Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 133(10): 1152-1163, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30591525

RESUMO

The plasma proteins factor XII (FXII) and prekallikrein (PK) undergo reciprocal activation to the proteases FXIIa and kallikrein by a process that is enhanced by surfaces (contact activation) and regulated by the serpin C1 inhibitor. Kallikrein cleaves high-molecular-weight kininogen (HK), releasing the vasoactive peptide bradykinin. Patients with hereditary angioedema (HAE) experience episodes of soft tissue swelling as a consequence of unregulated kallikrein activity or increased prekallikrein activation. Although most HAE cases are caused by reduced plasma C1-inhibitor activity, HAE has been linked to lysine/arginine substitutions for Thr309 in FXII (FXII-Lys/Arg309). Here, we show that FXII-Lys/Arg309 is susceptible to cleavage after residue 309 by coagulation proteases (thrombin and FXIa), resulting in generation of a truncated form of FXII (δFXII). The catalytic efficiency of δFXII activation by kallikrein is 15-fold greater than for full-length FXII. The enhanced rate of reciprocal activation of PK and δFXII in human plasma and in mice appears to overwhelm the normal inhibitory function of C1 inhibitor, leading to increased HK cleavage. In mice given human FXII-Lys/Arg309, induction of thrombin generation by infusion of tissue factor results in enhanced HK cleavage as a consequence of δFXII formation. The effects of δFXII in vitro and in vivo are reproduced when wild-type FXII is bound by an antibody to the FXII heavy chain (HC; 15H8). The results contribute to our understanding of the predisposition of patients carrying FXII-Lys/Arg309 to angioedema after trauma, and reveal a regulatory function for the FXII HC that normally limits PK activation in plasma.


Assuntos
Fator XII/química , Fator XIa/química , Angioedema Hereditário Tipo III/sangue , Angioedema Hereditário Tipo III/genética , Angioedemas Hereditários , Animais , Arginina/química , Coagulação Sanguínea , Bradicinina/sangue , Catálise , Proteína Inibidora do Complemento C1/química , Fator XIIa/química , Células HEK293 , Humanos , Cininogênios/sangue , Lisina/química , Camundongos , Camundongos Endogâmicos C57BL , Calicreína Plasmática/química , Pré-Calicreína/química , Ligação Proteica , Proteínas Recombinantes/química , Propriedades de Superfície , Trombina/genética
2.
Glycobiology ; 28(10): 754-764, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30016441

RESUMO

Plasma contact system is the initial part of both the intrinsic coagulation pathway and kallikrein-kinin pathway, which mainly involves three proteins: coagulation factor XII (FXII), prekallikrein (PK) and high-molecular weight kininogen. Fucosylated chondroitin sulfate (FCS) is a unique sulfated glycosaminoglycan (GAG) composed of a chondroitin sulfate-like backbone and sulfated fucose branches. The native FCS was preliminary found to cause undesired activation of the plasma contact system. How this unusual GAG functions in this process remains to be clarified. Herein, the relationship between its structure, plasma contact activation and its effects on the PK-FXII reciprocal activation loop were studied. The recalcification time assay indicated that the FCS at high concentration could be procoagulant which may be attributed to its contact activation activity. The structure-activity relationship study indicated that its high molecular weight and distinct fucose side chains are required for contact activation by FCS, although the sulfate substitution types of its side chains have less impact. In human plasma, the native FCSs potently induced FXII-dependent contact activation. However, in purified systems FCS did not significantly activate FXII per se or induce its autoactivation, whereas FCS significantly promoted the activation of PK by factor XIIa. Polysaccharide-protein interaction assays showed that FCS bound to PK with higher affinity than other contact system proteins. These data suggested that potent contact activation by FCS requires the positive feedback loop between PK and FXII. These findings contribute to better understanding of contact activation by complex GAG.


Assuntos
Sulfatos de Condroitina/sangue , Sulfatos de Condroitina/metabolismo , Fator XIIa/metabolismo , Cininogênios/metabolismo , Pré-Calicreína/metabolismo , Sulfatos de Condroitina/química , Fator XIIa/química , Humanos , Cininogênios/química , Pré-Calicreína/química , Relação Estrutura-Atividade
3.
J Thromb Haemost ; 12(9): 1513-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25039405

RESUMO

BACKGROUND: The mechanism underpinning factor XII autoactivation was originally characterized with non-physiological surfaces, such as dextran sulfate (DS), ellagic acid, and kaolin. Several 'natural' anionic activating surfaces, such as platelet polyphosphate (polyP), have now been identified. OBJECTIVE: To analyze the autoactivation of FXII by polyP of a similar length to that found in platelets (polyP70 ). METHODS AND RESULTS: PolyP70 showed similar efficacy to DS in stimulating autoactivation of FXII, as detected with amidolytic substrate. Western blotting revealed different forms of FXII with the two activating surfaces: two-chain αFXIIa was formed with DS, whereas single-chain FXII (scFXII; 80 kDa) was formed with polyP70 . Dissociation of scFXII from polyP70 abrogated amidolytic activity, suggesting reversible exposure of the active site. Activity of scFXII-polyP70 was enhanced by Zn(2+) and was sensitive to NaCl concentration. A bell-shaped concentration response to polyP70 was evident, as is typical of surface-mediated reactions. Reaction of scFXII-polyP70 with various concentrations of S2302 generated a sigmoidal curve, in contrast to a hyperbolic curve for αFXIIa, from which a Hill coefficient of 3.67 was derived, indicative of positive cooperative binding. scFXII-polyP70 was more sensitive to inhibition by H-d-Pro-Phe-Arg-chloromethylketone and corn trypsin inhibitor than αFXIIa, but inhibition profiles for C1-inhibitor were similar. Active scFXII-polyP70 was also able to cleave its physiological targets FXI and prekallikrein to their active forms. CONCLUSIONS: Autoactivation of FXII by polyP, of the size found in platelets, proceeds via an active single-chain intermediate. scFXII-polyP70 shows activity towards physiological substrates, and may represent the primary event in initiating contact activation in vivo.


Assuntos
Fator XII/química , Polifosfatos/química , Clorometilcetonas de Aminoácidos/química , Ânions/química , Arginina/química , Plaquetas/metabolismo , Domínio Catalítico , Sulfato de Dextrana/química , Dissulfetos/química , Ácido Elágico/química , Precursores Enzimáticos/química , Hemostasia , Humanos , Caulim/química , Proteínas de Plantas/química , Pré-Calicreína/química , Ligação Proteica , RNA/química , Propriedades de Superfície
4.
PLoS One ; 7(11): e50399, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226277

RESUMO

Nucleic acids, especially extracellular RNA, are exposed following tissue- or vessel damage and have previously been shown to activate the intrinsic blood coagulation pathway in vitro and in vivo. Yet, no information on structural requirements for the procoagulant activity of nucleic acids is available. A comparison of linear and hairpin-forming RNA- and DNA-oligomers revealed that all tested oligomers forming a stable hairpin structure were protected from degradation in human plasma. In contrast to linear nucleic acids, hairpin forming compounds demonstrated highest procoagulant activities based on the analysis of clotting time in human plasma and in a prekallikrein activation assay. Moreover, the procoagulant activities of the DNA-oligomers correlated well with their binding affinity to high molecular weight kininogen, whereas the binding affinity of all tested oligomers to prekallikrein was low. Furthermore, four DNA-aptamers directed against thrombin, activated protein C, vascular endothelial growth factor and nucleolin as well as the naturally occurring small nucleolar RNA U6snRNA were identified as effective cofactors for prekallikrein auto-activation. Together, we conclude that hairpin-forming nucleic acids are most effective in promoting procoagulant activities, largely mediated by their specific binding to kininogen. Thus, in vivo application of therapeutic nucleic acids like aptamers might have undesired prothrombotic or proinflammatory side effects.


Assuntos
Aptâmeros de Nucleotídeos/química , Coagulação Sanguínea , Coagulantes/química , Sequências Repetidas Invertidas , Cininogênio de Alto Peso Molecular/química , Oligorribonucleotídeos/química , Testes de Coagulação Sanguínea , Humanos , Conformação de Ácido Nucleico , Fosfoproteínas/química , Pré-Calicreína/química , Ligação Proteica , Proteína C/química , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/genética , Proteínas de Ligação a RNA/química , Relação Estrutura-Atividade , Trombina/química , Fator A de Crescimento do Endotélio Vascular/química , Nucleolina
5.
Blood Coagul Fibrinolysis ; 23(8): 781-3, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22964768

RESUMO

Diagnostic ultrasound activates the contact phase of human coagulation. This has been seen in human blood or plasma or with purified factor 12. The present work aimed to quantify a possibly triggering action of ultrasound on purified prekallikrein, the second of the two main triggers of the intrinsic hemostasis cascade. Either 2.7 µg/ml human prekallikrein or for control 1 µg/ml kallikrein in 26% glycerol - 0.54% NaCl-10.6 mmol/l Na3 citrate pH 7.4, in emptied polypropylene coagulation monovettes (Sarstedt) were exposed to diagnostic ultrasound (Siemens Acouson Antares, 5 MHz, 0.6 TIB, 0.6 TIS) for 0-5 min at room temperature (RT). Fifty microliter samples were withdrawn in duplicate and placed into an U-wells high quality microtiter plate (Brand 781600). Then 10 µl 2 mmol/l chromogenic substrate HD-CHG-Ala-Arg-pNA in 0.45% NaCl were added, and the increase in absorbance with time (ΔA405 nm /t at 37°C) was determined by a microtiterplate photometer with a 1 mA resolution (PHOmo; anthos). Exposure to diagnostic ultrasound biphasically increased the chromogenic activity of a prekallikrein solution in 26% glycerol. About 3-4 min ultrasound at 23 °C generated about 0.02 µg/ml kallikrein, that means that about 1% of pure prekallikrein in glycerol was converted into kallikrein. Thus, diagnostic ultrasound activates purified human prekallikrein to kallikrein. The ultrasound energy seems to fold the latent proenzyme prekallikrein into the active enzyme kallikrein. This contributes to explain the triggering action of ultrasound on the contact system of plasmatic human coagulation. Conversion of only 1% of prekallikrein into kallikrein is absolutely sufficient to start the intrinsic coagulation cascade. The clinical consequence of this action of ultrasound on intrinsic coagulation is that patients at risk for thrombosis, for example, patients with insufficiencies of hepatocytes, AT-3, C1-ina, or fibrinolysis should be protected by low-molecular-weight-heparin prior to the exposure of ultrasound, especially upon its prolonged exposure.


Assuntos
Pré-Calicreína/efeitos da radiação , Som/efeitos adversos , Testes de Coagulação Sanguínea , Soluções Tampão , Compostos Cromogênicos/química , Colorimetria , Glicerol , Humanos , Calicreínas/química , Calicreínas/efeitos da radiação , Oligopeptídeos/química , Pré-Calicreína/química , Dobramento de Proteína , Ultrassonografia/efeitos adversos
6.
Anal Biochem ; 390(1): 88-90, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19351526

RESUMO

A simple methodology for the identification of hemostatic proteins that are subjected to posttranslational tyrosine sulfation was developed. The procedure involves sequence analysis of members of the three hemostatic pathways using the Sulfinator prediction algorithm, followed by [(35)S]sulfate labeling of cultured HepG2 human hepatoma cells, immunoprecipitation of targeted [(35)S]sulfate-labeled hemostatic proteins, and tyrosine O-[(35)S]sulfate analysis of immunoprecipitated proteins. Three new tyrosine-sulfated hemostatic proteins-protein S, prekallikrein, and plasminogen-were identified. Such a target-specific approach will allow investigation of tyrosine-sulfated proteins of other biochemical/physiological pathways/processes and contribute to a better understanding of the functional role of posttranslational tyrosine sulfation.


Assuntos
Eletroforese em Gel Bidimensional/métodos , Plasminogênio/química , Pré-Calicreína/química , Proteína S/química , Sulfatos/química , Tirosina/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Isótopos de Enxofre/química
7.
Biochemistry ; 46(50): 14450-60, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18020374

RESUMO

The zymogen, factor XI, and the enzyme, factor XIa, interact specifically with functional receptors on the surface of activated platelets. These studies were initiated to identify the molecular subdomain within factor XIa that binds to activated platelets. Both factor XIa (Ki approximately 1.4 nM) and a chimeric factor XIa containing the Apple 3 domain of prekallikrein (Ki approximately 2.7 nM) competed with [125I]factor XIa for binding sites on activated platelets, suggesting that the factor XIa binding site for platelets is not located in the Apple 3 domain which mediates factor XI binding to platelets. The recombinant catalytic domain (Ile370-Val607) inhibited the binding of [125I]factor XIa to the platelets (Ki approximately 3.5 nM), whereas the recombinant factor XI heavy chain did not, demonstrating that the platelet binding site is located in the light chain of factor XIa. A conformationally constrained cyclic peptide (Cys527-Cys542) containing a high-affinity (KD approximately 86 nM) heparin-binding site within the catalytic domain of factor XIa also displaced [125I]factor XIa from the surface of activated platelets (Ki approximately 5.8 nM), whereas a scrambled peptide of identical composition was without effect, suggesting that the binding site in factor XIa that interacts with the platelet surface resides in the catalytic domain near the heparin binding site of factor XIa. These data support the conclusion that a conformational transition accompanies conversion of factor XI to factor XIa that conceals the Apple 3 domain factor XI (zymogen) platelet binding site and exposes the factor XIa (enzyme) platelet binding site within the catalytic domain possibly comprising residues Cys527-Cys542.


Assuntos
Plaquetas/metabolismo , Fator XIa/química , Fator XIa/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Catálise , Linhagem Celular , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Ensaio de Imunoadsorção Enzimática , Fator XI/química , Fator XI/genética , Fator XI/metabolismo , Fator XIIa/química , Fator XIIa/genética , Fator XIIa/metabolismo , Fator XIa/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Pré-Calicreína/química , Pré-Calicreína/genética , Pré-Calicreína/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
8.
J Thromb Haemost ; 3(5): 969-75, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15869593

RESUMO

Phospholipid binding proteins, including factor XII (FXII), are known to be targeted by antiphospholipid antibodies (aPA). Factor XII antibodies (FXIIab) have been described in some patients with the antiphospholipid syndrome (APS) and have been shown to lead to reduced levels of FXII. The antigenic binding site(s) and the pathophysiological effects of FXIIab are unknown. In an attempt to elucidate the binding site of these antibodies, immobilized plasma kallikrein was used to cleave FXII into its 52-kDa heavy-chain (HCFXII) and 28-kDa light-chain (LCFXII) components. Plasma samples from 12 female patients with definite APS and FXIIab were investigated for the presence of antibodies to FXII, HCFXII and LCFXII. All but one patient's plasma reacted to FXII, HCFXII and LCFXII in a similar manner. One patient gave markedly reduced positivity to HCFXII and LCFXII, suggesting that the FXIIab in this patient had a higher affinity for the intact FXII molecule. To further investigate the antigenic binding site(s) of FXII, 150 biotinylated peptides of the known FXII sequence were synthesized using a Multipin(TM) peptide synthesis procedure. The IgG and IgM fractions of the 12 patients' plasma were purified by affinity chromatography. The synthesized peptides were captured on streptavidin plates and individual patients' purified FXIIab assayed against the peptides in a modified enzyme-linked immunosorbent assay (ELISA). Two regions were identified as possible antigenic binding site(s) for FXIIab: one in the growth factor domain and the other in the catalytic domain.


Assuntos
Anticorpos/química , Síndrome Antifosfolipídica/imunologia , Fator XII/química , Fator XII/imunologia , Sequência de Aminoácidos , Antígenos/química , Síndrome Antifosfolipídica/metabolismo , Sítios de Ligação , Biotinilação , Domínio Catalítico , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/química , Imunoglobulina M/química , Substâncias Macromoleculares/química , Dados de Sequência Molecular , Complexos Multiproteicos/química , Peptídeos/química , Pré-Calicreína/química , Conformação Proteica , Reprodutibilidade dos Testes , Coloração pela Prata
9.
Blood ; 104(1): 128-34, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15026311

RESUMO

The bleeding diathesis associated with hereditary factor XI (fXI) deficiency is prevalent in Ashkenazi Jews, in whom the disorder appears to be an autosomal recessive condition. The homodimeric structure of fXI implies that the product of a single mutant allele could confer disease in a dominant manner through formation of heterodimers with wild-type polypeptide. We studied 2 unrelated patients with fXI levels less than 20% of normal and family histories indicating dominant disease transmission. Both are heterozygous for single amino acid substitutions in the fXI catalytic domain (Gly400Val and Trp569Ser). Neither mutant is secreted by transfected fibroblasts. In cotransfection experiments with a wild-type fXI construct, constructs with mutations common in Ashkenazi Jews (Glu117Stop and Phe283Leu) and a variant with a severe defect in dimer formation (fXI-Gly350Glu) have little effect on wild-type fXI secretion. In contrast, cotransfection with fXI-Gly400Val or fXI-Trp569Ser reduces wild-type secretion about 50%, consistent with a dominant negative effect. Immunoprecipitation of cell lysates confirmed that fXI-Gly400Val forms intracellular dimers. The data support a model in which nonsecretable mutant fXI polypeptides trap wild-type polypeptides within cells through heterodimer formation, resulting in lower plasma fXI levels than in heterozygotes for mutations that cause autosomal recessive fXI deficiency.


Assuntos
Deficiência do Fator XI/genética , Fator XI/genética , Adulto , Substituição de Aminoácidos , Animais , Domínio Catalítico , Linhagem Celular , Besouros/enzimologia , Dimerização , Fator XI/química , Fator XI/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Genes Dominantes , Heterozigoto , Humanos , Espaço Intracelular/metabolismo , Luciferases/genética , Luciferases/metabolismo , Masculino , Pessoa de Meia-Idade , Linhagem , Pré-Calicreína/química , Pré-Calicreína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
10.
J Thromb Haemost ; 1(11): 2340-7, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14629467

RESUMO

The coagulation protease zymogen factor (F)XI is a disulfide bond-linked homodimer, a configuration that is necessary for protein secretion and function. The non-catalytic portion of the FXI polypeptide contains four repeats called apple domains (A1-A4). It is clear that FXI A4 plays a key role in dimer formation, however, the importance of other apple domains to this process has not been examined. We prepared recombinant FXI molecules in which apple domains were exchanged with those of the structurally homologous monomeric protein prekallikrein (PK). As expected, FXI/PK chimeras containing FXI A4 are dimers, while those with PK A4 are monomers. FXI A4 contains cysteine at position 321 that forms the interchain disulfide bond, while Cys321 in PK is unavailable for interchain bond formation because it is paired with Cys326. FXI/PK chimeras containing PK A4 were modified by changing Cys326 to glycine, leaving Cys321 unpaired (PKA4-Gly326). FXI with a PK A4 domain is a monomer, however, introducing PKA4-Gly326 results in a disulfide bond-linked dimer. This indicates that dimer formation can occur in the absence of FXI A4. In proteins containing PKA4-Gly326, replacing FXI A3 with PK A3 partially interferes with dimer formation, while substitution of A2, or A2 and A3 prevents dimer formation. PKA4-Gly326 cannot induce the native PK molecule to dimerize. The data indicate that FXI A2 and A3 make contributions to dimer formation. As these domains are involved in activities that require dimeric protein, it seems reasonable that they stabilize this conformation.


Assuntos
Fator IX/química , Sítios de Ligação , Cromatografia em Gel , Clonagem Molecular , Dimerização , Dissulfetos , Eletroforese em Gel de Poliacrilamida , Fator IX/genética , Humanos , Pré-Calicreína/química , Pré-Calicreína/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão
11.
Biochem J ; 367(Pt 1): 49-56, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12084014

RESUMO

Human factor XI, a plasma glycoprotein required for normal haemostasis, is a homodimer (160 kDa) formed by a single interchain disulphide bond linking the Cys-321 of each Apple 4 domain. Bovine, porcine and murine factor XI are also disulphide-linked homodimers. Rabbit factor XI, however, is an 80 kDa polypeptide on non-reducing SDS/PAGE, suggesting that rabbit factor XI exists and functions physiologically either as a monomer, as does prekallikrein, a structural homologue to factor XI, or as a non-covalent homodimer. We have investigated the structure and function of rabbit factor XI to gain insight into the relation between homodimeric structure and factor XI function. Characterization of the cDNA sequence of rabbit factor XI and its amino acid translation revealed that in the rabbit protein a His residue replaces the Cys-321 that forms the interchain disulphide linkage in human factor XI, explaining why rabbit factor XI is a monomer in non-reducing SDS/PAGE. On size-exclusion chromatography, however, purified plasma rabbit factor XI, like the human protein and unlike prekallikrein, eluted as a dimer, demonstrating that rabbit factor XI circulates as a non-covalent dimer. In functional assays rabbit factor XI and human factor XI behaved similarly. Both monomeric and dimeric factor XI were detected in extracts of cells expressing rabbit factor XI. We conclude that the failure of rabbit factor XI to form a covalent homodimer due to the replacement of Cys-321 with His does not impair its functional activity because it exists in plasma as a non-covalent homodimer and homodimerization is an intracellular process.


Assuntos
Fator XI/química , Fator XI/genética , Sequência de Aminoácidos , Animais , Cromatografia , Clonagem Molecular , Cisteína/química , DNA Complementar/metabolismo , Dimerização , Eletroforese em Gel de Poliacrilamida , Humanos , Cinética , Dados de Sequência Molecular , Pré-Calicreína/química , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 99(2): 896-900, 2002 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11792853

RESUMO

Bradykinin is a major mediator of swelling in C1 inhibitor deficiency as well as the angioedema seen with ACE inhibitors and may contribute to bronchial hyperreactivity in asthma. Formation of bradykinin occurs in the fluid phase and along cell surfaces requiring interaction of factor XII, prekallikrein, and high M(r) kininogen (HK). Recent data suggest that activation of the kinin-forming cascade can occur on the surface of endothelial cells, even in the absence of factor XII. We sought to further define this factor XII-independent mechanism of kinin formation. Both cytosolic and membrane fractions from endothelial cells possessed the ability to catalyze prekallikrein conversion to kallikrein, and activation depended on the presence of HK and zinc ion. We fractionated the cytosol by ion exchange chromatography and affinity chromatography by using corn trypsin inhibitor as ligand. The fractions with peak activity were subjected to SDS gel electrophoresis and ligand blot with biotinylated corn trypsin inhibitor, and positive bands were sequenced. Heat shock protein 90 (Hsp90) was identified as the protein responsible for zinc-dependent prekallikrein activation in the presence of HK. Zinc-dependent activation of the prekallikrein-HK complex also depended on addition of either alpha and beta isoforms of Hsp90 and the activation on endothelial cells was inhibited on addition of polyclonal Ab to Hsp90 in a dose-dependent manner. Although the mechanism by which Hsp90 activates the kinin-forming cascade is not understood, this protein represents the cellular contribution to the reaction and may become the dominant mechanism in pathologic circumstances in which Hsp90 is highly expressed or secreted.


Assuntos
Fator XII/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Cininogênios/metabolismo , Pré-Calicreína/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Endotélio Vascular/metabolismo , Proteínas de Choque Térmico HSP90/química , Humanos , Cinética , Cininogênios/química , Substâncias Macromoleculares , Pré-Calicreína/química
13.
Vopr Med Khim ; 47(1): 20-42, 2001.
Artigo em Russo | MEDLINE | ID: mdl-11385996

RESUMO

The kallikrein-kinin system (KKS) is the key proteolytic system participating in control of a wide spectrum of physiological functions and the development of many pathological conditions. This explains great interest in structures, functions and molecular biology of separate components of the system, molecular mechanisms of their interaction and relationship with other regulatory systems. The information in this field for the last two decades clarifies the role of KKS in morphogenesis of cells, regulation of smooth muscular contractility of some organs, decrease of blood pressure, increase of vascular permeability, the development of inflammation, transformation of cells and the other functions of both physiological and pathological processes. Essential progress in understanding of functions KKS was made by the discovery and study of bradykinin receptors, cloning of kininogen and kallikrein encoding genes, revealing of domain structure of kininogen, prekallikrein and some kininase and decoding of mechanisms of contact phase of proteolytic system activation in blood plasma.


Assuntos
Sistema Calicreína-Cinina/fisiologia , Sequência de Aminoácidos , Humanos , Cininogênios/sangue , Dados de Sequência Molecular , Pré-Calicreína/química , Receptor B1 da Bradicinina , Receptores da Bradicinina/fisiologia
14.
Blood ; 97(10): 3117-22, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11342438

RESUMO

Human coagulation factor XI (FXI) is a plasma serine protease composed of 2 identical 80-kd polypeptides connected by a disulfide bond. This dimeric structure is unique among blood coagulation enzymes. The hypothesis was tested that dimeric conformation is required for normal FXI function by generating a monomeric version of FXI (FXI/PKA4) and comparing it to wild-type FXI in assays requiring factor IX activation by activated FXI (FXIa). FXI/PKA4 was made by replacing the FXI A4 domain with the A4 domain from prekallikrein (PK). A dimeric version of FXI/PKA4 (FXI/PKA4-Gly326) was prepared as a control. Activated FXI/PKA4 and FXI/PKA4-Gly326 activate factor IX with kinetic parameters similar to those of FXIa. In kaolin-triggered plasma clotting assays containing purified phospholipid, FXI/PKA4 and FXI/PKA4-Gly326 have coagulant activity similar to FXI. The surface of activated platelets is likely to be a physiologic site for reactions involving FXI/FXIa. In competition binding assays FXI/PKA4, FXI/PKA4-Gly326, and FXI have similar affinities for activated platelets (K(i) = 12-16 nM). In clotting assays in which phospholipid is replaced by activated platelets, the dimeric proteins FXI and FXI/PKA4-Gly326 promote coagulation similarly; however, monomeric FXI/PKA4 has greatly reduced activity. Western immunoblot analysis confirmed that activated monomeric FXI/PKA4 activates factor IX poorly in the presence of activated platelets. These findings demonstrate the importance of the dimeric state to FXI activity and suggest a novel model for factor IX activation in which FXIa binds to activated platelets by one chain of the dimer, while binding to factor IX through the other.


Assuntos
Plaquetas/química , Dimerização , Fator IX/fisiologia , Fator XIa/química , Fator XIa/fisiologia , Coagulação Sanguínea , Plaquetas/fisiologia , Western Blotting , Membrana Celular/química , Ativação Enzimática , Fator IXa/fisiologia , Fator XI/fisiologia , Fator XIa/genética , Humanos , Cinética , Ativação Plaquetária , Pré-Calicreína/química , Pré-Calicreína/genética , Conformação Proteica , Proteínas Recombinantes de Fusão
15.
Mol Biochem Parasitol ; 107(1): 91-102, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10717305

RESUMO

Microneme organelles are present in all apicomplexan protozoa and contain proteins that are critical for parasite motility and host cell invasion. One apicomplexan-wide family of microneme proteins has been identified with members that are characterised by the possession of thrombospondin type I repeats, conserved adhesive motifs which are implicated in binding to glycosaminoglycan chains. In this paper we describe a micronemal glycoprotein, EtMIC 5, from Eimeria tenella which contains eleven cysteine-rich motifs that have striking similarity to the adhesive Apple (A-) domains of blood coagulation factor XI and plasma pre-kallikrein. EtMIC 5 is confined to an intracellular location in resting sporozoites but is translocated to the parasite surface and secreted into the culture supernatant during parasite infection of MDBK cells. During intracellular replication, the protein is switched off in early schizogony and is then re-expressed within the apical tips of newly formed merozoites. A-domain sequences were also found in microneme proteins from Sarcocystis muris and Toxoplasma gondii and in a protein of unknown localisation from Eimeria acervulina. These studies suggest that A-domain containing proteins may comprise a novel apicomplexan-wide family of microneme adhesins.


Assuntos
Eimeria tenella/química , Fator XI/química , Glicoproteínas/química , Pré-Calicreína/química , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Clonagem Molecular , Eimeria tenella/crescimento & desenvolvimento , Dosagem de Genes , Glicoproteínas/biossíntese , Glicoproteínas/genética , Glicoproteínas/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas de Protozoários/biossíntese , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , RNA Mensageiro/biossíntese , RNA de Protozoário/biossíntese
16.
Immunopharmacology ; 36(2-3): 193-200, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9228546

RESUMO

HK31 (S565-K595) has previously been shown to encompass the binding domain for plasma prekallikrein (PK) within domain 6 of high molecular weight kininogen (HK). The complementary binding domain for HK within PK is mapped to PK56 (F56-G86), in the Apple 1 domain and to PK266 (K266-C295) in the Apple 4 domain. Isothermal titration calorimetry demonstrated that either PK peptide binds to HK31 in 1:1 stoichiometry. Binding of the alternate PK peptide into a ternary complex is facilitated nearly 2-fold. Fluorescence emission spectroscopy revealed that only the binding of PK56 caused a limited decrease in intrinsic tryptophane fluorescence emission intensity of HK31. We conclude that the two PK peptides bind to the HK peptide at different sites. To map the minimal sequence within HK31, truncated new peptides were tested for their ability to compete with HK for binding PK in a cell-free system. D567-T591, a 25-residue peptide which contains sufficient structural information for binding kallikrein in solution, blocked the binding of kallikrein to HK bound to endothelial cells and inhibited PK activation to kallikrein and the generation of kallikrein-activated urokinase on endothelial cell surfaces. HK-derived peptides could modulate excessive fibrinolysis and hypotension in sepsis and multiple trauma.


Assuntos
Cininogênios/sangue , Peptídeos/metabolismo , Ativadores de Plasminogênio/metabolismo , Pré-Calicreína/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/sangue , Sequência de Aminoácidos , Sítios de Ligação/efeitos dos fármacos , Proteínas Sanguíneas/metabolismo , Calorimetria , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fibrinolisina/biossíntese , Humanos , Cininogênios/química , Dados de Sequência Molecular , Peso Molecular , Biossíntese Peptídica , Peptídeos/química , Peptídeos/farmacologia , Pré-Calicreína/química , Espectrometria de Fluorescência , Relação Estrutura-Atividade , Triptofano/metabolismo , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
18.
Biochemistry ; 35(39): 12945-9, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8841140

RESUMO

HK31 (S565-K595) has previously been shown to encompass the binding domain for plasma prekallikrein (PK) within domain 6 of high molecular weight kininogen (HK). The complementary binding domain for HK within PK is mapped to PK56 (F56-G86), in the apple 1 domain, and to PK266 (K266-C295), in the apple 4 domain. Isothermal titration calorimetry was used to directly monitor binding among HK31, PK56, and PK266. Either PK peptide binds to HK31 in 1:1 stoichiometry, regardless of whether a binary complex is first formed between PK266 and HK31 or between PK56 and HK31. Binding of the alternate PK peptide into a ternary complex is facilitated nearly 2-fold. The ternary complex consists of 1:1:1 HK31:PK56:PK266. Furthermore, binary and ternary complex formation is entropically driven and thermodynamically favored, suggesting that the conformational changes accompany binding. Fluorescence emission spectroscopy revealed that binding of PK56 caused a limited decrease in intrinsic tryptophan fluorescence emission intensity of HK31 while binding of PK266 to HK31 or the complex of HK31/PK56 had no such effect. We conclude that the two PK peptides bind to the HK peptide at different sites. The binding between HK and PK is likely due to conformational changes which serve to juxtapose the PK binding domain within HK with the HK binding site involving two spatial proximity segments.


Assuntos
Cininogênios/química , Pré-Calicreína/química , Sequência de Aminoácidos , Sítios de Ligação , Calorimetria , Cininogênios/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Pré-Calicreína/metabolismo , Ligação Proteica , Conformação Proteica , Espectrometria de Fluorescência , Termodinâmica , Triptofano
19.
J Biol Chem ; 271(22): 13061-7, 1996 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-8662705

RESUMO

Prekallikrein, the precursor to the serine proteinase kallikrein, circulates in plasma in an equimolar complex with H-kininogen. The binding to H-kininogen is mediated by the kallikrein heavy chain consisting of four "apple" domains, A1-A4, which attaches to H-kininogen with high specificity and affinity (KD = 83 nM). At least two distinct portions of the kallikrein heavy chain form this H-kininogen binding site: a proximal segment located in the NH2-terminal fragment of the heavy chain encompassing A1, and distal segment(s) located in COOH-terminal fragment spanning domains A2-A4. The proximal binding segment has been located to amino acid positions 56-86 of A1. To precisely map the distal binding segment, we have identified monoclonal antibodies directed to the COOH-terminal fragment which interfere with the H-kininogen-prekallikrein complex formation. Monoclonal antibody 13G11 binds to recombinant apple domain A4 but not to domain A3 of the prekallikrein heavy chain. Deletion mutagenesis of domain A4 narrowed down the target epitope of 13G11 to the center portion of domain A4, positions 284-331. Direct binding studies of H-kininogen to various domain A4 constructs revealed that the distal H-kininogen binding portion is located on a segment of 48 residues, which overlaps the 13G11 epitope. Hence the tight interaction of H-kininogen and prekallikrein is mediated by at least two separate sequence segments located in domains A1 and A4, respectively, of the prekallikrein heavy chain. The isolated distal binding segment significantly prolongs the partial thromboplastin time of reconstituted Williams plasma thus stressing the critical role of the prekallikrein-H-kininogen complex formation in the initiation of the endogenous blood coagulation cascade.


Assuntos
Cininogênios/metabolismo , Pré-Calicreína/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Sequência de Bases , Sítios de Ligação , Western Blotting , Linhagem Celular , Cricetinae , Brometo de Cianogênio , Primers do DNA , Epitopos/química , Epitopos/imunologia , Humanos , Dados de Sequência Molecular , Tempo de Tromboplastina Parcial , Mapeamento de Peptídeos , Pré-Calicreína/química , Pré-Calicreína/imunologia
20.
Arch Biochem Biophys ; 314(1): 159-64, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7944388

RESUMO

The C-terminal end of the heavy chain of human plasma prekallikrein or kallikrein contains a binding site for high-molecular-weight kininogen, the nonenzymatic procofactor of contact activation. To further map this binding site, a series of overlapping peptides were synthesized. The amount of kallikrein that bound to kininogen-coated microtiter plate wells in the presence of increasing concentrations of each peptide was determined by kallikrein amidolytic activity. A peptide encompassing Lys266-Gly295 of kallikrein, conformationally constrained by a disulfide bond, displayed the lowest Kd value (approximately 67 microM). The linear peptide, Leu262-Gly295, displayed lower affinity (129 microM). N-terminal or C-terminal truncation/extension peptides of this sequence diminished binding activity. Since the closely related protein, factor XI, has been shown to bind kininogen, a kallikrein-based peptide (Phe56-Gly86) homologous to the binding domain of FXI, was examined and found to possess less, but significant, binding affinity for kininogen (Kd 530 microM). Isothermal titration calorimetry was used to assess binding between the kallikrein-based peptides and a peptide encompassing the kallikrein binding domain in kininogen (Ser565-Lys595). Leu262-Gly295 possesses potent binding activity (Kd 52 microM), while Phe56-Gly86 displays poorer binding activity (Kd 400 microM). These interactions are endothermic and entropically favored, suggesting that a conformational rearrangement takes place upon binding. We conclude that the binding site for kininogen within prekallikrein is composed of discontinuous linear segments that form a contiguous surface in the folded protein.


Assuntos
Calicreínas/química , Cininogênios/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Ligação Competitiva , Calorimetria , Humanos , Calicreínas/metabolismo , Cininogênios/química , Dados de Sequência Molecular , Peso Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Pré-Calicreína/química , Pré-Calicreína/metabolismo , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA