Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1784(5): 753-63, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18329389

RESUMEN

Factor VIII is activated and inactivated by plasmin by limited proteolysis. In our one-stage clotting assay, these plasmin-catalyzed reactions were inhibited by the addition of isolated factor VIII A2 subunits and by Glu-Gly-Arg-active-site modified factor IXa. SDS-PAGE analysis showed that an anti-A2 monoclonal antibody, recognizing the factor IXa-interactive site (residues 484-509), blocked the plasmin-catalyzed cleavage at Arg(336) and Arg(372) but not at Arg(740). Surface plasmon resonance-based assays and ELISA demonstrated that the A2 subunit bound to active-site modified anhydro-plasmin with high affinity (K(d): 21 nM). Both an anti-A2 monoclonal antibody and a peptide comprising of A2 residues 479-504 blocked A2 binding by approximately 80% and approximately 55%, respectively. Mutant A2 molecules where the basic residues in A2 were converted to alanine were evaluated for binding of anhydro-plasmin. Among the tested mutants, the R484A A2 mutant possessed approximately 250-fold lower affinity than the wild-type A2. The affinities of K377A, K466A, and R471A mutants were decreased by 10-20-fold. The inhibitory effect of R484A mutant on plasmin-catalyzed inactivation of factor VIIIa was approximately 20% of that of wild-type A2. In addition, the inactivation rate by plasmin of factor VIIIa reconstituted with R484A mutant was approximately 3-fold lower than that with wild-type A2. These findings demonstrate that Arg(484) plays a key role within the A2 plasmin-binding site, responsible for plasmin-catalyzed factor VIII(a) inactivation.


Asunto(s)
Factor VIII/química , Factor VIII/metabolismo , Fibrinolisina/metabolismo , Mapeo de Interacción de Proteínas , Ácido Aminocaproico/farmacología , Anticuerpos/farmacología , Sitios de Unión , Catálisis/efectos de los fármacos , Factor IXa/metabolismo , Humanos , Cinética , Proteínas Mutantes/metabolismo , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Subunidades de Proteína/metabolismo , Resonancia por Plasmón de Superficie
2.
Thromb Haemost ; 102(4): 645-55, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19806249

RESUMEN

We have recently demonstrated that protein S impairs the intrinsic tenase complex, independent of activated protein C, in competitive interactions between the A2 and A3 domains of factor VIIIa and factor IXa. In the present study, we have identified a protein S-interactive site in the A2 domain of factor VIIIa. Anti-A2 monoclonal antibody recognising a factor IXa-functional region (residues 484-509) on A2, and synthetic peptide inhibited the A2 binding to protein S by approximately 60% and approximately 70%, respectively, in solid-phase binding assays. The 484-509 peptide directly bound to protein S dose-dependently. Covalent cross-linking was observed between the 484-509 peptide and protein S following reaction with EDC (1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide). The cross-linked adduct was consistent with 1:1 stoichiometry of reactants. Cross-linking formation was blocked by addition of the 484-497 peptide, but not by the 498-509 peptide. Furthermore, N-terminal sequence analysis of the 484-509 peptide-protein S adduct showed that three sequential residues (S488, R489, and R490) in A2 were not identified, suggesting that these residues participate in cross-link formation. Mutant A2 molecules where these residues were converted to alanine were evaluated for the binding of protein S. The S488A, R489A, and R490A mutants demonstrated approximately four-fold lower affinity than wild-type A2. These results indicate that the 484-509 region in the A2 domain of factor VIIIa, in particular sequential residues at positions 488-490, contributes to a unique protein S-interactive site.


Asunto(s)
Dominio Catalítico/genética , Factor VIII/metabolismo , Fragmentos de Péptidos/metabolismo , Proteína S/metabolismo , Proteínas Recombinantes/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Dominio Catalítico/inmunología , Factor VIII/genética , Factor VIII/inmunología , Humanos , Datos de Secuencia Molecular , Complejos Multienzimáticos , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/genética , Unión Proteica/inmunología , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/inmunología , Multimerización de Proteína/genética , Proteína S/química , Proteína S/inmunología , Proteínas Recombinantes/genética , Resonancia por Plasmón de Superficie
3.
Semin Thromb Hemost ; 35(8): 735-51, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20169510

RESUMEN

Development of inhibitory antibodies (inhibitors) to factor VIII (FVIII) is the most serious adverse event in replacement therapy of hemophilia A patients. The etiology and management of this condition remain major challenges for both researchers and clinicians. In the present review, we discuss recent advances in understanding the molecular mechanisms by which inhibitors inactivate FVIII and experimental approaches used for the mapping of inhibitor epitopes. We also present a comparative analysis of treatment of hemophilia A patients with inhibitors with currently available bypassing agents-activated prothrombin complex concentrate (FEIBA VH; Baxter Healthcare Corp., Westlake Village, CA) and recombinant activated factor VII (NovoSeven; Novo Nordisk, Princeton, NJ)-and describe some ongoing research programs aimed at developing new treatment options for these patients. Availability of sensitive and standardized laboratory assays that would assist in monitoring the effectiveness of bypass therapies is essential for designing customized treatment regimens and improvement in the management of health conditions of hemophilia patients with inhibitors.


Asunto(s)
Hemofilia A/tratamiento farmacológico , Animales , Anticuerpos Catalíticos/inmunología , Inhibidores de Factor de Coagulación Sanguínea/genética , Inhibidores de Factor de Coagulación Sanguínea/inmunología , Factores de Coagulación Sanguínea/uso terapéutico , Ensayo de Inmunoadsorción Enzimática/métodos , Mapeo Epitopo/métodos , Epítopos/inmunología , Factor VIII/inmunología , Factor VIIa/uso terapéutico , Hemofilia A/diagnóstico , Hemofilia A/inmunología , Hemorragia/tratamiento farmacológico , Humanos , Biblioteca de Péptidos , Estructura Terciaria de Proteína/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico , Tromboelastografía
4.
Br J Haematol ; 140(4): 433-43, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18081893

RESUMEN

FVIII is activated by cleavage at Arg(372), Arg(740), and Arg(1689) by thrombin. This study showed that an anti-A2 monoclonal antibody, with a specific epitope for residues 484-509, and anti-FVIII inhibitor alloantibodies with similar A2 epitopes, inhibited thrombin-catalyzed FVIII activation. Sodium dodecyl sulphate polyacrylamide gel electrophoresis analysis showed that cleavage at Arg(372) but not at Arg(740) occurred at approximately fourfold decreased rate in the presence of anti-A2 antibody. Peptide 484-509 also inhibited co-factor activation, consistent with inhibition of cleavage at Arg(372). Direct binding studies using active-site modified thrombin showed that a 484-509 peptide as well as the anti-A2 antibodies blocked the A2-thrombin binding. Furthermore, covalent cross-linking was observed between the 484-509 peptide and thrombin following reaction with 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide. Mutant A2 molecules in which the clustered basic residues in this sequence were converted to alanine were used to assess the binding reactions in a surface plasmon resonance-based assay. Mutants R484A, R489A, R490A, H497A and K499A possessed two to fivefold lower affinity than wild-type A2. These findings demonstrate that clustered basic residues within the 484-509 region of the A2 domain play a part of key role in thrombin-binding, which is responsible for thrombin-catalyzed FVIII activation by cleavage at Arg(372).


Asunto(s)
Factor VIII/química , Trombina/química , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Unión Competitiva , Catálisis , Electroforesis en Gel de Poliacrilamida/métodos , Factor VIII/inmunología , Humanos , Isoanticuerpos , Fragmentos de Péptidos , Unión Proteica , Proteínas Recombinantes/química
5.
Br J Haematol ; 143(3): 409-20, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18759761

RESUMEN

Protein S functions as an activated protein C (APC)-independent anticoagulant in the inhibition of intrinsic factor X activation, although the precise mechanisms remain to be fully investigated. In the present study, protein S diminished factor VIIIa/factor IXa-dependent factor X activation, independent of APC, in a functional Xa generation assay. The presence of protein S resulted in an c. 17-fold increase in K(m) for factor IXa with factor VIIIa in the factor Xase complex, but an c. twofold decrease in K(m) for factor X. Surface plasmon resonance-based assays showed that factor VIII, particularly the A2 and A3 domains, bound to immobilized protein S (K(d); c. 10 nmol/l). Competition binding assays using Glu-Gly-Arg-active-site modified factor IXa showed that factor IXa inhibited the reaction between protein S and both the A2 and A3 domains. Furthermore, Sodium dodecyl sulphate polyacrylamide gel electrophoresis revealed that the cleavage rate of factor VIIIa at Arg(336) by factor IXa was c. 1.8-fold lower in the presence of protein S than in its absence. These data indicate that protein S not only down-regulates factor VIIIa activity as a cofactor of APC, but also directly impairs the assembly of the factor Xase complex, independent of APC, in a competitive interaction between factor IXa and factor VIIIa.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Factor VIIIa/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína S/farmacología , Unión Competitiva , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Factor IXa/metabolismo , Factor IXa/farmacología , Factor X/metabolismo , Factor X/farmacología , Humanos , Fosfolípidos/metabolismo , Proteína C/fisiología , Proteína S/metabolismo , Proteínas Recombinantes/metabolismo , Factor de von Willebrand/metabolismo
6.
Haematologica ; 93(1): 83-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18166789

RESUMEN

BACKGROUND: The development of factor VIII (FVIII) inhibitors remains the major hurdle in the clinical management of patients with hemophilia A. FVIII uptake by professional antigen-presenting cells (APC) is the first step involved in initiation of immune responses to FVIII. Studies on FVIII catabolism have highlighted the role played by CD91/LRP as a potential target for increasing FVIII half-life in patients and prolonging treatment efficiency. We investigated the involvement of CD91 in FVIII endocytosis by human dendritic cells (DC), a model of professional APC. DESIGN AND METHODS: Immature DC were generated from circulating monocytes from healthy donors. Surface expression of CD91 was assessed by flow cytometry. Uptake of fluorescein isothiocyanate-conjugated ligands by immature DC was studied in the presence of various blocking agents. RESULTS: CD91 was expressed on approximately 20% of DC and mediated the internalization of its model ligand, alpha2-macroglobulin. DC internalized FVIII and activated a human FVIII-specific T-cell clone in a dose-dependent manner. FVIII uptake by DC and subsequent T-cell activation were not inhibited by receptor-associated protein. CONCLUSIONS: Our results indicate that CD91 and other members of the LDL receptor family are not strongly implicated in FVIII internalization by monocyte-derived DC, and suggest the involvement of alternative divalent ion-dependent endocytic receptors.


Asunto(s)
Antígenos CD/biosíntesis , Células Dendríticas/citología , Factor VIII/biosíntesis , Linfocitos T/citología , Animales , Separación Celular , Endocitosis , Factor VIII/metabolismo , Hemofilia A/genética , Humanos , Leucocitos Mononucleares/citología , Ligandos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Activación de Linfocitos , Ratones , Monocitos/citología , Monocitos/metabolismo
7.
Blood Coagul Fibrinolysis ; 19(6): 543-55, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18685438

RESUMEN

Coagulation factor VIII interacts with several members of the low-density lipoprotein receptor family including low-density lipoprotein receptor-related protein, low-density lipoprotein receptor, and very low-density lipoprotein receptor. The present study was aimed to compare the mechanisms of factor VIII interaction with low-density lipoprotein receptor-related protein, megalin, low-density lipoprotein receptor, and very low-density lipoprotein receptor in order to reveal a general mode of these interactions. Binding of plasma-derived factor VIII and its fragments to recombinant soluble ligand-binding domain of low-density lipoprotein receptor (sLDLR1-7) and purified megalin was studied in solid phase and surface plasmon resonance assays. Full-length factor VIII and its light chain bound to the receptors with similar affinities (KD = 260 +/- 9 and 156 +/- 4 nmol/l, respectively, for megalin and KD = 210 +/- 3 and 174 +/- 13 nmol/l, respectively, for sLDLR1-7). Von Willebrand factor inhibited factor VIII binding to both receptors. In contrast to the light chain, exposure of the high-affinity receptor-binding site within the heavy chain (KD = 22 +/- 4 nmol/l for megalin and 17 +/- 3 nmol/l for sLDLR1-7) required proteolytic cleavage by thrombin. This site was mapped to the A2 domain residues 484-509, based on the inhibitory effects of anti-A2 monoclonal antibody 413, and is shared by all four receptors. Using a panel of A2 mutants, we identified key amino acid residues- positively charged K466, R471, R489 and R490, and hydrophilic residues Y487 and S488- which form the frame of this 'consensus' binding site. We conclude that interaction of factor VIII with the members of the low-density lipoprotein receptor family follows the general mode, requires dissociation of factor VIII from von Willebrand factor, and is activation sensitive.


Asunto(s)
Factor VIII/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Receptores de LDL/metabolismo , Sustitución de Aminoácidos , Aminoácidos/fisiología , Sitios de Unión , Secuencia de Consenso , Factor VIII/química , Humanos , Modelos Moleculares , Mutación Missense , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Resonancia por Plasmón de Superficie , Trombina/farmacología , Factor de von Willebrand/metabolismo
8.
Blood Coagul Fibrinolysis ; 19(2): 166-77, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18277139

RESUMEN

Coagulation factor VIII (FVIII) is a ligand for two members of the low-density lipoprotein receptor family, low-density lipoprotein receptor-related protein (LRP) and low-density lipoprotein receptor, which cooperate in regulating clearance of FVIII from the circulation. This study was aimed to explore the mechanism of interaction of FVIII with very low density lipoprotein receptor (VLDLR), another member of the family, and map receptor-binding sites. Binding of plasma-derived FVIII and its fragments to recombinant soluble ectodomain of VLDLR (sVLDLR) was studied in solid-phase and surface plasmon resonance assays. Full-length FVIII and its light chain bound to sVLDLR with similar affinities (KD = 114 +/- 14 and 95 +/- 11 nmol/l, respectively); in contrast, exposure of high-affinity VLDLR-binding site within the heavy chain (KD = 30 +/- 2 nmol/l) required proteolytic cleavage by thrombin. The VLDLR-binding sites within heavy and light chains were mapped to the A2 domain residues 484-509 and the A3-C1 fragment, based on the inhibitory effects of anti-A2 monoclonal antibody 413 and anti-A3-C1 antibody fragment scFv KM33, respectively, previously shown to inhibit FVIII/LRP interaction. Soluble ligand-binding fragment of VLDLR inhibited activation of factor X by the intrinsic Xase in purified system. In cell culture, a higher Xase activity was associated with wild-type human embryonic kidney cells compared with transfected cells that express VLDLR on the cell surface. We conclude that the binding sites for VLDLR and LRP within FVIII overlap and the A2 site becomes exposed upon physiological activation of FVIII. A functional role of FVIII/VLDLR interaction may be related to regulation of intrinsic Xase activity.


Asunto(s)
Factor VIIIa/fisiología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Receptores de LDL/fisiología , Sitios de Unión/fisiología , Coagulación Sanguínea/fisiología , Células Cultivadas , Cisteína Endopeptidasas/fisiología , Factor VIIIa/química , Humanos , Lipoproteínas VLDL , Proteínas de Neoplasias/fisiología , Resonancia por Plasmón de Superficie
9.
J Med Chem ; 50(18): 4329-39, 2007 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-17691762

RESUMEN

Hemophilia A, one of the most severe bleeding disorders, results from an inherited deficiency of factor VIII (FVIII) function. Treatment by injection of FVIII has been a common procedure for decades. Nevertheless, the production and purification of FVIII remains a challenging task. Current procedures using immunoaffinity chromatography are expensive and suffer from the instability of the applied antibody ligands, which elute along with the product and contaminate it. Recently, FVIII was purified by use of octapeptide ligands, but their low protease-resistance limits their application. We here report the systematic rational and combinatorial optimization procedure that allowed us to transfer the octapeptide ligands into a small peptidomimetic. This compound is the smallest ligand known for separation of such a large protein (330 kDa). It not only binds and purifies FVIII with high efficiency but also is stable, protease-resistant, and cheap to produce in preparative scale. Hence it offers a valuable alternative to antibody-based purification procedures.


Asunto(s)
Factor VIII/aislamiento & purificación , Ácidos Indolacéticos/síntesis química , Oligopéptidos/química , Sustitución de Aminoácidos , Cromatografía de Afinidad/métodos , Estabilidad de Medicamentos , Factor VIII/química , Humanos , Ácidos Indolacéticos/química , Ligandos , Imitación Molecular , Oligopéptidos/síntesis química , Péptido Hidrolasas/química , Polímeros , Unión Proteica , Proteínas Recombinantes/química , Suero , Estereoisomerismo
10.
Thromb Haemost ; 98(6): 1170-81, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18064310

RESUMEN

Catabolism of coagulation factor VIII (FVIII) is mediated by low-density lipoprotein receptor-related protein (LRP). The ligand-binding sites of LRP are formed by complement-type repeats (CR), and CR clusters II and IV bind most of LRP ligands. FVIII contains two major LRP-binding sites located in the A2 and A3 domains. This study was aimed to identify specific complement-type repeats of LRP involved in interaction with the A2 site and to probe their functional importance in A2 catabolism. We generated individual LRP clusters II, III and IV, along with nine overlapping CR triplets encompassing clusters II and IV in a baculovirus expression system and studied their interaction with isolated A2. In surface plasmon resonance (SPR) assay, A2 bound to clusters II and IV with KDs 22 and 39 nM, respectively, and to the majority of CR triplets with affinities in the range of KDs 25-90 nM. Similar affinities were determined for A2 interaction with a panel of CR doublets overlapping cluster II (CR 3-4, 4-5, 5-6, 6-7 and 7-8). These LRP fragments inhibited the binding of 125I-A2 to LRP in solid-phase assay, LRP-mediated internalization of 125I-A2 in cell culture and 125I-A2 clearance from the mouse circulation. Point mutations of critical A2 residues of the LRPbinding site resulted in differential reduction or abolishment of its binding to LRP fragments. We conclude that A2 interacts with LRP via multiple binding sites spanning CR 3-8 in cluster II and CR 23-29 in cluster IV, and the minimal A2-binding unit of LRP is formed by two adjacent CR.


Asunto(s)
Factor VIII/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Baculoviridae/genética , Baculoviridae/metabolismo , Unión Competitiva , Línea Celular , Endocitosis , Factor VIII/química , Factor VIII/genética , Fibroblastos/metabolismo , Humanos , Radioisótopos de Yodo , Proteínas Relacionadas con Receptor de LDL/química , Proteínas Relacionadas con Receptor de LDL/genética , Ligandos , Ratones , Ratones Endogámicos BALB C , Chaperonas Moleculares/metabolismo , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Mutación Puntual , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie , Factores de Tiempo
11.
FEBS J ; 273(2): 374-87, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16403024

RESUMEN

Activation of coagulation factor X (fX) by activated factors IX (fIXa) and VIII (fVIIIa) requires the assembly of the enzyme-cofactor-substrate fIXa-fVIIIa-fX complex on negatively charged phospholipid membranes. Using flow cytometry, we explored formation of the intermediate membrane-bound binary complexes of fIXa, fVIIIa, and fX. Studies of the coordinate binding of coagulation factors to 0.8-microm phospholipid vesicles (25/75 phosphatidylserine/phosphatidylcholine) showed that fVIII (fVIIIa), fIXa, and fX bind to 32 700 +/- 5000 (33 200 +/- 14 100), 20 000 +/- 4500, and 30 500 +/- 1300 binding sites per vesicle with apparent K(d) values of 76 +/- 23 (71 +/- 5), 1510 +/- 430, and 223 +/- 79 nm, respectively. FVIII at 10 nm induced the appearance of additional high-affinity sites for fIXa (1810 +/- 370, 20 +/- 5 nm) and fX (12 630 +/- 690, 14 +/- 4 nm), whereas fX at 100 nm induced high-affinity sites for fIXa (541 +/- 67, 23 +/- 5 nm). The effects of fVIII and fVIIIa on the binding of fIXa or fX were similar. The apparent Michaelis constant of the fX activation by fIXa was a linear function of the fVIIIa concentration with a slope of 1.00 +/- 0.12 and an intrinsic K(m) value of 8.0 +/- 1.5 nm, in agreement with the hypothesis that the reaction rate is limited by the fVIIIa-fX complex formation. In addition, direct correlation was observed between the fX activation rate and formation of the fVIIIa-fX complex. Titration of fX, fVIIIa, phospholipid concentration and phosphatidylserine content suggested that at high fVIIIa concentration the reaction rate is regulated by the concentration of free fX rather than of membrane-bound fX. The obtained results reveal formation of high-affinity fVIIIa-fX complexes on phospholipid membranes and suggest their role in regulating fX activation by anchoring and delivering fX to the enzymatic complex.


Asunto(s)
Factor VIIIa/fisiología , Factor Intrinseco/metabolismo , Activación Enzimática , Factor VIIIa/metabolismo , Factor X/metabolismo , Humanos , Cinética , Fosfolípidos/metabolismo , Unión Proteica
12.
Blood Coagul Fibrinolysis ; 17(5): 379-88, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16788314

RESUMEN

Hereditary deficiency of factor VIII (FVIII), haemophilia A, is treated by plasma-derived FVIII (pd-FVIII) or recombinant FVIII (rFVIII) infusions. B-domain-deleted FVIII (BDD-rFVIII), although generally safe and effective, was less effective than pd-FVIII in prophylaxis -- evidenced by a 2.5-fold higher bleeding incidence. Assessment of BDD-rFVIII activity in chromogenic and one-stage clotting assays gives up to 50% difference in activity values. As earlier studies demonstrated identical activation and cofactor activity of BDD-rFVIII and pd-FVIII, we decided to study susceptibility of thrombin-activated pd-FVIII, full-length rFVIII and BDD-rFVIII to proteolytic inactivation by activated protein C (APC) and activated factor X (FXa) in a purified system. Proteolysis was monitored by Western blot using monoclonal antibodies C5 and R8B12 specific for the A1 and A2 domains, respectively. Inactivation was monitored by measuring the residual cofactor activity of FVIII forms in a one-stage clotting assay. Proteolysis of A1 and A2 domains of activated BDD-rFVIII proceeded 11 or 13 times faster than that of pd-FVIII or full-length rFVIII. Inactivation of activated BDD-rFVIII was two to three times faster by APC and five to six times faster by FXa. We suggest that differences in proteolytic inactivation may contribute to differences between BDD-rFVIII and pd-FVIII in assaying and in clinical use.


Asunto(s)
Factor VIII/fisiología , Factor VIIIa/metabolismo , Factor Xa/farmacología , Fragmentos de Péptidos/fisiología , Proteína C/farmacología , Western Blotting , Humanos , Fragmentos de Péptidos/farmacología , Factores de Tiempo
13.
Biochem J ; 381(Pt 3): 779-94, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15104540

RESUMEN

Intrinsic tenase consists of activated Factors IX (IXa) and VIII (VIIIa) assembled on a negatively charged phospholipid surface. In vivo, this surface is mainly provided by activated platelets. In vitro, phosphatidylcholine/phosphatidylserine vesicles are often used to mimic natural pro-coagulant membranes. In the present study, we developed a quantitative mathematical model of Factor X activation by intrinsic tenase. We considered two situations, when complex assembly occurs on either the membrane of phospholipid vesicles or the surface of activated platelets. On the basis of existing experimental evidence, the following mechanism for the complex assembly on activated platelets was suggested: (i) Factors IXa, VIIIa and X bind to their specific platelet receptors; (ii) bound factors form complexes on the membrane: platelet-bound Factor VIIIa provides a high-affinity site for Factor X and platelet-bound Factor IXa provides a high-affinity site for Factor VIIIa; (iii) the enzyme-cofactor-substrate complex is assembled. This mechanism allowed the explanation of co-operative effects in the binding of Factors IXa, VIIIa and X to platelets. The model was reduced to obtain a single equation for the Factor X activation rate as a function of concentrations of Factors IXa, VIIIa, X and phospholipids (or platelets). The equation had a Michaelis-Menten form, where apparent V(max) and K(m) were functions of the factors' concentrations and the internal kinetic constants of the system. The equation obtained can be used in both experimental studies of intrinsic tenase and mathematical modelling of the coagulation cascade. The approach of the present study can be applied to research of other membrane-dependent enzymic reactions.


Asunto(s)
Factor IXa/metabolismo , Factor VIIIa/metabolismo , Factor X/metabolismo , Plaquetas/química , Plaquetas/enzimología , Membrana Celular/química , Cisteína Endopeptidasas/metabolismo , Factor IX/metabolismo , Humanos , Cinética , Liposomas/química , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Fosfatidilcolinas/química , Fosfatidilserinas/química , Activación Plaquetaria/fisiología , Valores de Referencia
14.
Trends Cardiovasc Med ; 12(7): 317-24, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12458095

RESUMEN

Atherosclerosis is a disease recognized as the main cause of death in industrial countries. The current paradigm establishes thrombosis to be the major reason for complications of atherosclerosis, such as myocardial infarction and stroke, and the major factor responsible for atherosclerosis-related mortality. Development of adequate treatment of patients with risk of atherothrombosis requires the comprehensive understanding of mechanisms underlying coagulation processes at the site of atherosclerotic lesion. The present review discusses contribution of the extrinsic and intrinsic pathways of blood coagulation in thrombogenicity of atherosclerotic plaque and factors determining the overall procoagulant/anticoagulant balance.


Asunto(s)
Arteriosclerosis/complicaciones , Arteriosclerosis/fisiopatología , Coagulación Sanguínea/fisiología , Trombosis/etiología , Trombosis/fisiopatología , Animales , Humanos
15.
Curr Gene Ther ; 3(1): 27-41, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12553533

RESUMEN

Hemophilia A, the most common inherited bleeding disorder, is caused by deficiency or functional defects in coagulation factor VIII (fVIII). Conventional treatment for this disease involves intravenous infusions of plasma-derived or recombinant fVIII products. Although replacement therapy effectively stops the bleeding episodes, it has a risk of transmission of viral blood-borne diseases and development of neutralizing antibodies that inactivate the administered fVIII protein. Hemophilia A is an attractive candidate for application of gene therapy approaches because the therapeutic window is wide and even modest elevation of fVIII levels will correct the hemophilic phenotype. Ongoing preclinical investigations utilize animal models of hemophilia A, including genetically fVIII-deficient mice and naturally fVIII-deficient dogs, to optimize vectors, transgenes and target cell populations for Phase I clinical trials. In this review, we outline the progress in understanding the mechanisms of fVIII turnover, which provides a basis for development of improved fVIII molecules with prolonged half-life in the circulation. We discuss the possibility of incorporating these improved fVIII molecules as transgenes into self-inactivating lentiviral vectors carrying chromatin insulator sequences, representing a new generation of gene delivery vehicle, to target hematopoietic stem cells and endothelial cells. The use of hematopoietic stem cells as the target cell population may prevent inhibitor formation to transduced fVIII by induction of immune tolerance. Alternatively, endothelial cells may support optimal synthesis of fVIII and myeloablative conditioning of patients with radiation or chemotherapy may not be required for efficient engraftment of the engineered cells. Collectively, these proposed advances represent promising prophylactic strategies toward long-term correction of the coagulation defect in this progressively debilitating, life-threatening disease.


Asunto(s)
Factor VIII/genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/terapia , Animales , Factor VIII/metabolismo , Humanos , Ratones , Transgenes
16.
Thromb Haemost ; 91(3): 619-25, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14983240

RESUMEN

Clotting factor VIII (fVIII)-inhibitory antibodies represent a major problem in the treatment of haemophilia A. To understand the inactivation mechanisms and to pave the way towards modifications of recombinant clotting factors that reduce their immunogenicity, the exact localization of immunodominant epitopes is required. Here, a random peptide phage display library was employed to identify epitopes of polyclonal fVIII antibodies isolated from patient's plasma by affinity chromatography. FVIII-binding specificity and inhibitory activity of the isolated fVIII antibodies were confirmed by ELISA and Bethesda assays. Phage selection on the individual samples yielded several phages which were displaced from binding to the respective antibody preparation by fVIII. Their homology with amino acid motifs of human fVIII and immunoprecipitation results with radioactively labelled fVIII fragments suggested putative epitopes in the A1, A2 and C1 domains of fVIII for one and in the C2 domain for another patient. Synthetic peptides corresponding to the A2, C1 and C2 domain epitopes blocked antibody binding to fVIII and partially neutralized the inhibitory activity of the respective plasma in Bethesda assays. These results provide the proof of principle that random peptide libraries can be used for the mapping of epitopes in a polyclonal antibody preparation.


Asunto(s)
Anticuerpos/química , Mapeo Epitopo/métodos , Factor VIII/química , Biblioteca de Péptidos , Secuencia de Aminoácidos , Aminoácidos/química , Preescolar , Cromatografía de Afinidad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Hemofilia A/terapia , Humanos , Lactante , Persona de Mediana Edad , Datos de Secuencia Molecular , Péptidos/química , Pruebas de Precipitina , Estructura Terciaria de Proteína , Radioinmunoensayo , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido
17.
Thromb Haemost ; 89(2): 235-42, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12574801

RESUMEN

Using time-lapse videomicroscopy, we studied the role of coagulation factor VIII (fVIII) in tissue factor-initiated spatial clot growth on fibroblast monolayers in a thin layer of non-stirred recalcified plasma from healthy donors or patients with severe Haemophilia A. Analysis of temporal evolution of light-scattering profiles from a growing clot revealed existence of two phases in the clot growth-initiation phase in a narrow (0.2 mm) zone adjacent to activator surface and elongation phase in plasma volume. While the initiation phase did not differ in normal and haemophilic plasmas, the rate of clot growth in the elongation phase in haemophilic plasma constituted only 30% of that in normal plasma. Supplementation of haemophilic plasma with 0.05 U/ml fVIII restored the normal clot growth rate (44.9 +/- 2.5 microm/min) at high but not at low fibroblast density. Our results indicate that the functioning of the intrinsic tenase complex is critical for normal spatial clot growth.


Asunto(s)
Coagulación Sanguínea/fisiología , Cisteína Endopeptidasas/fisiología , Factor VIII/fisiología , Hemofilia A/sangre , Proteínas de Neoplasias/fisiología , Recuento de Células , Células Cultivadas/citología , Relación Dosis-Respuesta a Droga , Factor VIII/administración & dosificación , Factor VIII/farmacología , Fibroblastos/citología , Humanos , Sustancias Macromoleculares , Nefelometría y Turbidimetría , Factores de Tiempo
18.
Thromb Haemost ; 88(5): 781-7, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12428094

RESUMEN

Thrombin-activated factor VIII (FVIIIa) is a heterotrimer with the A2 subunit in a weak ionic interaction with the A1 and A3-C1-C2 subunits. Dissociation of the A2 subunit correlates with inactivation of FVIIIa. A homology model (Blood 89:2413, 1997) of the triplicated A domains of factor VIII (FVIII) predicts a pseudo-threefold axis at the tightly packed hydrophobic core with several interdomain interactions. These lie at the interface of A1-A2, A2-A3 and A1-A3. We have previously demonstrated that hemophilia A mutations (R531H, A284E, S289L) within the predicted A1-A2 and A1-A3 interface disrupt potential intersubunit hydrogen bonds and have the molecular phenotype of increased rate of inactivation of FVIIIa due to increased rate of A2 subunit dissociation. Patients with these mutations exhibit a clinical phenotype where the FVIII activity by one-stage(1-st) assay is at least two-fold higher than by two-stage(2-st) assay. We have now also explored mutations within the predicted A2-A3 interface (N694I, R698W and R698L) that also have the phenotype of 1-st/2-st activity discrepancy. These mutations exhibit the same molecular mechanism of increased instability of FVIIIa as those mutations described along the A1-A2 and A1-A3 interfaces. This suggests that the entire tightly packed hydrophobic core within the predicted pseudo-threefold axis contributes to stabilization of FVIIIa.


Asunto(s)
Factor VIII/genética , Factor VIIIa/metabolismo , Hemofilia A/genética , Mutación Missense , Factor VIII/química , Factor VIIIa/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Mutagénesis Sitio-Dirigida , Fenotipo , Estructura Terciaria de Proteína , Subunidades de Proteína , Trombina/farmacología
19.
Thromb Haemost ; 87(3): 459-65, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11916079

RESUMEN

Factor VIII (FVIII) inhibitor alloantibodies react with the A2, C2, or A3-CI domains of FVIII and inactivate FVIII activity. We recently demonstrated that an anti-C2 monoclonal antibody with a Val2248-Gly2285 epitope, inhibited factor Xa (FXa)-catalyzed FVIII activation, and that a FXa binding site for FVIII was located within residues Thr2253-Gln2270. In this study, we investigated whether anti-C2 alloantibodies inhibit FXa-catalyzed FVIII activation. Anti-C2 alloantibodies from four patients inhibited FVIII activation by FXa in one-stage clotting assay. Furthermore, analysis by SDS-PAGE showed that all alloantibodies inhibited FVIII proteolytic cleavage by FXa independently of phospholipid. To confirm direct inhibition of FVIII and FXa interaction, we examined the effect of alloantibodies on FVIII binding to anhydro-FXa, a catalytically inactive FXa, in ELISA. All alloantibodies and C2-affinity purified F(ab)'2 preparations inhibited FVIII binding to anhydro-FXa dose-dependently. Our results revealed a new inhibitory mechanism of FVIII, mediated by inhibition of FXa in the presence of anti-C2 alloantibodies.


Asunto(s)
Epítopos/inmunología , Factor VIII/inmunología , Factor Xa/metabolismo , Isoanticuerpos/inmunología , Especificidad de Anticuerpos , Activación Enzimática/inmunología , Factor VIII/metabolismo , Hemofilia A/inmunología , Humanos , Fosfolípidos/farmacología , Unión Proteica/inmunología , Estructura Terciaria de Proteína
20.
Blood Coagul Fibrinolysis ; 15(2): 109-24, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15090997

RESUMEN

Factor VIII (FVIII) replacement therapy remains the mainstay in hemophilia A care. The major complication of replacement therapy is formation of antibodies, which inhibit FVIII activity, thus dramatically reducing treatment efficiency. The present review summarizes the accumulated knowledge on epitopes of FVIII inhibitors and mechanisms of their inhibitory effects. FVIII inhibitors most frequently target the A2, C2 and A3 domains of FVIII and interfere with important interactions of FVIII at various stages of its functional pathway; a class of FVIII inhibitors inactivates FVIII by proteolysis. We discuss therapeutic approaches currently used for treatment of hemophilia A patients with inhibitors and analyze the factors that influence the outcome. The choice between options should depend on the level of inhibitors and consideration of efficacy, safety, and availability of particular regimens. Advances of basic science open avenues for alternative targeted, specific and long-lasting treatments, such as the use of peptide decoys for blocking FVIII inhibitors, bypassing them with human/porcine FVIII hybrids, neutralizing FVIII-reactive CD4 T cells with anti-clonotypic antibodies, or inducing immune tolerance to FVIII with the use of universal CD4 epitopes or by genetic approaches.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/inmunología , Isoanticuerpos/inmunología , Animales , Anticuerpos Antiidiotipos/uso terapéutico , Anticuerpos Catalíticos/inmunología , Especificidad de Anticuerpos , Presentación de Antígeno , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Factores de Coagulación Sanguínea/uso terapéutico , Desensibilización Inmunológica , Mapeo Epitopo , Epítopos/inmunología , Factor VIII/antagonistas & inhibidores , Factor VIII/genética , Factor VIII/uso terapéutico , Terapia Genética , Hemofilia A/tratamiento farmacológico , Hemorragia/etiología , Hemorragia/prevención & control , Humanos , Tolerancia Inmunológica , Isoanticuerpos/biosíntesis , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/uso terapéutico , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA