Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Thromb Res ; 101(5): 377-85, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11297754

ABSTRACT

Factor VIII (fVIII) is a protein cofactor essential for blood coagulation, and it binds in the factor Xase complex to factors IXa, X, and phospholipid. In about 30% of severe hemophilia A patients, treatment with fVIII leads to production of anti-fVIII antibodies. Anti-fVIII autoantibodies also rarely appear in normal individuals. Those antibodies that inactivate fVIII (inhibitors) prevent optimal fVIII therapy. Inhibitor epitopes were previously localized to the fVIII A2, A3, and C2 domains and to an acidic amino acid region between A1 and A2. Such anti-fVIII antibodies interfere with fVIII binding to components of the factor Xase complex and prevent blood coagulation. When total anti-fVIII titers were determined for each fVIII domain in 43 inhibitor plasmas by immunoprecipitation (IP) and inhibitor neutralization assays, the anti-light chain (LCh) antibody titer was highest, anti-A2 was intermediate, and anti-A1 and anti-B were low. The relative immunogenicity of the fVIII domains in hemophilic and autoantibody inhibitor patients was similar.


Subject(s)
Autoantibodies/blood , Factor VIII/chemistry , Factor VIII/immunology , Hemophilia A/blood , Antibodies, Monoclonal , Blood Coagulation , Factor VIII/antagonists & inhibitors , Hemophilia A/immunology , Humans , Macromolecular Substances , Neutralization Tests , Peptide Fragments/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
2.
Thromb Haemost ; 85(1): 125-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11204564

ABSTRACT

Mice genetically deficient in factor VII (fVIII) are a model of hemophilia A. As a first step to reproduce in this mouse model what occurs over time in hemophilia A patients treated with human fVIII (hfVIII), we have investigated the time course and the characteristics of their immune response to hfVIII, after multiple intravenous injections. Anti-hfVIII antibodies appeared after four to five injections. They were IgG1 and to a lesser extent IgG2, indicating that they were induced by both Th2 and Th1 cells. Inhibitors appeared after six injections. CD4+ enriched splenocytes from hfVIII-treated mice proliferated in response to fVIII and secreted IL-10: in a few mice they secreted also IFN-gamma and in one mouse IL-4, but never IL-2. A hfVIII-specific T cell line derived from hfVIII-treated mice secreted both IL-4 and IFN-gamma, suggesting that it included both Th1 and Th2 cells. CD4+ enriched splenocytes of hfIII-treated mice recognized all hfVIII domains. Thus, hemophilic mice develop an immune response to hfVIII administered intravenously similar to that of hemophilia A patients. Their anti-hfVIII antibodies can be inhibitors and belong to IgG subclasses homologous to those of inhibitors in hemophilic patients; their anti-hfVIII CD4+ cells recognize a complex repertoire and both Th1 and Th2 cytokines, and especially IL-10, may drive the antibody synthesis.


Subject(s)
Factor VIII/immunology , Hemophilia A/blood , Mice, Mutant Strains/immunology , Animals , Antibodies/analysis , Antibodies/blood , Antibody-Producing Cells/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Division/drug effects , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Epitopes/analysis , Factor VIII/administration & dosage , Factor VIII/pharmacology , Hemophilia A/immunology , Humans , Immunoglobulin G/blood , Injections, Intravenous , Interleukin-10/metabolism , Mice , Spleen/cytology , Spleen/immunology , Time Factors
3.
Eur J Immunol ; 30(9): 2548-54, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11009088

ABSTRACT

Using the murine model of hemophilia A, we have examined the role of CD154 in the secondary immune response to factor VIII (FVIII). We previously reported that repeated i.v. injection of FVIII in hemophilia A mice induces a T cell-dependent anti-FVIII antibody formation. Herein, blocking of CD154 by a monoclonal antibody in FVIII-primed hemophilia A mice resulted in the disappearance of pre-existing spleen germinal centers (GC) in the white pulp within 24 h of treatment. Moreover, further expansion of GC in response to FVIII challenge was completely inhibited. In parallel, anti-FVIII antibody titers were markedly reduced and T cell responses to FVIII were abolished. The rapid disappearance of the GC after anti-CD154 treatment was not accompanied by increased B cell apoptosis; instead B cells accumulated in the peripheral zone of the splenic white pulp. Interestingly, repeated exposure to FVIII with anti-CD154 antibody administration blocked anti-FVIII antibody formation but failed to induce long-lasting unresponsiveness. Our data demonstrate that the CD40-CD154 interaction is critical for B cell homeostasis and the secondary immune response to FVIII. For potential clinical application, the data also suggest that therapies targeting the CD154 molecule may be useful for the treatment of high titer FVIII inhibitors in hemophilia A.


Subject(s)
Factor VIII/antagonists & inhibitors , Germinal Center/physiology , Membrane Glycoproteins/physiology , Animals , Antibodies, Monoclonal/immunology , Apoptosis , CD40 Ligand , Female , Hemophilia A/therapy , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
4.
Blood ; 95(4): 1324-9, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10666206

ABSTRACT

Inhibitory antibody formation is a major complication of factor VIII replacement therapy in patients with hemophilia A. To better understand the pathogenesis of this immunologic reaction, we evaluated the role of T-cell costimulatory signals for antifactor VIII antibody formation in a murine model of hemophilia A. Repeated intravenous injections of factor VIII in these factor VIII-deficient mice induced an antifactor VIII inhibitor antibody response. This response was shown to be T-cell dependent by its absence in hemophilic mice also deficient for the T-cell costimulatory ligand B7-2. In separate experiments, injection of murine CTLA4-Ig completely blocked the primary response to factor VIII in hemophilic mice with intact B7 function. This reagent also prevented or diminished further increases in antifactor VIII when given to hemophilic mice with low antifactor VIII antibody titers. These studies suggest that strategies targeting the B7-CD28 pathway are potential therapies to prevent and treat inhibitory antifactor VIII antibodies. Moreover, because the development of antibodies to replaced proteins may limit the success of many human gene therapy approaches, our results may be broadly applicable. (Blood. 2000;95:1324-1329)


Subject(s)
Antigens, Differentiation/therapeutic use , Autoantibodies/blood , Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/prevention & control , Hemophilia A/therapy , Immunoconjugates , Immunosuppressive Agents/therapeutic use , T-Lymphocytes/immunology , Abatacept , Animals , Antibody Formation , Antigens, CD/immunology , B7-2 Antigen , CTLA-4 Antigen , Female , Hemophilia A/immunology , Humans , Lymphocyte Activation , Male , Membrane Glycoproteins/immunology , Mice , Mice, Mutant Strains
5.
Haematologica ; 85(10 Suppl): 100-2, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11187859

ABSTRACT

Inhibitor antibody formation is a major complication of factor VIII replacement therapy in patients with hemophilia A. In order to understand the pathogenesis of this immunologic reaction better, we have characterized the immune response to human factor VIII in a murine model of hemophilia A. Mice with severe factor VIII deficiency caused by targeted gene disruptions were injected intravenously with human factor VIII. A human factor VIII-specific T-cell proliferative response was detected with spleen cells obtained three days after a single injection with human factor VIII and anti-factor VIII antibodies were detected after two intravenous injections. Subsequent exposures led to high titer anti-factor VIII antibodies in both ELISA and inhibitor assays. The anti-factor VIII inhibitor antibody response was shown to be T-cell dependent by its absence in hemophilic mice also deficient for the T-cell co-stimulatory ligand B7-2. In separate experiments, injection of murine CTLA4-Ig completely blocked the primary response to factor VIII in hemophilic mice with intact B7 function. This reagent also prevented or diminished further increases in anti-factor VIII when given to hemophilic mice with low anti-factor VIII antibody titers.


Subject(s)
Factor VIII/immunology , Hemophilia A/immunology , Immunoconjugates , Isoantibodies/blood , Abatacept , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , Antigens, CD , Antigens, Differentiation/immunology , CTLA-4 Antigen , Disease Models, Animal , Factor VIII/administration & dosage , Hemophilia A/complications , Hemophilia A/etiology , Humans , Immunosuppressive Agents/immunology , Mice , Mice, Knockout , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
Thromb Haemost ; 81(2): 240-4, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10063999

ABSTRACT

In order to understand better the mechanism of inhibitor formation in hemophilia A patients, we have characterized the immune response to human factor VIII in a murine model of hemophilia A. Mice with severe factor VIII deficiency caused by targeted gene disruptions in exons 16 and 17 were injected intravenously with human factor VIII. Anti-factor VIII was absent or was detected at only very low levels in hemophilic mice of both strains after a single injection of 0.2 microg factor VIII, but it was present in most mice after a second exposure. Subsequent exposures led to high titer anti-factor VIII antibodies in both ELISA and inhibitor assays. A human factor VIII-specific T cell proliferative response was detected with spleen cells obtained three days after a single injection with human factor VIII, before mice had detectable anti-factor VIII antibodies. Subsequent exposures to factor VIII were followed by an increased T cell proliferative response. These studies indicate that murine hemophilia A is a good model for the study of the immune response to human factor VIII, especially the role of the T cell in the early steps in inhibitor antibody formation.


Subject(s)
Factor VIII/immunology , Hemophilia A/therapy , Isoantibodies/biosynthesis , T-Lymphocytes/immunology , Animals , Antibody Specificity , Drug Administration Schedule , Factor VIII/genetics , Factor VIII/therapeutic use , Female , Hemophilia A/complications , Hemophilia A/immunology , Hemorrhage/etiology , Humans , Isoantibodies/immunology , Lymphocyte Activation , Lymphocyte Cooperation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology
7.
Nat Biotechnol ; 15(10): 971-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9335047

ABSTRACT

Deficiency or abnormality of coagulation factor VIII (FVIII) causes a bleeding disorder called hemophilia A. Treatment involves FVIII concentrates prepared from pooled human plasma or recombinant FVIII (rFVIII) prepared from mammalian cell culture. The cost of highly purified FVIII or rFVIII is a major factor in hemophilia therapy and restricts prophylaxis. We have sought to generate a new source of rFVIII by targeting expression of the human FVIII cDNA to the mammary gland of transgenic pigs using the regulatory sequences of the mouse whey acidic protein gene. The identity of processed heterodimeric rFVIII was confirmed using specific antibodies, by thrombin digestion and activity assays. The secretion of as much as 2.7 micrograms/ml of rFVIII in milk was over tenfold higher than in normal plasma. Up to 0.62 U/ml of rFVIII was detected in an assay in which rFVIII restored normal clotting activity to FVIII-deficient human plasma.


Subject(s)
DNA, Complementary/biosynthesis , Factor VIII/biosynthesis , Mammary Glands, Animal/metabolism , Milk/chemistry , Swine/genetics , Animals , Animals, Genetically Modified , Blood Coagulation/drug effects , Dimerization , Factor VIII/genetics , Factor VIII/pharmacology , Female , Gene Expression Regulation/genetics , Hemophilia A/drug therapy , Hemophilia A/economics , Humans , Mice , Milk Proteins/genetics , Recombinant Proteins/biosynthesis , Thrombin
8.
Am J Hum Genet ; 60(3): 565-73, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9042915

ABSTRACT

Although the molecular defect in patients in a Japanese family with mild to moderately severe hemophilia A was a deletion of a single nucleotide T within an A8TA2 sequence of exon 14 of the factor VIII gene, the severity of the clinical phenotype did not correspond to that expected of a frameshift mutation. A small amount of functional factor VIII protein was detected in the patient's plasma. Analysis of DNA and RNA molecules from normal and affected individuals and in vitro transcription/translation suggested a partial correction of the molecular defect, because of the following: (i) DNA replication/RNA transcription errors resulting in restoration of the reading frame and/or (ii) "ribosomal frameshifting" resulting in the production of normal factor VIII polypeptide and, thus, in a milder than expected hemophilia A. All of these mechanisms probably were promoted by the longer run of adenines, A10 instead of A8TA2, after the delT. Errors in the complex steps of gene expression therefore may partially correct a severe frameshift defect and ameliorate an expected severe phenotype.


Subject(s)
Factor VIII/genetics , Frameshift Mutation , Hemophilia A/genetics , DNA Replication , Exons , Factor VIII/analysis , Female , Frameshifting, Ribosomal , Gene Expression , Humans , Male , Pedigree , Protein Biosynthesis , Reading Frames , Templates, Genetic , Transcription, Genetic
9.
Blood ; 86(5): 1811-9, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7544643

ABSTRACT

The finding that human factor VIII (fVIII) inhibitor antibodies with C2 domain epitopes interfere with the binding of fVIII to phosphatidylserine (PS) suggested that this is the mechanism by which they inactivate fVIII. We constructed a recombinant C2 domain polypeptide and demonstrated that it bound to all six human inhibitors with fVIII light chain specificity. Thus, some antibodies within the polyclonal anti-light chain population require only amino acids within C2 for binding. Recombinant C2 also partially or completely neutralized the inhibitor titer of these plasmas, demonstrating that anti-C2 antibodies inhibit fVIII activity. Immunoblotting of a series of C2 deletion polypeptides, expressed in Escherichia coli, with inhibitor plasmas showed that the epitopes for human inhibitors consist of a common core of amino acid residues 2248 through 2312 with differing extensions for individual inhibitors. The epitope of inhibitory monoclonal antibody (MoAb) ESH8 was localized to residues 2248 through 2285. Three human antibodies and anti-C2 MoAb NMC-VIII/5 bound to a synthetic peptide consisting of amino acids 2303 through 2332, a PS-binding site, but MoAb ESH8 did not. These antibodies also inhibited the binding of fVIII to synthetic phospholipid membranes of PS and phosphatidylcholine, confirming that the blocked epitopes contribute to membrane binding as well as binding to PS. In contrast, MoAb ESH8 did not inhibit binding. As the maximal function of activated fVIII in the intrinsic factor Xase complex requires its binding to a phospholipid membrane, we propose that fVIII inhibition by anti-C2 antibodies is related to the overlap of their epitopes with the PS-binding site. MoAb ESH8 did not inhibit fVIII binding to PS-containing membranes, suggesting the existence of a second mechanism of fVIII inhibition by anti-C2 antibodies.


Subject(s)
Antibodies, Monoclonal , Antibodies , Epitopes/analysis , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Antibodies/pharmacology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Binding, Competitive , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Flow Cytometry , Humans , Immunoblotting , Immunoglobulin G/pharmacology , Kinetics , Molecular Sequence Data , Peptide Fragments/analysis , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/immunology
10.
Curr Opin Hematol ; 2(5): 365-71, 1995 Sep.
Article in English | MEDLINE | ID: mdl-9372021

ABSTRACT

The development of a Factor VIII inhibitor, an antibody that blocks its procoagulant function, is one of the most serious complications of hemophilia A treatment. Similar antibodies are also recognized as a rare cause of bleeding in previously healthy individuals who develop autoimmune anti-Factor VIII antibodies. Recent studies have yielded important information about these antibodies in four different areas: better understanding of the incidence of inhibitors following Factor VIII treatment; identification of patients at highest risk of inhibitor formation; characterization of anti-Factor VIII; and the development of better therapies.


Subject(s)
Factor VIII/antagonists & inhibitors , Hemophilia A/therapy , Antibody Formation , Autoantibodies/blood , Genetic Predisposition to Disease , Hemophilia A/immunology , Humans
11.
Blood ; 86(6): 2183-90, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7662967

ABSTRACT

Porcine factor VIII has been used successfully to treat factor VIII inhibitor patients whose plasmas have minimal cross-reactivity to porcine factor VIII. However, some inhibitor plasmas do inhibit porcine factor VIII, and the extent of procoagulant inhibition often increases after treatment with porcine factor VIII. Because there is no information about the porcine factor VIII epitopes with which these antibodies react, we have compared the immunoblot and enzyme-linked immunosorbent assay (ELISA) reactivities with porcine and human factor VIII for 20 inhibitor plasmas (11 from hemophilia A patients and 9 autoantibodies). Immunoblots identified binding to porcine factor VIII for only 2 of the 12 plasmas from patients who had not received porcine factor VIII, but this reactivity could not be predicted from the inhibitor titer to porcine factor VIII. Immunoblot reactivity with porcine factor VIII was detected for 7 of 8 inhibitor plasmas from patients who had been previously treated with porcine factor VIII, and the strength of this reactivity was generally related to the inhibitor titer. Of the 5 plasmas that were immunoblot positive with the porcine factor VIII A2 domain, 4 had inhibitor titers greater than 45 Bethesda units when tested with porcine factor VIII, whereas only 1 of 15 of the other plasmas had this level of inhibitor activity with porcine factor VIII. In contrast, immunoblot reactivity to the porcine factor VIII A1 domain did not correlate with the antiporcine VIII inhibitor titer. We also determined the effect of preincubation with human or porcine factor VIII on immunoblot reactivity. In one case, immunoblot reactivity with porcine factor VIII was absorbed with porcine, but not human, factor VIII, which is consistent with antibody formation after treatment with porcine factor VIII. In no cases did human factor VIII reduce the reactivity of inhibitor plasmas with the porcine A1 domain, suggesting that these antibodies are directed at unique porcine factor VIII determinants. The reactivity to porcine A2 in 2 plasmas probably represented cross-reactivity of similar A2 determinants, because it was absorbed by both human and porcine factor VIII. Although the ELISA assays with porcine factor VIII detected antibodies in some plasmas that could not be identified by inhibitor assay or immunoblot, the level of ELISA reactivity was generally consistent with the titers of the other assays.


Subject(s)
Autoantibodies/immunology , Factor VIII/immunology , Hemophilia A/immunology , Isoantibodies/immunology , Swine/immunology , Animals , Blotting, Western , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Factor VIII/antagonists & inhibitors , Factor VIII/chemistry , Hemophilia A/blood , Humans , Peptide Fragments/immunology , Recombinant Proteins/immunology , Species Specificity , Thrombin/metabolism
13.
J Biol Chem ; 270(24): 14505-9, 1995 Jun 16.
Article in English | MEDLINE | ID: mdl-7540171

ABSTRACT

The A2 domain (residues 373-740) of human blood coagulation factor VIII (fVIII) contains a major epitope for inhibitory alloantibodies and autoantibodies. We took advantage of the differential reactivity of inhibitory antibodies with human and porcine fVIII and mapped a major determinant of the A2 epitope by using a series of active recombinant hybrid human/porcine fVIII molecules. Hybrids containing a substitution of porcine sequence at segment 410-508, 445-508, or 484-508 of the human A2 domain were not inhibited by a murine monoclonal antibody A2 inhibitory, mAb 413, whereas hybrids containing substitutions at 387-403, 387-444, and 387-468 were inhibited by mAb 413. This indicates that the segment bounded by Arg484 and Ile508 contains a major determinant of the A2 epitope. mAb 413 did not inhibit two more hybrids that contained porcine substitutions at residues 484-488 and 489-508, indicating that amino acid side chains on both sides of the Ser488-Arg489 bond within the Arg484-Ile508 segment contribute to the A2 epitope. The 484-508, 484-488, and 489-508 porcine substitution hybrids displayed decreased inhibition by A2 inhibitors from four patient plasmas, suggesting that there is little variation in the structure of the A2 epitope in the inhibitor population.


Subject(s)
Epitopes/chemistry , Factor VIII/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Base Sequence , DNA Primers , Factor VIII/antagonists & inhibitors , Factor VIII/genetics , Humans , Immunoglobulin Heavy Chains/physiology , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Swine
14.
Adv Exp Med Biol ; 386: 35-45, 1995.
Article in English | MEDLINE | ID: mdl-8851013

ABSTRACT

1. Many factors must be considered when retrospective studies are compared, for the intensity of F.VIII treatment and the frequency of inhibitor evaluation have a marked effect on inhibitor incidence. 2. The incidence of F.VIII inhibitors in patients treated with cryoprecipitate and/or intermediate purity concentrates varies greatly in different studies, with cumulative risks of 7-32% after 10-15 years of follow-up. 3. The rate of inhibitor development appears to be higher for patients treated with recombinant F.VIII, but the cumulative incidence is not greater. This may change, however, when there has been an equivalent follow-up period for the patients receiving recombinant F.VIII. 4. Cumulative risk figures may inappropriately overstate the magnitude of the problem since some inhibitors disappear after short periods of time. 5. Only prospective, long-term studies will provide more satisfactory incidence and prevalence data.


Subject(s)
Factor VIII/antagonists & inhibitors , Hemophilia A/blood , Adolescent , Age Factors , Child , Child, Preschool , Humans , Risk Factors
15.
Nucleic Acids Res ; 22(22): 4851-68, 1994 Nov 11.
Article in English | MEDLINE | ID: mdl-7984443

ABSTRACT

A large number of different mutations in the factor VIII (F8) gene have been identified as a cause of haemophilia A. This compilation lists known single base-pair substitutions, deletions and insertions in the F8 gene and reviews the status of the inversional events which account for a substantial proportion of mutations causing severe haemophilia A.


Subject(s)
Databases, Factual , Factor VIII/genetics , Hemophilia A/genetics , Mutation/genetics , Base Sequence , Chromosome Inversion , Female , Gene Frequency , Genetic Linkage , Humans , Male , Molecular Sequence Data , Point Mutation/genetics , Sequence Deletion/genetics , Sex Factors
16.
Nucleic Acids Res ; 22(17): 3511-33, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7937051

ABSTRACT

A large number of different mutations in the factor VIII (F8) gene have been identified as a cause of haemophilia A. This compilation lists known single base-pair substitutions, deletions and insertions in the F8 gene and reviews the status of the inversional events which account for a substantial proportion of mutations causing severe haemophilia A.


Subject(s)
Databases, Factual , Factor VIII/genetics , Hemophilia A/genetics , Mutation , Base Sequence , Chromosome Inversion , DNA Transposable Elements , Female , Gene Deletion , Gene Rearrangement , Genetic Linkage , Humans , Male , Molecular Sequence Data , Oligodeoxyribonucleotides , Sequence Deletion , Sex Ratio
17.
J Clin Invest ; 93(6): 2497-504, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8200986

ABSTRACT

Human inhibitory alloantibodies and autoantibodies to Factor VIII (FVIII) are usually directed toward the A2 and/or C2 domains of the FVIII molecule. Anti-C2 antibodies block the binding of FVIII to phospholipid, but the mechanism of action of anti-A2 antibodies is not known. We investigated the properties of a patient autoantibody, RC, and a monoclonal antibody, 413, that bind to the region which contains the epitopes of all anti-A2 alloantibodies or autoantibodies studied to date. mAb 413 and RC were noncompetitive inhibitors of a model intrinsic Factor X activation complex (intrinsic FXase) consisting of Factor IXa, activated FVIII (FVIIIa), and synthetic phospholipid vesicles, since they decreased the Vmax of intrinsic FXase by > 95% at saturating concentrations without altering the Km. This indicates that RC and mAb 413 either block the binding of FVIIIa to FIXa or phospholipid or interfere with the catalytic function of fully assembled intrinsic FXase, but they do not inhibit the binding of the substrate Factor X. mAb 413 did not inhibit the increase in fluorescence anisotropy that results from the binding of Factor VIIIa to fluorescein-5-maleimidyl-D-phenylalanyl-prolyl-arginyl-FIXa (Fl-M-FPR-FIXa) on phospholipid vesicles in the absence of Factor X, indicating it does not inhibit assembly of intrinsic FXase. Addition of Factor X to Fl-M-FPR-FIXa, FVIIIa, and phospholipid vesicles produced a further increase in fluorescence anisotropy and a decrease in fluorescence intensity. This effect was blocked completely by mAb 413. We conclude that anti-A2 antibodies inhibit FVIIIa function by blocking the conversion of intrinsic FXase/FX complex to the transition state, rather than by interfering with formation of the ground state Michaelis complex.


Subject(s)
Antibodies, Monoclonal/immunology , Autoantibodies/immunology , Factor VIIIa/antagonists & inhibitors , Animals , Factor VIIIa/metabolism , Factor X/pharmacology , Fluorescence Polarization , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...