Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 27(4)2022 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-35209023

RESUMO

In the modern world, complications caused by disorders in the blood coagulation system are found in almost all areas of medicine. Thus, the development of new, more advanced drugs that can prevent pathological conditions without disrupting normal hemostasis is an urgent task. The blood coagulation factor XIIa is one of the most promising therapeutic targets for the development of anticoagulants based on its inhibitors. The initial stage of drug development is directly related to computational methods of searching for a lead compound. In this study, docking followed by quantum chemical calculations was used to search for noncovalent low-molecular-weight factor XIIa inhibitors in a focused library of druglike compounds. As a result of the study, four low-molecular-weight compounds were experimentally confirmed as factor XIIa inhibitors. Selectivity testing revealed that two of the identified factor XIIa inhibitors were selective over the coagulation factors Xa and XIa.


Assuntos
Proteínas Sanguíneas , Fator XIIa , Simulação de Acoplamento Molecular , Proteínas Sanguíneas/síntese química , Proteínas Sanguíneas/química , Fator XIIa/antagonistas & inibidores , Fator XIIa/química , Humanos
2.
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
3.
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
4.
Blood ; 121(19): 3962-9, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23515926

RESUMO

Factor XI (fXI) is a homodimeric zymogen that is converted to a protease with 1 (1/2-fXIa) or 2 (fXIa) active subunits by factor XIIa (fXIIa) or thrombin. It has been proposed that the dimeric structure is required for normal fXI activation. Consistent with this premise, fXI monomers do not reconstitute fXI-deficient mice in a fXIIa-dependent thrombosis model. FXI activation by fXIIa or thrombin is a slow reaction that can be accelerated by polyanions. Phosphate polymers released from platelets (poly-P) can enhance fXI activation by thrombin and promote fXI autoactivation. Poly-P increased initial rates of fXI activation 30- and 3000-fold for fXIIa and thrombin, respectively. FXI monomers were activated more slowly than dimers by fXIIa in the presence of poly-P. However, this defect was not observed when thrombin was the activating protease, nor during fXI autoactivation. The data suggest that fXIIa and thrombin activate fXI by different mechanisms. FXIIa may activate fXI through a trans-activation mechanism in which the protease binds to 1 subunit of the dimer, while activating the other subunit. For activation by thrombin, or during autoactivation, the data support a cis-activation mechanism in which the activating protease binds to and activates the same fXI subunit.


Assuntos
Fator XI/química , Fator XI/metabolismo , Fator XIa/metabolismo , Animais , Trombose das Artérias Carótidas/genética , Trombose das Artérias Carótidas/metabolismo , Fator XI/genética , Deficiência do Fator XI/genética , Deficiência do Fator XI/metabolismo , Fator XIIa/química , Fator XIIa/metabolismo , Fator XIa/química , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína
5.
Biologicals ; 40(1): 79-83, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22122985

RESUMO

The aim of this study was to establish the first national standard for prekallikrein activator (PKA) calibrating to the first international standard PKA. A collaborative study among five laboratories, including three manufacturers and two national control laboratories, was carried out to evaluate the suitability of a candidate to serve as a national standard of PKA. The candidate was manufactured in GMP facility following approved human serum albumin fractionation procedure and freeze-dried 5% albumin solution containing PKA. Participants were provided with sufficient samples and asked to use lab-made prekallikrein substrate prepared in accordance with European Pharmacopeia and also to use a commercial prekallikrein provided as part of the study. The PKA concentration of the candidate was 61.8 IU per vial using lab-made prekallikrein. However, the concentration was 54.2 IU per vial using commercial prekallikrein. The variability obtained at each laboratory ranged from 1.9% to 5.1% for within-a-day and from 5.6% to 9.0% for day-to-day. The candidate showed excellent stability from accelerated degradation study and real-time stability study. As a conclusion, the candidate preparation was suitable to serve as a Korean National Standard for PKA.


Assuntos
Bioensaio/normas , Fator XIIa/normas , Calibragem , Fator XIIa/análise , Fator XIIa/química , Feminino , Humanos , Masculino , Padrões de Referência , Reprodutibilidade dos Testes , República da Coreia
6.
J Mater Sci Mater Med ; 21(3): 931-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19851837

RESUMO

Biomaterial induced coagulation encompasses plasmatic and cellular processes. The functional loss of biomedical devices possibly resulting from these thrombotic reactions motivates the need for a better understanding of processes occurring at blood-biomaterial interfaces. Well defined model surfaces providing specific chemical-physical properties (self assembled monolayers (SAMs)) displaying hydrophobic or/and acidic terminal groups were used to uncover initial mechanisms of biomaterial induced coagulation. We investigated the influence of electrical charge and wettability on platelet- and contact activation, the two main actors of blood coagulation, which are often considered as separate mechanisms in biomaterials research. Our results show a dependence of contact activation on acidic surface groups and a correlation of platelet adhesion to surface hydrophobicity. Clot formation resulting from the interplay of blood platelets and contact activation was only found on surfaces combining both acidic and hydrophobic surface groups but not on monolayers displaying extreme hydrophobic/acidic properties.


Assuntos
Ânions , Coagulação Sanguínea , Interações Hidrofóbicas e Hidrofílicas , Adsorção , Materiais Biocompatíveis/química , Sangue/metabolismo , Plaquetas/química , Plaquetas/metabolismo , Cristalização , Fator XIIa/química , Fibrinogênio/química , Humanos , Técnicas In Vitro , Calicreínas/química , Adesividade Plaquetária , Molhabilidade
7.
Comput Math Methods Med ; 2020: 2852051, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32549905

RESUMO

Human coagulation factor XIIa (FXIIa) is a trypsin-like serine protease that is involved in pathologic thrombosis. As a potential target for designing safe anticoagulants, FXIIa has received a great deal of interest in recent years. In the present study, we employed virtual high-throughput screening of 500,064 compounds within Enamine database to acquire the most potential inhibitors of FXIIa. Subsequently, 18 compounds with significant binding energy (from -65.195 to -15.726 kcal/mol) were selected, and their ADMET properties were predicted to select representative inhibitors. Three compounds (Z1225120358, Z432246974, and Z146790068) exhibited excellent binding affinity and druggability. MD simulation for FXIIa-ligand complexes was carried out to reveal the stability and inhibition mechanism of these three compounds. Through the inhibition of activated factor XIIa assay, we tested the activity of five compounds Z1225120358, Z432246974, Z45287215, Z30974175, and Z146790068, with pIC50 values of 9.3∗10-7, 3.0∗10-5, 7.8∗10-7, 8.7∗10-7, and 1.3∗10-6 M, respectively; the AMDET properties of Z45287215 and Z30974175 show not well but have better inhibition activity. We also found that compounds Z1225120358, Z45287215, Z30974175, and Z146790068 could be more inhibition of FXIIa than Z432246974. Collectively, compounds Z1225120358, Z45287215, Z30974175, and Z146790068 were anticipated to be promising drug candidates for inhibition of FXIIa.


Assuntos
Anticoagulantes/química , Anticoagulantes/farmacologia , Fator XIIa/antagonistas & inibidores , Fator XIIa/química , Sítios de Ligação , Biologia Computacional , Bases de Dados de Produtos Farmacêuticos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/estatística & dados numéricos , Fator XIIa/metabolismo , Ensaios de Triagem em Larga Escala/estatística & dados numéricos , Humanos , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Interface Usuário-Computador
8.
Eur J Med Chem ; 208: 112753, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-32883641

RESUMO

Coagulation factor XII (FXII), a S1A serine protease, was discovered more than fifty years ago. However, its in vivo functions and its three-dimensional structure started to be disclosed in the last decade. FXII was found at the crosstalk of several physiological pathways including the intrinsic coagulation pathway, the kallikrein-kinin system, and the immune response. The FXII inhibition emerges as a therapeutic strategy for the safe prevention of artificial surface-induced thrombosis and in patients suffering from hereditary angioedema. The anti-FXII antibody garadacimab discovered by phage-display library technology is actually under phase II clinical evaluation for the prophylactic treatment of hereditary angioedema. The implication of FXII in neuro-inflammatory and neurodegenerative disorders is also an emerging research field. The FXII or FXIIa inhibitors currently under development include peptides, proteins, antibodies, RNA-based technologies, and, to a lesser extent, small-molecular weight inhibitors. Most of them are proteins, mainly isolated from hematophagous arthropods and plants. The discovery and development of these FXII inhibitors and their potential indications are discussed in the review.


Assuntos
Anticoagulantes/farmacologia , Fator XII/antagonistas & inibidores , Fator XIIa/antagonistas & inibidores , Inibidores de Serina Proteinase/farmacologia , Animais , Anticoagulantes/química , Descoberta de Drogas , Fator XII/química , Fator XIIa/química , Humanos , Inibidores de Serina Proteinase/química
9.
Thromb Haemost ; 120(3): 400-411, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31940673

RESUMO

Factor XII (FXII) zymogen activation requires cleavage after arginine 353 located in the activation loop. This cleavage can be executed by activated FXII (autoactivation), plasma kallikrein (PKa), or plasmin. Previous studies proposed that the activation loop of FXII is shielded to regulate FXII activation and subsequent contact activation. In this study, we aimed to elucidate this mechanism by expressing and characterizing seven consecutive N-terminally truncated FXII variants as well as full-length wild-type (WT) FXII. As soon as the fibronectin type II domain is lacking (FXII Δ1-71), FXII cleavage products appear on Western blot. These fragments display spontaneous amidolytic activity, indicating that FXII without the fibronectin type II domain is susceptible to autoactivation. Additionally, truncated FXII Δ1-71 is more easily activated by PKa or plasmin than full-length WT FXII. To exclude a contribution of autoactivation, we expressed active-site incapacitated FXII truncation variants (S544A). FXII S544A Δ1-71 is highly susceptible to cleavage by PKa, indicating exposure of the activation loop. In surface binding experiments, we found that the fibronectin type II domain is non-essential for binding to kaolin or polyphosphate, whereas the following epidermal growth factor-like domain is indispensable. Binding of full-length FXII S544A to kaolin or polyphosphate increases its susceptibility to cleavage by PKa. Moreover, the activation of full-length WT FXII by PKa increases approximately threefold in the presence of kaolin. Deletion of the fibronectin type II domain eliminates this effect. Combined, these findings suggest that the fibronectin type II domain shields the activation loop of FXII, ensuring zymogen quiescence.


Assuntos
Precursores Enzimáticos/química , Fator XII/química , Fibrinolisina/química , Fibronectinas/química , Calicreínas/química , Animais , Sítios de Ligação , Coagulação Sanguínea , Bradicinina/química , Domínio Catalítico , Bovinos , Fator XIIa/química , Fibronectinas/sangue , Células HEK293 , Humanos , Calicreínas/sangue , Caulim/química , Polifosfatos/química , Ligação Proteica , Domínios Proteicos
10.
J Mol Graph Model ; 89: 225-233, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30921556

RESUMO

Thrombosis represents a major cause of morbidity and mortality around the world. Peptides isolated from natural sources have been proven to have anticoagulant and antithrombotic properties. VITPOR AI, a 16-mer peptide, isolated from Porphyra yezoensis was reported to have anticoagulant property. In this study, the coagulation factor XIIa activity in the presence of VITPOR AI was determined. Molecular modelling was performed to investigate the interaction between peptide and FXIIa. The structure of the peptide was predicted using PEP-FOLD3 server and simulated by molecular dynamics (MD) using GROMACS package. Molecular docking was carried out using peptide-protein docking software, pepATTRACT and its stability was confirmed by MD simulations. The chromogenic substrate assay revealed that the peptide inhibited the amidolytic activity of FXIIa with IC50 of 70.24 µM. The docking result showed peptide interactions through hydrogen bonds with Pro 96, Tyr 99, Glu 146, Gly 193 and Ser 195 of FXIIa. The MD simulation demonstrated that the peptide's binding with the FXIIa was stable as it did not move away from its binding region throughout the simulation period of 100 ns Moreover, MM/PBSA analysis also indicated a stable binding between the protein and peptide. These results suggest that the inhibition of the FXIIa activity might be due to binding of the peptide to oxyanion hole of the catalytic site. Thus, VITPOR AI could be explored as a potent anticoagulant which inhibits only intrinsic pathway of coagulation cascade but does not affect the extrinsic pathway.


Assuntos
Anticoagulantes/química , Anticoagulantes/farmacologia , Fator XIIa/antagonistas & inibidores , Fator XIIa/química , Peptídeos/química , Peptídeos/farmacologia , Porphyra/química , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Conformação Molecular , Extratos Vegetais , Relação Estrutura-Atividade
11.
Acta Crystallogr D Struct Biol ; 75(Pt 6): 578-591, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-31205020

RESUMO

Coagulation factor XII (FXII) is a key initiator of the contact pathway, which contributes to inflammatory pathways. FXII circulates as a zymogen, which when auto-activated forms factor XIIa (FXIIa). Here, the production of the recombinant FXIIa protease domain (ßFXIIaHis) with yields of ∼1-2 mg per litre of insect-cell culture is reported. A second construct utilized an N-terminal maltose-binding protein (MBP) fusion (MBP-ßFXIIaHis). Crystal structures were determined of MBP-ßFXIIaHis in complex with the inhibitor D-Phe-Pro-Arg chloromethyl ketone (PPACK) and of ßFXIIaHis in isolation. The ßFXIIaHis structure revealed that the S2 and S1 pockets were occupied by Thr and Arg residues, respectively, from an adjacent molecule in the crystal. The Thr-Arg sequence mimics the P2-P1 FXIIa cleavage-site residues present in the natural substrates prekallikrein and FXII, and Pro-Arg (from PPACK) mimics the factor XI cleavage site. A comparison of the ßFXIIaHis structure with the available crystal structure of the zymogen-like FXII protease revealed large conformational changes centred around the S1 pocket and an alternate conformation for the 99-loop, Tyr99 and the S2 pocket. Further comparison with activated protease structures of factors IXa and Xa, which also have the Tyr99 residue, reveals that a more open form of the S2 pocket only occurs in the presence of a substrate mimetic. The FXIIa inhibitors EcTI and infestin-4 have Pro-Arg and Phe-Arg P2-P1 sequences, respectively, and the interactions that these inhibitors make with ßFXIIa are also described. These structural studies of ßFXIIa provide insight into substrate and inhibitor recognition and establish a scaffold for the structure-guided drug design of novel antithrombotic and anti-inflammatory agents.


Assuntos
Fator XIIa , Proteínas Ligantes de Maltose , Proteínas Recombinantes de Fusão/química , Clorometilcetonas de Aminoácidos/química , Animais , Sítios de Ligação , Linhagem Celular , Cristalização , Cristalografia por Raios X/métodos , Drosophila melanogaster , Fator XIIa/química , Fator XIIa/metabolismo , Proteínas Ligantes de Maltose/química , Proteínas Ligantes de Maltose/metabolismo , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Especificidade por Substrato
12.
Chem Biol Drug Des ; 94(5): 1905-1918, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31148409

RESUMO

The experimental inhibitory potency of benzamidine (BEN) paved way for further design and development of inhibitors that target ß-FXIIa. Structural dynamics of the loops and catalytic residues that encompass the binding pocket of ß-FXIIa and all serine proteases are crucial to their overall activity. Employing molecular dynamics and post-MD analysis, this study sorts to unravel the structural and molecular events that accompany the inhibitory activity of BEN on human ß-FXIIa upon selective non-covalent binding. Analysis of conformational dynamics of crucial loops revealed prominent alterations of the original conformational posture of FXIIa, evidenced by increased flexibility, decreased compactness, and an increased exposure to solvent upon binding of BEN, which could have in turn interfered with the essential roles of these loops in enhancing their procoagulation interactions with biological substrates and cofactors, altogether resulting in the consequential inactivation of FXIIa. A sustained interaction of the catalytic triad residues and key residues of the autolysis loop impeded their roles in catalysis which equally enhanced the inhibitory potency of BEN toward ß-FXIIa evidenced by a favorable binding. Findings provide essential structural and molecular insights that could facilitate the structure-based design of novel antithrombotic compounds with enhanced inhibitory activities and low therapeutic risk.


Assuntos
Benzamidinas/química , Coagulação Sanguínea/efeitos dos fármacos , Fator XIIa/antagonistas & inibidores , Fibrinolíticos/química , Sequência de Aminoácidos , Domínio Catalítico , Desenho de Fármacos , Fator XIIa/química , Humanos , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Termodinâmica
13.
J Med Chem ; 51(11): 3077-80, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18459730

RESUMO

Recently, FXIIa was highlighted as an original attractive target for the development of new anticoagulant drugs with low rates of therapy-related hemorrhages. In this work, we describe the development of a new series of 3-carboxamide-coumarins that are the first potent and selective nonpeptidic inhibitors of FXIIa.


Assuntos
Amidas/síntese química , Anticoagulantes/síntese química , Cumarínicos/síntese química , Fator XIIa/antagonistas & inibidores , Amidas/química , Anticoagulantes/química , Cumarínicos/química , Fator XIIa/química , Relação Estrutura-Atividade
14.
Biomaterials ; 27(28): 4840-5, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16765435

RESUMO

Autohydrolysis of blood factor XII (FXII+FXIIa-->2FXIIa) is found to be a facile reaction in neat-buffer buffer solutions of FXII but an insignificant reaction in the presence of plasma proteins. Autohydrolysis causes a chromogenic assay for FXIIa in buffer solution to strongly deviate from the traditional plasma-coagulation assay. Autohydrolysis can be accommodated by performing chromogenic detection of FXIIa as a rate assay in swamping concentrations of FXII. Rate-assay results performed in this way are shown to be in analytical agreement with the plasma-coagulation assay. Autohydrolysis can be used as a means of amplifying FXIIa produced by contacting neat-buffer solutions of FXII with biomaterials, suggesting a route to highly sensitive measurement of biomaterial hemocompatibility.


Assuntos
Bioensaio/métodos , Compostos Cromogênicos/metabolismo , Fator XIIa/metabolismo , Compostos Cromogênicos/química , Fator XIIa/química , Humanos , Hidrólise , Plasma/química , Plasma/metabolismo , Estatística como Assunto
15.
Blood Coagul Fibrinolysis ; 16(6): 447-52, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16093737

RESUMO

Our aim was to establish whether differences in clotting times for recalcified blood and plasma samples might be explained by the use of different blood collection tubes. Samples obtained from different plastic vacuum tubes were recalcified and clotting times determined by free oscillation rheometry. The clotting times for blood collected in Vacutainer (Becton Dickinson, Rutherford, New Jersey, USA) or Vacuette (Greiner Bio-One, Kremsmünster, Austria) tubes decreased with time, with maximal effect after 30 min. Blood from Monovette (Sarstedt, Nümbrecht, Germany) tubes displayed longer clotting times, which did not decrease with time. Clotting times for plasma prepared after 1 h storage in Vacutainer or Vacuette tubes were unaffected by subsequent addition of corn trypsin inhibitor to inhibit factor XIIa, although an antibody against factor XI prolonged the clotting time markedly. In Monovette plasma, both corn trypsin inhibitor and anti-factor XI effectively prolonged the clotting time. When corn trypsin inhibitor or anti-factor XI was added to the tubes before blood collection, but both additions clearly prolonged the clotting times in all types of tubes, even though corn trypsin inhibitor was less effective in whole blood. Antibodies against human tissue factor did not affect the clotting times. The amounts of platelet or leukocyte microparticles in plasma were low and similar in all tubes. This indicates that blood collection in Vacutainer or Vacuette tubes induces a rapid activation of factor XII and factor XI.


Assuntos
Preservação de Sangue , Coleta de Amostras Sanguíneas , Anticorpos/química , Anticorpos/imunologia , Testes de Coagulação Sanguínea , Preservação de Sangue/normas , Coleta de Amostras Sanguíneas/instrumentação , Coleta de Amostras Sanguíneas/normas , Fator XI/química , Fator XI/imunologia , Fator XIIa/antagonistas & inibidores , Fator XIIa/química , Humanos , Proteínas de Plantas/química , Reprodutibilidade dos Testes , Tromboplastina/química , Tromboplastina/imunologia
16.
J Thromb Haemost ; 13(4): 580-91, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25604127

RESUMO

BACKGROUND: Coagulation factor XII is a serine protease that is important for kinin generation and blood coagulation, cleaving the substrates plasma kallikrein and FXI. OBJECTIVE: To investigate FXII zymogen activation and substrate recognition by determining the crystal structure of the FXII protease domain. METHODS AND RESULTS: A series of recombinant FXII protease constructs were characterized by measurement of cleavage of chromogenic peptide and plasma kallikrein protein substrates. This revealed that the FXII protease construct spanning the light chain has unexpectedly weak proteolytic activity compared to ß-FXIIa, which has an additional nine amino acid remnant of the heavy chain present. Consistent with these data, the crystal structure of the light chain protease reveals a zymogen conformation for active site residues Gly193 and Ser195, where the oxyanion hole is absent. The Asp194 side chain salt bridge to Arg73 constitutes an atypical conformation of the 70-loop. In one crystal form, the S1 pocket loops are partially flexible, which is typical of a zymogen. In a second crystal form of the deglycosylated light chain, the S1 pocket loops are ordered, and a short α-helix in the 180-loop of the structure results in an enlarged and distorted S1 pocket with a buried conformation of Asp189, which is critical for P1 Arg substrate recognition. The FXII structures define patches of negative charge surrounding the active site cleft that may be critical for interactions with inhibitors and substrates. CONCLUSIONS: These data provide the first structural basis for understanding FXII substrate recognition and zymogen activation.


Assuntos
Precursores Enzimáticos/química , Fator XII/química , Coagulação Sanguínea , Domínio Catalítico , Cristalografia por Raios X , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Fator XII/genética , Fator XII/metabolismo , Fator XIIa/química , Fator XIIa/metabolismo , Humanos , Calicreínas/química , Calicreínas/metabolismo , Cinética , Modelos Moleculares , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
17.
Transfus Apher Sci ; 29(3): 195-203, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14572810

RESUMO

Hereditary angioedema (HAE), which is characterized by episodic localized angioedema of the skin or mucosa, results from heterozygous deficiency of the plasma protease inhibitor, C1 inhibitor (C1INH). The most obvious biologic role of C1INH, therefore, is prevention of excessive vascular permeability. A variety of data indicate that this role is primarily a product of regulation of the contact system proteases, factor XIIa and plasma kallikrein. The C1INH deficient mouse, although it does not have episodes of cutaneous angioedema, does have increased vascular permeability which is reversed by treatment with C1INH, with the plasma kallikrein inhibitor, DX88, and with the bradykinin 2 receptor (Bk2R) antagonist, Hoe140. In addition, mice deficient in both C1INH and the Bk2R do not have increased vascular permeability. These analyses strengthen the argument that angioedema is mediated by bradykinin. This mouse also provides a system to test new potential therapeutic approaches. In addition to its role in the regulation of vascular permeability, C1INH also is an important modulator of inflammatory responses via regulation of activation of both the contact and the complement systems, and very likely via activities unrelated to protease inhibition.


Assuntos
Angioedema/genética , Angioedema/patologia , Proteínas Inativadoras do Complemento 1/genética , Inibidores de Proteases/farmacologia , Animais , Bradicinina/metabolismo , Ativação do Complemento , Proteínas Inativadoras do Complemento 1/deficiência , Modelos Animais de Doenças , Fator XIIa/química , Humanos , Calicreínas/química , Camundongos , Modelos Biológicos
18.
J Thromb Haemost ; 12(10): 1678-86, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25142753

RESUMO

BACKGROUND: Corn trypsin inhibitor (CTI), an inhibitor of FXIIa, is used to prevent plasma coagulation by contact activation, to specifically investigate tissue factor (TF)-initiated coagulation. OBJECTIVE: In the present work the specificity of CTI for factor (F) XIIa is questioned. METHODS AND RESULTS: In the commercial available plasma coagulation assays CTI was found to double activated partial thromboplastin time (APTT) at a plasma concentration of 7.3 ± 1.5 µm CTI (assay concentration 2.4 µm). No effect was found on the prothrombin time (PT) when high TF concentrations were used. Also, with specific antibodies for FXIIa and for FXIa only APTT was found to be extended but not PT. With specific enzyme assays using chromogenic substrates CTI was shown to be a strong inhibitor of FXIIa and a competitive inhibitor of FXIa with Ki  = 8.1 ± 0.3 µm, without effect on the coagulation factors FVIIa, FIXa, FXa and thrombin. In thrombin generation and coagulation (free oscillation rheometry, FOR) assays, initiated with low TF concentrations, no effect of CTI (plasma concentrations of 4.4 and 13.6 µm CTI, 25 resp. 100 mg L(-1) in blood) was found with ≥ 1 pm TF. At ≤ 0.1 pm TF in the FOR whole blood assay the coagulation time (CT) concentration dependently increased while the plasma CT became longer than the observation time. CONCLUSION: To avoid inhibition of FXIa and the thrombin feedback loop we recommend that for coagulation assays the concentration of CTI in blood should be below 20 mg L(-1) (1.6 µm) and in plasma below 3 µm.


Assuntos
Testes de Coagulação Sanguínea , Coagulação Sanguínea/efeitos dos fármacos , Fator XIIa/química , Fator XIa/química , Proteínas de Plantas/química , Anticorpos Monoclonais/química , Ligação Competitiva , Calibragem , Fator XIIa/imunologia , Fator XIa/imunologia , Hematócrito , Humanos , Oscilometria , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Trombina/química
19.
Thromb Haemost ; 110(3): 423-33, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23494059

RESUMO

The kallikrein kinin system (KKS) consists of serine proteases involved in the production of peptides called kinins, principally bradykinin and Lys-bradykinin (kallidin). The KKS contributes to a variety of physiological processes including inflammation, blood pressure control and coagulation. Here we review the protein structural data available for these serine proteases and examine the molecular mechanisms of zymogen activation and substrate recognition focusing on plasma kallikrein (PK) and tissue kallikrein (KLK1) cleavage of kininogens. PK circulates as a zymogen bound to high-molecular-weight kininogen (HK). PK is activated by coagulation factor XIIa and then cleaves HK to generate bradykinin and factor XII to generate further XIIa.A structure has been described for the activated PK protease domain in complex with the inhibitor benzamidine. Kallikrein-related peptidases (KLKs) have a distinct domain structure and exist as a family of 15 genes which are differentially expressed in many tissues and the central nervous system.They cleave a wide variety of substrates including low-molecular-weight kininogen (LK) and matrix proteins. Crystal structures are available for KLK1, 3, 4, 5, 6 and 7 activated protease domains typically in complex with S1 pocket inhibitors. A substrate mimetic complex is described for KLK3 which provides insight into substrate recognition. A zymogen crystal structure determined for KLK6 reveals a closed S1 pocket and a novel mechanism of zymogen activation. Overall these structures have proved highly informative in understanding the molecular mechanisms of the KKS and provide templates to design inhibitors for treatment of a variety of diseases.


Assuntos
Calicreína Plasmática/química , Calicreínas Teciduais/química , Sequência de Aminoácidos , Animais , Pressão Sanguínea , Catálise , Domínio Catalítico , Precursores Enzimáticos/química , Fator XIIa/química , Humanos , Inflamação , Sistema Calicreína-Cinina , Cininogênios/química , Modelos Moleculares , Dados de Sequência Molecular , Peptídeo Hidrolases/química , Homologia de Sequência de Aminoácidos , Serina Proteases/química , Especificidade por Substrato
20.
Thromb Haemost ; 110(3): 399-407, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23846131

RESUMO

Plasma prekallikrein is the liver-derived precursor of the trypsin-like serine protease plasma kallikrein (PK) and circulates in plasma bound to high molecular weight kininogen. The zymogen is converted to PK by activated factor XII. PK drives multiple proteolytic reaction cascades in the cardiovascular system such as the intrinsic pathway of coagulation, the kallikrein-kinin system, the fibrinolytic system, the renin-angiotensin system and the alternative complement pathway. Here, we review the biochemistry and cell biology of PK and focus on recent in vivo studies that have established important functions of the protease in procoagulant and proinflammatory disease states. Targeting PK offers novel strategies not previously appreciated to interfere with thrombosis and vascular inflammation in a broad variety of diseases.


Assuntos
Bradicinina/metabolismo , Calicreína Plasmática/metabolismo , Animais , Aprotinina/química , Coagulação Sanguínea , Hemorragia Cerebral/metabolismo , Proteínas do Sistema Complemento , Cisteína/química , Retinopatia Diabética/metabolismo , Dissulfetos/química , Fator XIIa/química , Fibrinólise , Hemostasia , Humanos , Inflamação , Sistema Calicreína-Cinina , Calicreínas/química , Cininas/química , Camundongos , Oligonucleotídeos Antissenso/química , Peptídeos/química , Estrutura Terciária de Proteína , Proteólise , Sistema Renina-Angiotensina , Transdução de Sinais , Trombose/metabolismo , Tripsina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA