Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Haemophilia ; 17(5): 721-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21649795

RESUMO

The possibility of alloimmunization in patients receiving protein replacement therapy depends on (at least) three risk factors, which are necessary concomitantly but insufficient alone. The first is the degree of structural difference between the therapeutic protein and the patient's own endogenous protein, if expressed. Such differences depend on the nature of the disease mutation and the pre-mutation endogenous protein structure as well as on post-translational changes and sequence-engineered alterations in the therapeutic protein. Genetic variations in the recipients' immune systems comprise the second set of risk determinants for deleterious immune responses. For example, the limited repertoire of MHC class II isomers encoded by a given person's collection of HLA genes may or may not be able to present a 'foreign' peptide(s) produced from the therapeutic protein - following its internalization and proteolytic processing - on the surface of their antigen-presenting cells (APCs). The third (and least characterized) variable is the presence or absence of immunologic 'danger signals' during the display of foreign-peptide/MHC-complexes on APCs. A choice between existing therapeutic products or the manufacture of new proteins, which may be less immunogenic in some patients or patient populations, may require prior definition of the first two of these variables. This leads then to the possibility of developing personalized therapies for disorders due to genetic deficiencies in endogenous proteins, such as haemophilia A and B. [Correction made after online publication 11 July 2011: several critical corrections have been made to the abstract].


Assuntos
Fator VIII , Hemofilia A , Farmacoeconomia , Fator VIII/genética , Fator VIII/imunologia , Fator VIII/uso terapêutico , Predisposição Genética para Doença , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemofilia A/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Tolerância Imunológica/genética , Isoanticorpos/imunologia , Fatores de Risco
2.
Haemophilia ; 16(102): 44-55, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20536985

RESUMO

An HLA-DRA-DRB1*0101-restricted T-cell epitope in the factor VIII (FVIII) C2 domain occurred in a mild haemophilia A patient with missense substitution FVIII-A2201P. His T cells responded to synthetic peptides FVIII(2186-2205) and FVIII(2194-2213) (J Thromb Haemost 2007; 5: 2399). T cells from family members with genotype FVIII-A2201P were analysed to determine if FVIII-specific T cells occur in individuals with a haemophilic mutation but no clinically significant inhibitor response. Fluorescent MHC class II tetramers corresponding to subjects'HLA-DRB1 types were loaded with 20-mer peptides and utilized to label antigen-specific CD4+ T cells. T-cell responses to peptides spanning the FVIII-C2 sequence were evaluated. T cells recognizing specific peptides were cloned, and antigen specificity was verified by proliferation assays. Plasma and/or purified IgG samples were tested for FVIII inhibitory activity. CD4+ T cells and T-cell clones from two brothers who shared the DRB1*0101 allele responded to FVIII(2194-2213). A haemophilic cousin's HLA-DRA-DRB1*1104-restricted response to FVIII(2202-2221) was detected only when CD4+CD25+ cells were depleted. A great uncle and two obligate carriers had no detectable FVIII-C2-specific T cells. Concentrated IgG from the brother without a clinical inhibitor response showed a low-titre FVIII inhibitor. FVIII-specific T cells and inhibitory IgG were found in a previously infused, haemophilic subject who had a sub-clinical FVIII inhibitor. CD4+CD25+ depleted T cells from a non-infused haemophilic cousin recognized an overlapping FVIII epitope, indicating a latent HLA-DRA-DRB1*1104-restricted T-cell response to FVIII. Specific T-cell responses to FVIII can occur without clinically significant inhibitors.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Fator VII/genética , Fator VII/imunologia , Antígenos HLA-DR/genética , Hemofilia A/genética , Hemofilia A/imunologia , Linfócitos T/imunologia , Proliferação de Células , Mapeamento de Epitopos , Epitopos , Genótipo , Antígenos HLA-DR/análise , Cadeias HLA-DRB1 , Humanos , Mutação de Sentido Incorreto , Linfócitos T/citologia
3.
Haemophilia ; 15(4): 942-51, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19473408

RESUMO

Haemophilia A (HA) is a bleeding disorder caused by mutations within the X-linked F8 gene. A series of 42 unrelated Moldovan patients with HA had their disease-causative mutation determined to provide clinically valuable genotyping information for a historically underserved population and to utilize factor VIII (FVIII) structural information to analyse the effects of haemophilic missense substitutions. DNA samples were analysed to detect intron 22 and intron 1 inversions followed by heteroduplex analysis of PCR-amplified fragments containing all exonic sequences. Missense sites identified by DNA sequencing were visualized in the recently described crystal structures of human FVIII. Of the 26 different point mutations, 12 were novel. Gel electrophoresis identified samples with a second major DNA band that migrated abnormally; these amplified products were sequenced. Thirteen intron 22 inversions and one intron 1 inversion were found. Two patients had large, partial gene deletions and there were six frameshift, two non-sense, two splicing and 16 missense genotypes. Two subjects with an intron 22 inversion and one with a large, partial gene deletion developed an alloimmune inhibitor. Their localization suggests intra- and possibly inter-molecular interactions that are important for the structural integrity and/or procoagulant function of FVIII.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/genética , DNA Recombinante/genética , Fator VIII/genética , Hemofilia A/genética , Mutação Puntual/genética , Inibidores dos Fatores de Coagulação Sanguínea/fisiologia , DNA Recombinante/ultraestrutura , Fator VIII/ultraestrutura , Feminino , Deleção de Genes , Genótipo , Hemofilia A/epidemiologia , Humanos , Masculino , Moldávia/epidemiologia , Mutação de Sentido Incorreto/genética , Fenótipo
4.
J Thromb Haemost ; 5(12): 2399-407, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18034765

RESUMO

BACKGROUND: Antibodies that neutralize factor (F) VIII activity, clinically referred to as 'inhibitors', complicate the treatment of hemophilia A patients; current tolerance and bypass strategies are extremely costly and sometimes ineffective. The development of inhibitors requires T-cell help. OBJECTIVES: We characterized T-cell responses of a subject with mild hemophilia A with missense genotype A2201P for one year following his initial inhibitor response, with the goals of defining the primary epitope(s) and its (their) MHC Class II restriction. We investigated the possible involvement of regulatory T cells in modulating immune responses. PATIENTS/METHODS: The subject developed high-titer FVIII-neutralizing antibodies (250 BU mL(-1)) that declined over time to 8 BU ml(-1). His clotting activity was initially impaired (3%) but returned to baseline (8-10%) within four weeks. MHC Class II tetramers were used to analyze his CD4 T cells, which were stimulated with peptides spanning the C2 domain. Responses of total and CD25-depleted CD4 cells to sequences containing A2201 (native), P2201 (hemophilic), and other predicted T-cell epitopes were evaluated. RESULTS AND CONCLUSIONS: An HLA-DRA-DRB1*0101 restricted T-cell epitope containing the wild-type A2201 sequence was identified. Interestingly, peptides containing A2201 were recognized by CD4 T cells at all time points, whereas a P2201 peptide was recognized only near the initial peak response. The responsiveness of CD25-depleted CD4 cells to an A2201 peptide was enhanced 11 and 19 weeks following inhibitor detection, suggesting the possible involvement of CD4+CD25+ regulatory T cells in modulating immune responses. Patient-derived T-cell clones proliferated in response to C2 protein and to peptides containing A2201 but not P2201.


Assuntos
Autoanticorpos/sangue , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Fator VIII/imunologia , Antígenos HLA-A/imunologia , Hemofilia A/imunologia , Tolerância a Antígenos Próprios , Adulto , Coagulação Sanguínea , Mapeamento de Epitopos , Fator VIII/genética , Genótipo , Antígenos HLA-A/genética , Cadeias HLA-DRB1 , Hemofilia A/sangue , Hemofilia A/genética , Humanos , Subunidade alfa de Receptor de Interleucina-2/análise , Masculino , Mutação de Sentido Incorreto , Fenótipo , Índice de Gravidade de Doença , Fatores de Tempo
5.
J Thromb Haemost ; 4(10): 2172-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16824190

RESUMO

BACKGROUND: The development of anti-factor VIII (FVIII) neutralizing antibodies (inhibitors) is a significant obstacle to FVIII replacement therapy. OBJECTIVE: As mucosal administration of an antigen may induce immune tolerance we have evaluated the efficacy of mucosal antigen exposure to achieve tolerance to FVIII. METHODS: We investigated the effects of oral and nasal administration of the purified FVIII C2 domain (FVIII-C2) to FVIII-deficient BALB/c mice prior to FVIII protein challenge. Mice received oral or nasal doses of FVIII-C2, followed by a subcutaneous challenge of either FVIII-C2 or FVIII. The development of anti-FVIII inhibitors, cytokine production by splenocytes in vitro, and adoptive transfer assays were analyzed. RESULTS AND CONCLUSIONS: Mucosal administration of FVIII-C2 decreases the titer of anti-FVIII-C2 inhibitors after FVIII-C2 challenge, and decreases the percentage of FVIII-C2 specific antibodies after challenge with full-length FVIII. Tolerance induction to FVIII-C2 is associated with increased IL-10 production by splenocytes in vitro, and can be adoptively transferred to naïve mice. This study is the first to demonstrate that tolerance to the FVIII-C2 domain can be induced via the mucosal route. Based on these results, the potential use of FVIII-specific mucosal tolerance induction as an immunotherapy treatment for anti-FVIII inhibitor development warrants further investigation.


Assuntos
Fator VIII/genética , Fator VIII/metabolismo , Fator VIII/uso terapêutico , Tolerância Imunológica , Mucosa/metabolismo , Administração Intranasal , Animais , Linfócitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Fator VIII/química , Feminino , Hemofilia A/sangue , Hemofilia A/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucosa/patologia , Estrutura Terciária de Proteína , Baço/citologia , Fatores de Tempo
6.
Structure ; 5(1): 125-38, 1997 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9016719

RESUMO

BACKGROUND: Blood coagulation occurs by a cascade of zymogen activation resulting from minor proteolysis. The final stage of coagulation involves thrombin generation and limited proteolysis of fibrinogen to give spontaneously polymerizing fibrin. The resulting fibrin network is covalently crosslinked by factor XIIIa to yield a stable blood clot. Fibrinogen is a 340 kDa glycoprotein composed of six polypeptide chains, (alphabetagamma)2, held together by 29 disulfide bonds. The globular C terminus of the gamma chain contains a fibrin-polymerization surface, the principal factor XIIIa crosslinking site, the platelet receptor recognition site, and a calcium-binding site. Structural information on this domain should thus prove helpful in understanding clot formation. RESULTS: The X-ray crystallographic structure of the 30 kDa globular C terminus of the gamma chain of human fibrinogen has been determined in one crystal form using multiple isomorphous replacement methods. The refined coordinates were used to solve the structure in two more crystal forms by molecular replacement; the crystal structures have been refined against diffraction data to either 2.5 A or 2.1 A resolution. Three domains were identified in the structure, including a C-terminal fibrin-polymerization domain (P), which contains a single calcium-binding site and a deep binding pocket that provides the polymerization surface. The overall structure has a pronounced dipole moment, and the C-terminal residues appear highly flexible. CONCLUSIONS: The polymerization domain in the gamma chain is the most variable among a family of fibrinogen-related proteins and contains many acidic residues. These residues contribute to the molecular dipole moment in the structure, which may allow electrostatic steering to guide the alignment of fibrin monomers during the polymerization process. The flexibility of the C-terminal residues, which contain one of the factor XIIIa crosslinking sites and the platelet receptor recognition site, may be important in the function of this domain.


Assuntos
Fibrinogênio/química , Sequência de Aminoácidos , Sítios de Ligação , Cálcio/metabolismo , Reagentes de Ligações Cruzadas/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Alinhamento de Sequência , Transglutaminases/farmacologia
7.
J Thromb Haemost ; 11(1): 47-55, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23140211

RESUMO

Only a fraction of patients with hemophilia A develop a neutralizing antibody (inhibitor) response to therapeutic infusions of factor VIII. Our present understanding of the underlying causes of the immunogenicity of this protein is limited. In the past few years, insights into the uptake and processing of FVIII by antigen-presenting cells (APCs) have expanded significantly. Although the mechanism of endocytosis remains unclear, current data indicate that FVIII enters APCs via its C1 domain. Its subsequent processing within endolysosomes allows for presentation of a heterogeneous collection of FVIII-derived peptides on major histocompatibility complex (MHC) class II, and this peptide-MHC class II complex may then be recognized by cognate effector CD4(+) T cells, leading to anti-FVIII antibody production. Here we aim to summarize recent knowledge gained about FVIII processing and presentation by APCs, as well as the diversity of the FVIII-specific T-cell repertoire in mice and humans. Moreover, we discuss possible factors that can drive FVIII immunogenicity. We believe that increasing understanding of the immune recognition of FVIII and the cellular mechanisms of anti-FVIII antibody production will lead to novel therapeutic approaches to prevent inhibitor formation in patients with hemophilia A.


Assuntos
Anticorpos Neutralizantes/sangue , Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Coagulantes/imunologia , Fator VIII/imunologia , Hemofilia A/tratamento farmacológico , Animais , Coagulantes/administração & dosagem , Coagulantes/química , Coagulantes/metabolismo , Endocitose , Epitopos , Fator VIII/administração & dosagem , Fator VIII/química , Fator VIII/metabolismo , Hemofilia A/sangue , Hemofilia A/imunologia , Humanos , Ativação Linfocitária , Modelos Moleculares , Conformação Proteica , Fator de von Willebrand/metabolismo
8.
J Thromb Haemost ; 9(4): 689-99, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21251204

RESUMO

BACKGROUND: Development of neutralizing anti-factor (F)VIII antibodies ('inhibitors') is a serious clinical problem in hemophilia A. Increased inhibitor risk has been associated with certain FVIII missense substitutions, including R593C in the A2 domain. OBJECTIVES: The aim of the present study was to identify T-cell epitopes in FVIII and characterize T-cell responses in two unrelated hemophilia A subjects sharing F8-R593C and HLA-DRB1*1101 genotypes. We hypothesized that the hemophilic substitution site coincides with an important T-cell epitope. PATIENTS/METHODS: The binding affinities of peptides for recombinant HLA-DR proteins were measured and compared with epitope prediction results. CD4+ T cells were stimulated using peptides and stained with fluorescent, peptide-loaded tetramers. RESULTS: The inhibitor subjects, but not HLA-matched controls, had high-avidity HLA-DRB1*1101-restricted T-cell responses against FVIII(589-608), which contains the hemophilic missense site. Antigen-specific T cells secreted Th1 and Th2 cytokines and proliferated in response to FVIII and FVIII(592-603). FVIII(589-608) bound with physiologically relevant (micromolar) IC(50) values to recombinant DR0101, DR1101 and DR1501 proteins. CONCLUSIONS: Hemophilia A patients with R593C missense substitutions and these HLA haplotypes had an increased incidence of inhibitors in our cohorts, supporting a paradigm in which presentation of FVIII epitopes containing the wild-type R593 influences inhibitor risk in this hemophilia A sub-population.


Assuntos
Epitopos/imunologia , Fator VIII/genética , Hemofilia A/imunologia , Mutação de Sentido Incorreto , Linfócitos T/imunologia , Sequência de Aminoácidos , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Antígenos HLA-DR/genética , Cadeias HLA-DRB1 , Humanos , Dados de Sequência Molecular , Linfócitos T/citologia
9.
Haemophilia ; 13(3): 317-22, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17498082

RESUMO

Inhibitors of FVIII are usually IgG polyclonal antibodies that develop as alloimmune responses in patients with congenital haemophilia A or as autoimmune responses resulting in acquired haemophilia. Their recognition can be difficult, especially when the titre is low. Furthermore, results from a Bethesda assay often require several days as samples are referred to a specialty laboratory. The aim of this study is to assess the utility of an ELISA system for detecting immune responses to FVIII. A total of 246 plasma samples submitted from 176 individuals with immune responses to FVIII, as verified with the Bethesda assay, and samples from 50 control subjects were tested for the presence of FVIII-specific IgG using an ELISA-based assay. Paired sera from 18 of the patients were also tested by the ELISA. Of the 246 samples that were positive for a FVIII inhibitor by the Bethesda assay, 235 (95.5%) were also positive by ELISA. The regression coefficient, using Log BU was r = 0.82. The correlation data were strengthened when 27 inhibitor samples were diluted further. There was a strong correlation between ELISA results for the 18-paired serum and plasma samples (r = 0.99). There is a strong correlation between the ELISA and Bethesda methods in detecting immune responses to FVIII. The ELISA provides rapid screening that could be available well in advance of confirmation by the Bethesda assay.


Assuntos
Autoanticorpos/sangue , Fator VIII/imunologia , Hemofilia A/sangue , Autoanticorpos/imunologia , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática/métodos , Ensaio de Imunoadsorção Enzimática/normas , Hemofilia A/imunologia , Humanos
10.
Curr Opin Drug Discov Devel ; 3(5): 516-26, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19649880

RESUMO

Factor VIII is a plasma glycoprotein that becomes activated during blood coagulation. Through its association with other procoagulant components at the wound site, it leads to a tremendous acceleration in the production of thrombin, and thus to the rate at which bleeding is staunched. Deficiencies in Factor VIII can result in hemophilia A, the most common hereditary bleeding disorder. Advances in molecular biology and protein chemistry led to characterization of Factor VIII and its gene in the early 1980s. Over the past 15 years, PCR methods combined with careful clinical studies have identified many of the molecular defects associated with hemophilia A. Concurrently, basic research utilizing both plasma-derived Factor VIII and recombinant constructs has led to a greatly improved understanding of the functions of specific regions within this large glycoprotein. This review focuses on recent advances in elucidating the roles of the Factor VIII carboxy terminal C2 domain. Particular emphasis is placed upon interpreting biophysical measurements and clinical data in light of the recently obtained high-resolution crystal structure of the recombinant C2 domain. Some interesting directions for future experiments are also suggested.

11.
J Biol Chem ; 272(38): 23792-8, 1997 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-9295325

RESUMO

The carboxyl-terminal region of the gamma chain of fibrinogen is involved in calcium binding, fibrin polymerization, factor XIIIa-mediated cross-linking, and binding to the platelet fibrin(ogen) receptor. Protein fragments encoding amino acids Val143 to Val411 (rFbggammaC30) or Val143 to Leu427 (gamma'C30) from the carboxyl end of the gamma or gamma' chains, respectively, of human fibrinogen were expressed in yeast (Pichia pastoris) and characterized as to their cross-linking by factor XIIIa, polymerization pocket, and calcium-binding site. rFbggammaC30 and gamma'C30 were both readily cross-linked by factor XIIIa, but only rFbggammaC30 was capable of inhibiting thrombin-induced platelet aggregation. Two mutants, gammaC30-Q329R and gammaC30-D364A, which were based on the three-dimensional structure of the polymerization pocket within rFbggammaC30 and on information derived from naturally occurring mutant fibrinogens, were also expressed and characterized. rFbggammaC30 inhibited (desAA)fibrin polymerization in a dose-dependent manner, while the two mutant forms did not. Similarly, rFbggammaC30 and gamma'C30 were protected from plasmin degradation by the presence of Ca2+ or the peptide Gly-Pro-Arg-Pro, indicating that a functional Ca2+-binding site and polymerization pocket are contained within each of these fragments. The mutant fragments, however, were protected from plasmin only by metal ions, while no protective effect was conferred by GPRP or by any other peptide tested. These results indicate that the polymerization pocket "a", which binds the peptide GPRP, functions independently from the nearby calcium-binding site and that amino acids Gln329 and Asp364 play a crucial role in fibrin polymerization.


Assuntos
Cálcio/metabolismo , Fibrinogênio/química , Sequência de Aminoácidos , Sítios de Ligação , Biopolímeros , Fibrinogênio/metabolismo , Fibrinolisina/metabolismo , Humanos , Hidrólise , Masculino , Agregação Plaquetária , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Trombina/metabolismo
12.
Proc Natl Acad Sci U S A ; 94(14): 7176-81, 1997 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-9207064

RESUMO

After vascular injury, a cascade of serine protease activations leads to the conversion of the soluble fibrinogen molecule into fibrin. The fibrin monomers then polymerize spontaneously and noncovalently to form a fibrin gel. The primary interaction of this polymerization reaction is between the newly exposed N-terminal Gly-Pro-Arg sequence of the alpha chain of one fibrin molecule and the C-terminal region of a gamma chain of an adjacent fibrin(ogen) molecule. In this report, the polymerization pocket has been identified by determining the crystal structure of a 30-kDa C-terminal fragment of the fibrin(ogen) gamma chain complexed with the peptide Gly-Pro-Arg-Pro. This peptide mimics the N terminus of the alpha chain of fibrin. The conformational change in the protein upon binding the peptide is subtle, with electrostatic interactions primarily mediating the association. This is consistent with biophysical experiments carried out over the last 50 years on this fundamental polymerization reaction.


Assuntos
Fibrina/química , Oligopeptídeos/química , Dimerização , Fibrina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Oligopeptídeos/metabolismo , Conformação Proteica
13.
Nature ; 402(6760): 439-42, 1999 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-10586887

RESUMO

Human factor VIII is a plasma glycoprotein that has a critical role in blood coagulation. Factor VIII circulates as a complex with von Willebrand factor. After cleavage by thrombin, factor VIIIa associates with factor IXa at the surface of activated platelets or endothelial cells. This complex activates factor X (refs 6, 7), which in turn converts prothrombin to thrombin in the presence of factor Va (refs 8, 9). The carboxyl-terminal C2 domain of factor VIII contains sites that are essential for its binding to von Willebrand factor and to negatively charged phospholipid surfaces. Here we report the structure of human factor VIII C2 domain at 1.5 A resolution. The structure reveals a beta-sandwich core, from which two beta-turns and a loop display a group of solvent-exposed hydrophobic residues. Behind the hydrophobic surface lies a ring of positively charged residues. This motif suggests a mechanism for membrane binding involving both hydrophobic and electrostatic interactions. The structure explains, in part, mutations in the C2 region of factor VIII that lead to bleeding disorders in haemophilia A.


Assuntos
Fator VIII/química , Cristalografia por Raios X , Eletroquímica , Fator VIII/genética , Hemofilia A/genética , Humanos , Modelos Moleculares , Mutação Puntual , Conformação Proteica , Estrutura Terciária de Proteína
14.
Blood ; 98(1): 13-9, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11418455

RESUMO

The development of an immune response to infused factor VIII is a complication affecting many patients with hemophilia A. Inhibitor antibodies bind to antigenic determinants on the factor VIII molecule and block its procoagulant activity. A patient-derived inhibitory immunoglobulin G4kappa antibody (BO2C11) produced by an immortalized memory B-lymphocyte cell line interferes with the binding of factor VIII to phospholipid surfaces and to von Willebrand factor. The structure of a Fab fragment derived from this antibody complexed with the factor VIII C2 domain was determined at 2.0 A resolution. The Fab interacts with solvent-exposed basic and hydrophobic side chains that form a membrane-association surface of factor VIII. This atomic resolution structure suggests a variety of amino acid substitutions in the C2 domain of factor VIII that might prevent the binding of anti-C2 inhibitor antibodies without significantly compromising the procoagulant functions of factor VIII.


Assuntos
Complexo Antígeno-Anticorpo/química , Fator VIII/imunologia , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Linfócitos B/imunologia , Ligação Competitiva , Linhagem Celular , Cristalografia por Raios X , Mapeamento de Epitopos , Hemofilia A/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Fragmentos Fab das Imunoglobulinas/metabolismo , Cinética , Modelos Moleculares , Ligação Proteica , Estrutura Terciária de Proteína , Fator de von Willebrand/metabolismo
15.
Blood ; 96(3): 979-87, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910913

RESUMO

Factor VIII C domains contain key binding sites for von Willebrand factor (vWF) and phospholipid membranes. Hemophilic patients were screened for factor VIII C-domain mutations to provide a well-characterized series. Mutated residues were localized to the high-resolution C2 structure and to a homology model of C1. Of 30 families found with mutations in the C domains, there were 14 missense changes, and 9 of these were novel. Of the missense mutations, 10 were associated with reduced vWF binding and 8 were at residues with surface-exposed side chains. Six of the 10 mutants had nearly equivalent factor VIII clotting activity and antigen level, suggesting that reduced vWF binding could cause hemophilia by reducing factor VIII stability in circulation. When the present series was combined with previously described mutations from an online international database, 11 C1 and C2 mutations in patients with mild or moderately severe hemophilia A were associated with antibody-inhibitor development in at least one affected individual. Of these substitutions, 6 occurred at surface-exposed residues. As further details of the C1 structure and its interface with C2 become available, and as binding studies are performed on the plasma of more patients with hemophilic C-domain mutations, prediction of surface binding sites should improve, allowing confirmation by site-specific mutagenesis of surface-exposed residues.


Assuntos
Fator VIII/química , Fator VIII/genética , Sequência de Aminoácidos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Conformação Proteica , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa