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










Base de dados
Intervalo de ano de publicação
1.
Thromb Haemost ; 102(4): 645-55, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19806249

RESUMO

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.


Assuntos
Domínio Catalítico/genética , Fator VIII/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteína S/metabolismo , Proteínas Recombinantes/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Domínio Catalítico/imunologia , Fator VIII/genética , Fator VIII/imunologia , Humanos , Dados de Sequência Molecular , Complexos Multienzimáticos , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Ligação Proteica/imunologia , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/imunologia , Multimerização Proteica/genética , Proteína S/química , Proteína S/imunologia , Proteínas Recombinantes/genética , Ressonância de Plasmônio de Superfície
4.
J Biol Chem ; 284(11): 6934-45, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19126539

RESUMO

We have recently reported that plasmin likely associates with the factor VIII light chain to proteolyze at Lys36 within the A1 domain. In this study, we determined that the rate of plasmin-catalyzed inactivation on the forms of factor VIIIa containing A1-(1-336) and 1722A3C1C2, reflecting Lys36 cleavage, was reduced by approximately 60%, compared with those containing 1649A3C1C2 and 1690A3C1C2. SDS-PAGE analysis revealed that Lys36 cleavage of factor VIIIa with 1722A3C1C2 was markedly slower than those with 1649A3C1C2 and 1690A3C1C2. Surface plasmon resonance-based assays, using active site-modified anhydro-plasmin (Ah-plasmin) showed that 1722A3C1C2 bound to Ah-plasmin with an approximately 3-fold lower affinity than 1649A3C1C2 or 1690A3C1C2 (Kd, 176, 68.2, and 60.3 nM, respectively). Recombinant A3 bound to Ah-plasmin (Kd, 44.2 nM), whereas C2 failed to bind, confirming the presence of a plasmin-binding site within N terminus of A3. Furthermore, the Glu-Gly-Arg active site-modified factor IXa also blocked 1722A3C1C2 binding to Ah-plasmin by approximately 95%, supporting the presence of another plasmin-binding site overlapping the factor IXa-binding site in A3. In keeping with a major contribution of the lysine-binding sites in plasmin for interaction with the factor VIII light chain, analysis of the A3 sequence revealed two regions involving clustered lysine residues in 1690-1705 and 1804-1818. Two peptides based on these regions blocked 1649A3C1C2 binding to Ah-plasmin by approximately 60% and plasmin-catalyzed Lys36 cleavage of factor VIIIa with A1-(1-336) by approximately 80%. Our findings indicate that an extended surface, centered on residues 1690-1705 and 1804-1818 within the A3 domain, contributes to a unique plasmin-interactive site that promotes plasmin docking during cofactor inactivation by cleavage at Lys36.


Assuntos
Fator VIIIa/química , Fibrinolisina/química , Sítios de Ligação/fisiologia , Fator IXa/química , Fator IXa/genética , Fator IXa/metabolismo , Fator VIIIa/genética , Fator VIIIa/metabolismo , Fibrinolisina/genética , Fibrinolisina/metabolismo , Humanos , Lisina/química , Lisina/genética , Lisina/metabolismo , Mapeamento de Peptídeos , Estrutura Terciária de Proteína/fisiologia , Ressonância de Plasmônio de Superfície
5.
Semin Thromb Hemost ; 35(8): 735-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20169510

RESUMO

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.


Assuntos
Hemofilia A/tratamento farmacológico , Animais , Anticorpos Catalíticos/imunologia , Inibidores dos Fatores de Coagulação Sanguínea/genética , Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Fatores de Coagulação Sanguínea/uso terapêutico , Ensaio de Imunoadsorção Enzimática/métodos , Mapeamento de Epitopos/métodos , Epitopos/imunologia , Fator VIII/imunologia , Fator VIIa/uso terapêutico , Hemofilia A/diagnóstico , Hemofilia A/imunologia , Hemorragia/tratamento farmacológico , Humanos , Biblioteca de Peptídeos , Estrutura Terciária de Proteína/efeitos dos fármacos , Proteínas Recombinantes/uso terapêutico , Tromboelastografia
6.
Blood Coagul Fibrinolysis ; 19(8): 743-55, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19002040

RESUMO

Supraphysiological concentrations of recombinant activated factor VII (rVIIa, NovoSeven) are used to control bleeding in hemophilia. Current experimental evidence suggests that rVIIa may increase thrombin generation via two pathways: one being tissue factor (TF)-dependent and another being activated platelet-dependent. Contribution of TF to the rVIIa action may justify different administration profiles of rVIIa. In the present study, thrombin and fibrin generation and spatial clot formation assays in platelet-free hemophilia A and normal plasma were used to investigate this contribution. By varying the concentration of TF and the way it becomes available to plasma, we obtained the following results. Activation of clotting with less than 5 pmol/l of TF facilitates thrombin and fibrin generation at low, but not at supraphysiological rVIIa concentrations. Activation with more than 13 pmol/l of TF saturates thrombin and fibrin generation kinetics, making it insensitive to rVIIa. rVIIa minimally modulates clot growth on the surface of TF-expressing fibroblasts. On the contrary, rVIIa produces spontaneous clot formation in nonflowing platelet-free plasma far away from fibroblasts via plasma lipid particles. Therefore, both the concentration and the distribution of TF determine relevance of a particular experimental system for the studies of rVIIa action. The results indicate that 300-1600 nmol/l (megadoses) of rVIIa may deliver coagulation outside of the TF-rich areas of blood vessel damage via the platelet-derived microparticles. Therefore, rate and extent of platelet-derived microparticles generation might be important with regard to rVIIa treatment safety.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/fisiologia , Fator VIIa/farmacologia , Tromboplastina/farmacologia , Plaquetas/citologia , Micropartículas Derivadas de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Fibrina/metabolismo , Hemofilia A/sangue , Humanos , Cinética , Proteínas Recombinantes/farmacologia , Trombina/metabolismo
7.
Br J Haematol ; 143(3): 409-20, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18759761

RESUMO

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.


Assuntos
Cisteína Endopeptidases/metabolismo , Regulação para Baixo/efeitos dos fármacos , Fator VIIIa/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína S/farmacologia , Ligação Competitiva , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática/métodos , Fator IXa/metabolismo , Fator IXa/farmacologia , Fator X/metabolismo , Fator X/farmacologia , Humanos , Fosfolipídeos/metabolismo , Proteína C/fisiologia , Proteína S/metabolismo , Proteínas Recombinantes/metabolismo , Fator de von Willebrand/metabolismo
8.
Blood Coagul Fibrinolysis ; 19(6): 543-55, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18685438

RESUMO

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.


Assuntos
Fator VIII/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Receptores de LDL/metabolismo , Substituição de Aminoácidos , Aminoácidos/fisiologia , Sítios de Ligação , Sequência Consenso , Fator VIII/química , Humanos , Modelos Moleculares , Mutação de Sentido Incorreto , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Ressonância de Plasmônio de Superfície , Trombina/farmacologia , Fator de von Willebrand/metabolismo
9.
Biochim Biophys Acta ; 1784(5): 753-63, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18329389

RESUMO

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.


Assuntos
Fator VIII/química , Fator VIII/metabolismo , Fibrinolisina/metabolismo , Mapeamento de Interação de Proteínas , Ácido Aminocaproico/farmacologia , Anticorpos/farmacologia , Sítios de Ligação , Catálise/efeitos dos fármacos , Fator IXa/metabolismo , Humanos , Cinética , Proteínas Mutantes/metabolismo , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Subunidades Proteicas/metabolismo , Ressonância de Plasmônio de Superfície
10.
Blood Coagul Fibrinolysis ; 19(2): 166-77, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18277139

RESUMO

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.


Assuntos
Fator VIIIa/fisiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Receptores de LDL/fisiologia , Sítios de Ligação/fisiologia , Coagulação Sanguínea/fisiologia , Células Cultivadas , Cisteína Endopeptidases/fisiologia , Fator VIIIa/química , Humanos , Lipoproteínas VLDL , Proteínas de Neoplasias/fisiologia , Ressonância de Plasmônio de Superfície
11.
Haematologica ; 93(1): 83-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18166789

RESUMO

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.


Assuntos
Antígenos CD/biossíntese , Células Dendríticas/citologia , Fator VIII/biossíntese , Linfócitos T/citologia , Animais , Separação Celular , Endocitose , Fator VIII/metabolismo , Hemofilia A/genética , Humanos , Leucócitos Mononucleares/citologia , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Ativação Linfocitária , Camundongos , Monócitos/citologia , Monócitos/metabolismo
12.
Br J Haematol ; 140(4): 433-43, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18081893

RESUMO

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).


Assuntos
Fator VIII/química , Trombina/química , Anticorpos Monoclonais/imunologia , Sítios de Ligação , Ligação Competitiva , Catálise , Eletroforese em Gel de Poliacrilamida/métodos , Fator VIII/imunologia , Humanos , Isoanticorpos , Fragmentos de Peptídeos , Ligação Proteica , Proteínas Recombinantes/química
13.
Thromb Haemost ; 98(6): 1170-81, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18064310

RESUMO

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.


Assuntos
Fator VIII/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Ligação Competitiva , Linhagem Celular , Endocitose , Fator VIII/química , Fator VIII/genética , Fibroblastos/metabolismo , Humanos , Radioisótopos do Iodo , Proteínas Relacionadas a Receptor de LDL/química , Proteínas Relacionadas a Receptor de LDL/genética , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Chaperonas Moleculares/metabolismo , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Mutação Puntual , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Fatores de Tempo
14.
J Med Chem ; 50(18): 4329-39, 2007 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-17691762

RESUMO

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.


Assuntos
Fator VIII/isolamento & purificação , Ácidos Indolacéticos/síntese química , Oligopeptídeos/química , Substituição de Aminoácidos , Cromatografia de Afinidade/métodos , Estabilidade de Medicamentos , Fator VIII/química , Humanos , Ácidos Indolacéticos/química , Ligantes , Mimetismo Molecular , Oligopeptídeos/síntese química , Peptídeo Hidrolases/química , Polímeros , Ligação Proteica , Proteínas Recombinantes/química , Soro , Estereoisomerismo
15.
Blood Coagul Fibrinolysis ; 17(5): 379-88, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16788314

RESUMO

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.


Assuntos
Fator VIII/fisiologia , Fator VIIIa/metabolismo , Fator Xa/farmacologia , Fragmentos de Peptídeos/fisiologia , Proteína C/farmacologia , Western Blotting , Humanos , Fragmentos de Peptídeos/farmacologia , Fatores de Tempo
16.
Biochemistry ; 45(6): 1829-40, 2006 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-16460029

RESUMO

Regulation of the coagulation factor VIII (fVIII) level in circulation involves a hepatic receptor low-density lipoprotein receptor-related protein (LRP). One of two major LRP binding sites in fVIII is located within the A2 domain (A2), likely exposed within the fVIII complex with von Willebrand factor and contributing to regulation of fVIII via LRP. This work aimed to identify A2 residues forming its LRP-binding site, previously shown to involve residues 484-509. Isolated A2 was subjected to alanine-scanning mutagenesis followed by expression of a set of mutants in a baculovirus system. In competition and surface plasmon resonance assays, affinities of A2 mutants K466A, R471A, R484A, S488A, R489A, R490A, H497A, and K499A for LRP were found to be decreased by 2-4-fold. This correlated with 1.3-1.5-fold decreases in the degree of LRP-mediated internalization of the mutants in cell culture. Combining these mutations into pairs led to cumulative effects, i.e., 7-13-fold decrease in affinity for LRP and 1.6-2.2-fold decrease in the degree of LRP-mediated internalization in cell culture. We conclude that the residues mentioned above play a key role in formation of the A2 binding epitope for LRP. Experiments in mice revealed an approximately 4.5 times shorter half-life for A2 in the circulation in comparison with that of fVIII. The half-lives of A2 mutant R471A/R484A or A2 co-injected with receptor-associated protein, a classical ligand of LRP, were prolonged by approximately 1.9 and approximately 3.5 times, respectively, compared to that of A2. This further confirms the importance of the mutated residues for interaction of A2 with LRP and suggests the existence of an LRP-dependent mechanism for removing A2 as a product of dissociation of activated fVIII from the circulation.


Assuntos
Epitopos/metabolismo , Fator VIII/metabolismo , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Alanina/genética , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Sequência de Bases , Sítios de Ligação , Membrana Celular/metabolismo , Células Cultivadas , Epitopos/genética , Fator VIII/química , Fator VIII/genética , Fator VIII/isolamento & purificação , Meia-Vida , Fígado/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Receptores Imunológicos/metabolismo , Ressonância de Plasmônio de Superfície
17.
FEBS J ; 273(2): 374-87, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16403024

RESUMO

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.


Assuntos
Fator VIIIa/fisiologia , Fator Intrínseco/metabolismo , Ativação Enzimática , Fator VIIIa/metabolismo , Fator X/metabolismo , Humanos , Cinética , Fosfolipídeos/metabolismo , Ligação Proteica
18.
Biophys J ; 90(5): 1489-500, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16326897

RESUMO

Blood coagulation in vivo is a spatially nonuniform, multistage process: coagulation factors from plasma bind to tissue factor (TF)-expressing cells, become activated, dissociate, and diffuse into plasma to form enzymatic complexes on the membranes of activated platelets. We studied spatial regulation of coagulation using two approaches: 1), an in vitro experimental model of clot formation in a thin layer of plasma activated by a monolayer of TF-expressing cells; and 2), a computer simulation model. Clotting in factor VIII- and factor XI-deficient plasmas was initiated normally, but further clot elongation was impaired in factor VIII- and, at later stages, in factor XI-deficient plasma. The data indicated that clot elongation was regulated by factor Xa formation by intrinsic tenase, whereas factor IXa was formed by extrinsic tenase on activating cells and diffused into plasma, thus sustaining clot growth. Far from the activating cells, additional factor IXa was produced by factor XIa. Exogenously added TF had no effect on the clot growth rate, suggesting that plasma TF does not contribute significantly to the clot propagation process in a reaction-diffusion system without flow. Addition of thrombomodulin at 3-100 nM caused dose-dependent termination of clot elongation with a final clot size of 2-0.2 mm. These results identify roles of specific coagulation pathways at different stages of spatial clot formation (initiation, elongation, and termination) and provide a possible basis for their therapeutic targeting.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/citologia , Plaquetas/fisiologia , Modelos Biológicos , Proteína C/metabolismo , Transdução de Sinais/fisiologia , Tromboplastina/metabolismo , Fatores de Coagulação Sanguínea/metabolismo , Células Cultivadas , Simulação por Computador , Humanos
19.
J Lipid Res ; 46(8): 1721-31, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15863833

RESUMO

Apolipoprotein E (apoE) associates with lipoproteins and mediates their interaction with members of the LDL receptor family. ApoE exists as three common isoforms that have important distinct functional and biological properties. Two apoE isoforms, apoE3 and apoE4, are recognized by the LDL receptor, whereas apoE2 binds poorly to this receptor and is associated with type III hyperlipidemia. In addition, the apoE4 isoform is associated with the common late-onset familial and sporadic forms of Alzheimer's disease. Although the interaction of apoE with the LDL receptor is well characterized, the specificity of other members of this receptor family for apoE is poorly understood. In the current investigation, we have characterized the binding of apoE to the VLDL receptor and the LDL receptor-related protein (LRP). Our results indicate that like the LDL receptor, LRP prefers lipid-bound forms of apoE, but in contrast to the LDL receptor, both LRP and the VLDL receptor recognize all apoE isoforms. Interestingly, the VLDL receptor does not require the association of apoE with lipid for optimal recognition and avidly binds lipid-free apoE. It is likely that this receptor-dependent specificity for various apoE isoforms and for lipid-free versus lipid-bound forms of apoE is physiologically significant and is connected to distinct functions for these receptors.


Assuntos
Apolipoproteínas E/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Receptores de LDL/metabolismo , Apolipoproteína E3 , Apolipoproteína E4 , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Humanos , Metabolismo dos Lipídeos , Fragmentos de Peptídeos/metabolismo , Mapeamento de Interação de Proteínas , Ressonância de Plasmônio de Superfície
20.
Biochem J ; 381(Pt 3): 779-94, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15104540

RESUMO

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.


Assuntos
Fator IXa/metabolismo , Fator VIIIa/metabolismo , Fator X/metabolismo , Plaquetas/química , Plaquetas/enzimologia , Membrana Celular/química , Cisteína Endopeptidases/metabolismo , Fator IX/metabolismo , Humanos , Cinética , Lipossomos/química , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Fosfatidilcolinas/química , Fosfatidilserinas/química , Ativação Plaquetária/fisiologia , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...