure.


Subject(s)
1-Carboxyglutamic Acid , Antigen-Antibody Complex , Epitopes/analysis , Factor VII/immunology , Factor X/immunology , Protein C/immunology , Prothrombin/immunology , Vitamin K/physiology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Calcium , Cations, Divalent , Cattle , Humans , Molecular Sequence Data , Oligopeptides/chemical synthesis
20.
Blood ; 72(6): 1911-21, 1988 Dec.
Article in English | MEDLINE | ID: mdl-2461754

ABSTRACT

A monoclonal antibody (designated alpha BFX-2b) prepared against bovine factor X inhibited factor X activity in human, bovine, porcine, rabbit, and canine plasma. In assays using purified prothrombinase components, factor Xa, factor Va, phospholipid vesicles, and calcium ion with the fluorescent active site thrombin inhibitor dansylarginyl-N-(3-ethyl-1,5-pentanediyl)amide, the antibody inhibited the conversion of prothrombin to thrombin. Antibody alpha BFX-2b also blocked prothrombinase cleavage of the macromolecular substrates prethrombin 1 and prethrombin 2 but did not inhibit factor Xa hydrolysis of the synthetic substrate benzoyl-Ile-Glu-Gly-Arg-p-nitroanilide. The antibody also prevented the inactivation of factor Xa by antithrombin III but did not prevent the inactivation by soybean trypsin inhibitor. Antibody alpha BFX-2b bound factor Xa with a stoichiometry of 1:1 and an apparent dissociation constant of 9.0 x 10(-11) mol/L as estimated from its inhibition of prothrombinase activity. Antibody alpha BFX-2b did not prevent binding of factor Xa to factor Va-phospholipid as measured by using fluorescence polarization or high-pressure liquid gel chromatography with the fluorescent Factor Xa analogue dansyl-glutamyl-glycyl-arginyl-Xa. Immunoblotting of factor X following electrophoresis on sodium dodecyl sulphate-polyacrylamide gels and transfer to nitrocellulose indicated that the antigenic determinant recognized by antibody alpha BFX-2b was found on the heavy chain of factors X and Xa. From these observations it can be concluded that antibody alpha BFX-2b recognizes a highly conserved epitope on the factor X heavy chain that is remote from the topographic sites required for prothrombinase complex assembly and substrate hydrolysis but may be located at or near a portion of the macromolecular substrate binding site.


Subject(s)
Antibodies, Monoclonal/immunology , Factor X/immunology , Prothrombin/metabolism , Thromboplastin/metabolism , Animals , Antithrombin III/metabolism , Cattle , Dogs , Enzyme Precursors/metabolism , Epitopes/immunology , Factor X/antagonists & inhibitors , Factor Xa , Oligopeptides/metabolism , Protein Conformation , Rabbits , Serine Proteinase Inhibitors , Species Specificity , Swine , Thromboplastin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...