Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 22(5): 1336-1346, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38242207

RESUMEN

BACKGROUND: Factor (F)XI can be activated by proteases, including thrombin and FXIIa. The interactions of these enzymes with FXI are transient in nature and therefore difficult to study. OBJECTIVES: To identify the binding interface between thrombin and FXI and understand the dynamics underlying FXI activation. METHODS: Crosslinking mass spectrometry was used to localize the binding interface of thrombin on FXI. Molecular dynamics simulations were applied to investigate conformational changes enabling thrombin-mediated FXI activation after binding. The proposed trajectory of activation was examined with nanobody 1C10, which was previously shown to inhibit thrombin-mediated activation of FXI. RESULTS: We identified a binding interface of thrombin located on the light chain of FXI involving residue Pro520. After this initial interaction, FXI undergoes conformational changes driven by binding of thrombin to the apple 1 domain in a secondary step to allow migration toward the FXI cleavage site. The 1C10 binding site on the apple 1 domain supports this proposed trajectory of thrombin. We validated the results with known mutation sites on FXI. As Pro520 is conserved in prekallikrein (PK), we hypothesized and showed that thrombin can bind PK, even though it cannot activate PK. CONCLUSION: Our investigations show that the activation of FXI is a multistaged procedure. Thrombin first binds to Pro520 in FXI; thereafter, it migrates toward the activation site by engaging the apple 1 domain. This detailed analysis of the interaction between thrombin and FXI paves a way for future interventions for bleeding or thrombosis.


Asunto(s)
Factor XI , Simulación de Dinámica Molecular , Unión Proteica , Trombina , Trombina/metabolismo , Trombina/química , Humanos , Factor XI/metabolismo , Factor XI/química , Sitios de Unión , Multimerización de Proteína , Mutación , Conformación Proteica , Coagulación Sanguínea , Precalicreína/metabolismo , Precalicreína/química , Subunidades de Proteína/metabolismo , Activación Enzimática , Factor XIa/metabolismo , Factor XIa/química
2.
Blood Adv ; 7(7): 1156-1167, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36409609

RESUMEN

A dysregulated plasma contact system is involved in various pathological conditions, such as hereditary angioedema, Alzheimer disease, and sepsis. We previously showed that the 3E8 anti-high molecular weight kininogen (anti-HK) antibody blocks HK cleavage and bradykinin generation in human plasma ex vivo. Here, we show that 3E8 prevented not only HK cleavage but also factor XI (FXI) and prekallikrein (PK) activation by blocking their binding to HK in mouse plasma in vivo. 3E8 also inhibited contact system-induced bradykinin generation in vivo. Interestingly, FXII activation was also inhibited, likely because of the ability of 3E8 to block the positive feedback activation of FXII by kallikrein (PKa). In human plasma, 3E8 also blocked PK and FXI binding to HK and inhibited both thrombotic (FXI activation) and inflammatory pathways (PK activation and HK cleavage) of the plasma contact system activation ex vivo. Moreover, 3E8 blocked PKa binding to HK and dose-dependently inhibited PKa cleavage of HK. Our results reveal a novel strategy to inhibit contact system activation in vivo, which may provide an effective method to treat human diseases involving contact system dysregulation.


Asunto(s)
Precalicreína , Trombosis , Humanos , Animales , Ratones , Precalicreína/química , Precalicreína/metabolismo , Factor XI/metabolismo , Bradiquinina/farmacología , Bradiquinina/química , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo
3.
J Biol Chem ; 298(2): 101567, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35007530

RESUMEN

Skeletal muscle myosin (SkM) has been shown to possess procoagulant activity; however, the mechanisms of this coagulation-enhancing activity involving plasma coagulation pathways and factors are incompletely understood. Here, we discovered direct interactions between immobilized SkM and coagulation factor XI (FXI) using biolayer interferometry (Kd = 0.2 nM). In contrast, we show that prekallikrein, a FXI homolog, did not bind to SkM, reflecting the specificity of SkM for FXI binding. We also found that the anti-FXI monoclonal antibody, mAb 1A6, which recognizes the Apple (A) 3 domain of FXI, potently inhibited binding of FXI to immobilized SkM, implying that SkM binds FXI A3 domain. In addition, we show that SkM enhanced FXI activation by thrombin in a concentration-dependent manner. We further used recombinant FXI chimeric proteins in which each of the four A domains of the heavy chain (designated A1 through A4) was individually replaced with the corresponding A domain from prekallikrein to investigate SkM-mediated enhancement of thrombin-induced FXI activation. These results indicated that activation of two FXI chimeras with substitutions of either the A3 domains or A4 domains was not enhanced by SkM, whereas substitution of the A2 domain did not reduce the thrombin-induced activation compared with wildtype FXI. These data strongly suggest that functional interaction sites on FXI for SkM involve the A3 and A4 domains. Thus, this study is the first to reveal and support the novel intrinsic blood coagulation pathway concept that the procoagulant mechanisms of SkM include FXI binding and enhancement of FXI activation by thrombin.


Asunto(s)
Coagulación Sanguínea , Factor XI , Miosinas del Músculo Esquelético , Trombina , Anticuerpos Monoclonales/química , Sitios de Unión , Factor XI/química , Factor XI/genética , Factor XI/metabolismo , Precalicreína/química , Precalicreína/metabolismo , Dominios Proteicos , Proteínas Recombinantes de Fusión/química , Miosinas del Músculo Esquelético/metabolismo , Trombina/metabolismo
4.
Blood Adv ; 6(10): 3142-3154, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35086137

RESUMEN

Factor XII (FXII) is the zymogen of a plasma protease (FXIIa) that contributes to bradykinin generation by converting prekallikrein to the protease plasma kallikrein (PKa). FXII conversion to FXIIa by autocatalysis or PKa-mediated cleavage is enhanced when the protein binds to negatively charged surfaces such as polymeric orthophosphate. FXII is composed of noncatalytic (heavy chain) and catalytic (light chain) regions. The heavy chain promotes FXII surface-binding and surface-dependent activation but restricts activation when FXII is not surface bound. From the N terminus, the heavy chain contains fibronectin type 2 (FN2), epidermal growth factor-1 (EGF1), fibronectin type 1 (FN1), EGF2, and kringle (KNG) domains and a proline-rich region. It shares this organization with its homolog, pro-hepatocyte growth factor activator (Pro-HGFA). To study the importance of heavy chain domains in FXII function, we prepared FXII with replacements of each domain with corresponding Pro-HGFA domains and tested them in activation and activity assays. EGF1 is required for surface-dependent FXII autoactivation and surface-dependent prekallikrein activation by FXIIa. KNG and FN2 are important for limiting FXII activation in the absence of a surface by a process that may require interactions between a lysine/arginine binding site on KNG and basic residues elsewhere on FXII. This interaction is disrupted by the lysine analog ε-aminocaproic acid. A model is proposed in which an ε-aminocaproic acid-sensitive interaction between the KNG and FN2 domains maintains FXII in a conformation that restricts activation. Upon binding to a surface through EGF1, the KNG/FN2-dependent mechanism is inactivated, exposing the FXII activation cleavage site.


Asunto(s)
Factor XII , Precalicreína , Ácido Aminocaproico , Coagulación Sanguínea , Factor XII/química , Fibronectinas/química , Lisina , Precalicreína/química , Precalicreína/metabolismo
5.
Blood ; 135(8): 558-567, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31800958

RESUMEN

Prekallikrein (PK) is the precursor of the trypsin-like plasma protease kallikrein (PKa), which cleaves kininogens to release bradykinin and converts the protease precursor factor XII (FXII) to the enzyme FXIIa. PK and FXII undergo reciprocal conversion to their active forms (PKa and FXIIa) by a process that is accelerated by a variety of biological and artificial surfaces. The surface-mediated process is referred to as contact activation. Previously, we showed that FXII expresses a low level of proteolytic activity (independently of FXIIa) that may initiate reciprocal activation with PK. The current study was undertaken to determine whether PK expresses similar activity. Recombinant PK that cannot be converted to PKa was prepared by replacing Arg371 with alanine at the activation cleavage site (PK-R371A, or single-chain PK). Despite being constrained to the single-chain precursor form, PK-R371A cleaves high-molecular-weight kininogen (HK) to release bradykinin with a catalytic efficiency ∼1500-fold lower than that of kallikrein cleavage of HK. In the presence of a surface, PK-R371A converts FXII to FXIIa with a specific activity ∼4 orders of magnitude lower than for PKa cleavage of FXII. These results support the notion that activity intrinsic to PK and FXII can initiate reciprocal activation of FXII and PK in solution or on a surface. The findings are consistent with the hypothesis that the putative zymogens of many trypsin-like proteases are actually active proteases, explaining their capacity to undergo processes such as autoactivation and to initiate enzyme cascades.


Asunto(s)
Coagulación Sanguínea , Bradiquinina/metabolismo , Precalicreína/metabolismo , Sustitución de Aminoácidos , Animales , Factor XII/metabolismo , Células HEK293 , Humanos , Quininógeno de Alto Peso Molecular/metabolismo , Ratones Endogámicos C57BL , Precalicreína/química , Precalicreína/genética , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Blood ; 133(10): 1152-1163, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30591525

RESUMEN

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.


Asunto(s)
Factor XII/química , Factor XIa/química , Angioedema Hereditario Tipo III/sangre , Angioedema Hereditario Tipo III/genética , Angioedemas Hereditarios , Animales , Arginina/química , Coagulación Sanguínea , Bradiquinina/sangre , Catálisis , Proteína Inhibidora del Complemento C1/química , Factor XIIa/química , Células HEK293 , Humanos , Quininógenos/sangre , Lisina/química , Ratones , Ratones Endogámicos C57BL , Calicreína Plasmática/química , Precalicreína/química , Unión Proteica , Proteínas Recombinantes/química , Propiedades de Superficie , Trombina/genética
8.
Glycobiology ; 28(10): 754-764, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30016441

RESUMEN

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.


Asunto(s)
Sulfatos de Condroitina/sangre , Sulfatos de Condroitina/metabolismo , Factor XIIa/metabolismo , Quininógenos/metabolismo , Precalicreína/metabolismo , Sulfatos de Condroitina/química , Factor XIIa/química , Humanos , Quininógenos/química , Precalicreína/química , Relación Estructura-Actividad
9.
Hematology ; 23(6): 346-350, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29165051

RESUMEN

OBJECTIVE: To investigate the structure-function relation in prekallikrein (PK) deficiency. PK is one of the proteins of the contact phase of blood coagulation which at the present time is the object of a revival of interest. METHODS: All patients with PK deficiency who had been investigated by molecular biology techniques are the object of the present investigation. Details of patients were obtained from personal files and a time-unlimited PubMed search. Only cases with a molecular-biology-based diagnosis were included. RESULTS: Twelve families were included. The total number of missense mutation was 10, together with 3 stop codons and 2 insertions. These mutations involved mainly exons 11 and 14. There were eight proved homozygotes and three compound heterozygotes. In one instance, homozygosity was probable but not proved. In nine cases, the defect was Type I, whereas it was Type II in the remaining three. No bleeding manifestations were present in 11 of the 12 probands. One proband had epistaxis, but she had hypertension. Altogether, four patients had hypertension and one of them had also two myocardial infarctions. CONCLUSIONS: Despite the paucity of cases, it was established that the majority of mutations involved the catalytic domain. It is auspicable that future reports of patients with this disorder should include molecular studies. This would certainly contribute to the understanding of the contact phase of blood coagulation.


Asunto(s)
Trastornos de la Coagulación Sanguínea/genética , Precalicreína/química , Precalicreína/deficiencia , Precalicreína/genética , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/diagnóstico , Trastornos de la Coagulación Sanguínea/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Femenino , Genotipo , Humanos , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Persona de Mediana Edad , Mutación , Precalicreína/metabolismo , Relación Estructura-Actividad , Adulto Joven
10.
Bioorg Khim ; 41(5): 559-64, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26762093

RESUMEN

Effects of extracellular proteases of Aspergillus ochraceus and Aspergillus terreus on plasma hemostasis proteins, consist of initiating the activation of prothrombin complex proteins, was detected. Was discovered, that A. ochraceus proteases have a direct influence on protein C and coagulation factor X, and A. terreus proteases causes their activation indirectly through kallikrein system stimulation. The ability of extracellular proteases of micromycetes activate prekallikrein in human blood plasma on the example of A. terreus was first demonstrated.


Asunto(s)
Aspergillus ochraceus/enzimología , Proteínas Sanguíneas/análisis , Hemostasis , Péptido Hidrolasas/química , Proteínas Sanguíneas/química , Factor X/análisis , Factor X/metabolismo , Humanos , Péptido Hidrolasas/metabolismo , Plasma/química , Precalicreína/análisis , Precalicreína/química , Precalicreína/metabolismo , Proteína C/análisis , Proteína C/metabolismo , Especificidad por Sustrato
11.
Curr Mol Med ; 14(9): 1173-85, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25324000

RESUMEN

Prolylcarboxypeptidase isoform 1 (PRCP1) is capable of regulating numerous autocrines and hormones, such as angiotensin II, angiotensin III, αMSH1-13, and DesArg(9) bradykinin. It does so by cleaving a C-terminal PRO-X bond. Recent work also indicates that the human PRCP1 activates plasma prekallikrein (PK) to kallikrein on endothelial cells through an uncharacterized mechanism. This study aims to identify PRCP1 binding interaction and cleavage site on PK. Recently, a cDNA encoding a novel splice variant of the human PRCP1 was identified. This isoform differed only in the N-terminal region of the deduced amino acid sequence. Using structural and functional studies, a combination of peptide mapping and site-directed mutagenesis approaches were employed to investigate the interaction of PRCP1 with PK. Three PRCP peptides, in decreasing order of potency, from 1) the N-terminus of the secreted protein, 2) spanning the opening of the active site pocket, and 3) in the dimerization region inhibit PRCP activation of PK on endothelial cells. Investigations also tested the hypothesis that PRCP cleavage site on PK is between its C-terminal Pro 637 (P(637)) and Ala 638 (A(638)). Recombinant forms of PK with C-terminal alanine mutagenesis or a stop codon is activated equally as wild type PK by PRCP. In conclusion, PRCP1 interacts with PK at multiple sites for PK activation. PRCP1 also enhances FXIIa activation of PK, suggesting that its activation site on PK is not identical to that of FXIIa.


Asunto(s)
Carboxipeptidasas/química , Precalicreína/química , Secuencia de Aminoácidos , Dominio Catalítico , Línea Celular , Activación Enzimática , Pruebas de Enzimas , Humanos , Cinética , Datos de Secuencia Molecular , Proteolisis , Especificidad por Sustrato
12.
J Thromb Haemost ; 12(9): 1513-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25039405

RESUMEN

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.


Asunto(s)
Factor XII/química , Polifosfatos/química , Clorometilcetonas de Aminoácidos/química , Aniones/química , Arginina/química , Plaquetas/metabolismo , Dominio Catalítico , Sulfato de Dextran/química , Disulfuros/química , Ácido Elágico/química , Precursores Enzimáticos/química , Hemostasis , Humanos , Caolín/química , Proteínas de Plantas/química , Precalicreína/química , Unión Proteica , ARN/química , Propiedades de Superficie
13.
Chem Immunol Allergy ; 100: 205-13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24925400

RESUMEN

The formation of bradykinin in plasma requires interaction of three proteins, namely coagulation factor XII (Hageman factor), prekallikrein and high-molecular-weight kininogen (HK). Prekallikrein and HK circulate as a bimolecular complex. Initiation of the cascade upon binding to negatively charged surfaces (or macromolecules) is dependent on factor XII autoactivation, conversion of prekallikrein to kallikrein, and a feedback activation of factor XII by kallikrein. The latter reaction is extremely rapid relative to factor XII autoactivation. The kallikrein then digests HK to liberate bradykinin. The natural surface appears to be vascular endothelial cells which express binding proteins for factor XII and HK, and activation can proceed along the cell surface. Recent findings demonstrate that prekallikrein has enzymatic activity separate from that of kallikrein such that it can stoichiometrically bind and cleave HK to liberate bradykinin. It is normally prevented from doing so by the plasma C1 inhibitor. Release of heat shock protein 90 (HSP-90) from endothelial cells can convert prekallikrein to kallikrein (stoichiometrically) within the prekallikrein-HK complex, even in the absence of factor XII, and the prekallikrein-HK complex can autoactivate to generate kallikrein if phosphate is the buffering ion. The effects of phosphate ion and HSP-90 are additive. Thus, an active site appears to be induced in prekallikrein by binding to HK and any of the aforementioned reactions can generate kallikrein prior to factor XII activation by autoactivation of the HK-PK complex. This brief review highlights the major discoveries made over the past 50 years which have led to our current concepts regarding the constituents and mechanisms of activation of the plasma bradykinin-forming cascade.


Asunto(s)
Bradiquinina/metabolismo , Bradiquinina/química , Células Endoteliales/metabolismo , Factor XII/química , Factor XII/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Precalicreína/química , Precalicreína/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
14.
PLoS One ; 7(11): e50399, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226277

RESUMEN

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.


Asunto(s)
Aptámeros de Nucleótidos/química , Coagulación Sanguínea , Coagulantes/química , Secuencias Invertidas Repetidas , Quininógeno de Alto Peso Molecular/química , Oligorribonucleótidos/química , Pruebas de Coagulación Sanguínea , Humanos , Conformación de Ácido Nucleico , Fosfoproteínas/química , Precalicreína/química , Unión Proteica , Proteína C/química , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , Proteínas de Unión al ARN/química , Relación Estructura-Actividad , Trombina/química , Factor A de Crecimiento Endotelial Vascular/química , Nucleolina
15.
Blood Coagul Fibrinolysis ; 23(8): 781-3, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22964768

RESUMEN

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.


Asunto(s)
Precalicreína/efectos de la radiación , Sonido/efectos adversos , Pruebas de Coagulación Sanguínea , Tampones (Química) , Compuestos Cromogénicos/química , Colorimetría , Glicerol , Humanos , Calicreínas/química , Calicreínas/efectos de la radiación , Oligopéptidos/química , Precalicreína/química , Pliegue de Proteína , Ultrasonografía/efectos adversos
16.
Blood ; 115(13): 2569-77, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20110423

RESUMEN

Factor XI (FXI) is the zymogen of an enzyme (FXIa) that contributes to hemostasis by activating factor IX. Although bleeding associated with FXI deficiency is relatively mild, there has been resurgence of interest in FXI because of studies indicating it makes contributions to thrombosis and other processes associated with dysregulated coagulation. FXI is an unusual dimeric protease, with structural features that distinguish it from vitamin K-dependent coagulation proteases. The recent availability of crystal structures for zymogen FXI and the FXIa catalytic domain have enhanced our understanding of structure-function relationships for this molecule. FXI contains 4 "apple domains" that form a disk structure with extensive interfaces at the base of the catalytic domain. The characterization of the apple disk structure, and its relationship to the catalytic domain, have provided new insight into the mechanism of FXI activation, the interaction of FXIa with the substrate factor IX, and the binding of FXI to platelets. Analyses of missense mutations associated with FXI deficiency have provided additional clues to localization of ligand-binding sites on the protein surface. Together, these data will facilitate efforts to understand the physiology and pathology of this unusual protease, and development of therapeutics to treat thrombotic disorders.


Asunto(s)
Factor XI/fisiología , Coagulación Sanguínea , Plaquetas/metabolismo , Dominio Catalítico , Dimerización , Activación Enzimática , Evolución Molecular , Factor IX/química , Factor XI/antagonistas & inhibidores , Factor XI/química , Factor XI/genética , Deficiencia del Factor XI/sangre , Deficiencia del Factor XI/genética , Predicción , Humanos , Modelos Moleculares , Mutación , Glicoproteínas de Membrana Plaquetaria/fisiología , Precalicreína/química , Precalicreína/genética , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Relación Estructura-Actividad
17.
J Allergy Clin Immunol ; 124(1): 143-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19342086

RESUMEN

BACKGROUND: Bradykinin formation typically requires interaction of Factor XII, prekallikrein (PK), and high-molecular-weight kininogen (HK) with negatively charged exogenous initiators or cell-surface proteins. Approximately 85% of plasma PK circulates as a complex with HK. Nonenzymatic cell-derived initiators, such as heat shock protein 90, can activate the HK-PK complex to generate kallikrein, bradykinin, and cleaved HK, even in the absence of Factor XII. OBJECTIVE: We sought to determine whether PK, without activation to kallikrein, can digest HK to release bradykinin. METHODS: Kallikrein was measured by using a chromogenic assay, and bradykinin levels were determined by ELISA. Cleavage of PK and HK were assessed by SDS-PAGE and Western blot analysis. RESULTS: Cleavage of HK by PK is demonstrated without any conversion of PK to kallikrein. HK cleavage by PK is distinguished from that of kallikrein by the following: (1) stoichiometric activation of HK by PK with release of bradykinin proportional to the PK input; (2) inhibition of PK cleavage of HK by corn trypsin inhibitor, which has no effect on kallikrein; and (3) inhibition of PK cleavage of HK by a peptide derived from HK, which inhibits binding of PK to HK. The same peptide has no effect on kallikrein activation of HK. C1 inhibitor (C1INH), the major control protein of the plasma bradykinin-forming cascade, inhibits PK cleavage of HK. CONCLUSION: PK is an enzyme that can cleave HK to release bradykinin, and this reaction is inhibited by C1INH. This might account, in part, for circulating bradykinin levels and initiation of kinin formation in C1INH deficiency.


Asunto(s)
Proteína Inhibidora del Complemento C1/metabolismo , Factor XII/química , Quininógeno de Alto Peso Molecular/química , Quininógeno de Alto Peso Molecular/metabolismo , Precalicreína/metabolismo , Western Blotting , Bradiquinina/química , Catálisis , Electroforesis en Gel de Poliacrilamida , Humanos , Precalicreína/antagonistas & inhibidores , Precalicreína/química
18.
Anal Biochem ; 390(1): 88-90, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19351526

RESUMEN

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.


Asunto(s)
Electroforesis en Gel Bidimensional/métodos , Plasminógeno/química , Precalicreína/química , Proteína S/química , Sulfatos/química , Tirosina/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Isótopos de Azufre/química
19.
Thromb Res ; 122(2): 257-63, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18177925

RESUMEN

INTRODUCTION: This study was designed to investigate whether fibrinogen, soluble desAA-fibrin, and insoluble desAABB-fibrin are able to induce clotting by triggering the plasma contact activation system when adsorbed to polystyrene. MATERIALS AND METHODS: The above-mentioned substances were individually prepared on polystyrene meshwork squares, and then exposed to a purified FXII solution or non-calcium containing plasma (citrated and dialyzed normal pooled plasma) in polystyrene cuvettes coated with surface-immobilized heparin, to completely block contact activation and the coagulation mechanism that might be induced by the cuvette surfaces. Sodium glass beads were used as the reference material. RESULTS: On exposure to purified FXII solution and plasma, all the tested materials adsorbed and activated FXII to varying degrees. This activation led to the formation of FXIa in the exposed plasma, with the highest activation occurring upon exposure to glass, desAA-fibrin and desAABB-fibrin and the lowest upon exposure to fibrinogen-adsorbed or unmodified polystyrene meshwork squares. Following recalcification, in cuvettes with surface-immobilized heparin, a spectrophotometric assay showed that the surface-exposed plasma aliquots clotted within 5 min after contact with glass, within 10 to 15 min after contact with the two forms of fibrin, and somewhat longer after contact with adsorbed fibrinogen. The longest lag phase, close to 20 min, occurred in plasma exposed to unmodified polystyrene meshwork. Whole blood deposited in surface heparinized cuvettes directly from the cubital vein did not clot during the observation time (2 h). CONCLUSIONS: These results indicate that domains induced by conformational changes in adsorbed fibrinogen and fibrin are capable of activating adsorbed proenzymes and that various forms of fibrin are considerably stronger activators of the contact activation system than are adsorbed fibrinogen or a polystyrene meshwork. The delayed coagulation in plasma exposed to the unmodified polystyrene meshwork can be explained by a two-step process: first, adsorption of fibrinogen, and second, activation of FXII. Under our experimental conditions, the adsorption and activation of FXII on fibrinogen and fibrin seems to be an important mechanism for triggering coagulation.


Asunto(s)
Fibrina/química , Fibrinógeno/química , Fragmentos de Péptidos/química , Adsorción , Coagulación Sanguínea , Factor XII/química , Vidrio , Heparina/química , Humanos , Poliestirenos/química , Precalicreína/química , Sodio/química , Propiedades de Superficie , Trombina/antagonistas & inhibidores
20.
Biochemistry ; 46(50): 14450-60, 2007 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-18020374

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Factor XIa/química , Factor XIa/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Línea Celular , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Ensayo de Inmunoadsorción Enzimática , Factor XI/química , Factor XI/genética , Factor XI/metabolismo , Factor XIIa/química , Factor XIIa/genética , Factor XIIa/metabolismo , Factor XIa/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Precalicreína/química , Precalicreína/genética , Precalicreína/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...