Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
J Thromb Haemost ; 21(6): 1567-1579, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36863563

RESUMEN

BACKGROUND: During plasma contact activation, factor XII (FXII) binds to surfaces through its heavy chain and undergoes conversion to the protease FXIIa. FXIIa activates prekallikrein and factor XI (FXI). Recently, we showed that the FXII first epidermal growth factor-1 (EGF1) domain is required for normal activity when polyphosphate is used as a surface. OBJECTIVES: The aim of this study was to identify amino acids in the FXII EGF1 domain required for polyphosphate-dependent FXII functions. METHODS: FXII with alanine substitutions for basic residues in the EGF1 domain were expressed in HEK293 fibroblasts. Wild-type FXII (FXII-WT) and FXII containing the EGF1 domain from the related protein Pro-HGFA (FXII-EGF1) were positive and negative controls. Proteins were tested for their capacity to be activated, and to activate prekallikrein and FXI, with or without polyphosphate, and to replace FXII-WT in plasma clotting assays and a mouse thrombosis model. RESULTS: FXII and all FXII variants were activated similarly by kallikrein in the absence of polyphosphate. However, FXII with alanine replacing Lys73, Lys74, and Lys76 (FXII-Ala73,74,76) or Lys76, His78, and Lys81 (FXII-Ala76,78,81) were activated poorly in the presence of polyphosphate. Both have <5% of normal FXII activity in silica-triggered plasma clotting assays and have reduced binding affinity for polyphosphate. Activated FXIIa-Ala73,74,76 displayed profound defects in surface-dependent FXI activation in purified and plasma systems. FXIIa-Ala73,74,76 reconstituted FXII-deficient mice poorly in an arterial thrombosis model. CONCLUSION: FXII Lys73, Lys74, Lys76, and Lys81 form a binding site for polyanionic substances such as polyphosphate that is required for surface-dependent FXII function.


Asunto(s)
Factor XII , Trombosis , Humanos , Animales , Ratones , Factor XII/metabolismo , Precalicreína/metabolismo , Polifosfatos , Células HEK293 , Factor XI/metabolismo , Factor XIIa/metabolismo
2.
J Thromb Thrombolysis ; 54(1): 11-14, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34993714

RESUMEN

The contact system activation can play a role in microthrombus formation of disseminated intravascular coagulation (DIC). This study investigated whether the activity of prekallikrein and high-molecular-weight kininogen (HMWK) correlated DIC progression. Contact system factors (prekallikrein, HMWK, activated factor XII), coagulation factors (IX, XI, XII) and tissue factor were measured in 140 patients who clinically suspected of having DIC. Prekallikrein and HMWK activity levels showed significant linear relationships with DIC score and antithrombin level, whereas prekallikrein and HMWK antigen levels did not. The activated factor XII, factor XII, factor XI and tissue factor were significant risk factors of overt-DIC. This finding suggests that consumption of prekallikrein and HMWK contributes to microvascular thrombosis in DIC. Measurements of prekallikrein and HMWK activity could be used as potential diagnostic markers for overt-DIC.


Asunto(s)
Coagulación Intravascular Diseminada , Trombosis , Coagulación Intravascular Diseminada/diagnóstico , Factor XIIa , Humanos , Quininógeno de Alto Peso Molecular , Quininógenos/fisiología , Precalicreína , Factores de Riesgo , Tromboplastina
3.
Blood ; 139(18): 2816-2829, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35100351

RESUMEN

Patients with hereditary angioedema (HAE) experience episodes of bradykinin (BK)-induced swelling of skin and mucosal membranes. The most common cause is reduced plasma activity of C1 inhibitor, the main regulator of the proteases plasma kallikrein (PKa) and factor XIIa (FXIIa). Recently, patients with HAE were described with a Lys311 to glutamic acid substitution in plasminogen (Plg), the zymogen of the protease plasmin (Plm). Adding tissue plasminogen activator to plasma containing Plg-Glu311 vs plasma containing wild-type Plg (Plg-Lys311) results in greater BK generation. Similar results were obtained in plasma lacking prekallikrein or FXII (the zymogens of PKa and FXIIa) and in normal plasma treated with a PKa inhibitor, indicating Plg-Glu311 induces BK generation independently of PKa and FXIIa. Plm-Glu311 cleaves high and low molecular weight kininogens (HK and LK, respectively), releasing BK more efficiently than Plm-Lys311. Based on the plasma concentrations of HK and LK, the latter may be the source of most of the BK generated by Plm-Glu311. The lysine analog ε-aminocaproic acid blocks Plm-catalyzed BK generation. The Glu311 substitution introduces a lysine-binding site into the Plg kringle 3 domain, perhaps altering binding to kininogens. Plg residue 311 is glutamic acid in most mammals. Glu311 in patients with HAE, therefore, represents reversion to the ancestral condition. Substantial BK generation occurs during Plm-Glu311 cleavage of human HK, but not mouse HK. Furthermore, mouse Plm, which has Glu311, did not liberate BK from human kininogens more rapidly than human Plg-Lys311. This indicates Glu311 is pathogenic in the context of human Plm when human kininogens are the substrates.


Asunto(s)
Angioedemas Hereditarios , Angioedemas Hereditarios/genética , Angioedemas Hereditarios/patología , Animales , Bradiquinina/metabolismo , Factor XIIa/metabolismo , Fibrinolisina , Ácido Glutámico , Humanos , Quininógenos/metabolismo , Lisina , Mamíferos/metabolismo , Ratones , Calicreína Plasmática , Plasminógeno/genética , Plasminógeno/metabolismo , Activador de Tejido Plasminógeno
4.
J Thromb Haemost ; 20(4): 821-832, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34967109

RESUMEN

BACKGROUND: Previously, we showed that histidine-rich glycoprotein (HRG) binds factor (F) XIIa with high affinity, inhibits FXII autoactivation and FXIIa-mediated activation of FXI, and attenuates ferric chloride-induced arterial thrombosis in mice. Therefore, HRG downregulates the contact pathway in vitro and in vivo. OBJECTIVE: To identify the domains on HRG responsible for contact pathway inhibition. METHODS: Recombinant HRG domain constructs (N-terminal [N1, N2, and N1N2], proline-rich regions, histidine-rich region [HRR], and C-terminal) were expressed and purified. The affinities of plasma-derived HRG, HRG domain constructs, and synthetic HRR peptides for FXII, FXIIa, ß-FXIIa, and polyphosphate (polyP) were determined using surface plasmon resonance, and their effects on polyP-induced FXII autoactivation, FXIIa-mediated activation of FXI and prekallikrein, the activated partial thromboplastin time (APTT), and thrombin generation were examined. RESULTS: HRG and HRG domain constructs bind FXIIa, but not FXII or ß-FXII. HRR, N1, and N1N2 bind FXIIa with affinities comparable with that of HRG, whereas the remaining domains bind with lower affinity. Synthetic HRR peptides bind FXIIa and polyP with high affinity. HRG and HRR significantly inhibit FXII autoactivation and prolong the APTT. Like HRG, synthetic HRR peptides inhibit FXII autoactivation, attenuate FXIIa-mediated activation of prekallikrein and FXI, prolong the APTT, and attenuate thrombin generation. CONCLUSION: The interaction of HRG with FXIIa and polyP is predominantly mediated by the HRR domain. Like intact HRG, HRR downregulates the contact pathway and contributes to HRG-mediated down regulation of coagulation.


Asunto(s)
Precalicreína , Trombina , Animales , Factor XII/metabolismo , Factor XIIa/metabolismo , Humanos , Ratones , Péptidos/farmacología , Polifosfatos , Precalicreína/metabolismo , Proteínas , Trombina/metabolismo
5.
J Am Chem Soc ; 143(44): 18481-18489, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34723512

RESUMEN

Cyclotides are plant-derived peptides with complex structures shaped by their head-to-tail cyclic backbone and cystine knot core. These structural features underpin the native bioactivities of cyclotides, as well as their beneficial properties as pharmaceutical leads, including high proteolytic stability and cell permeability. However, their inherent structural complexity presents a challenge for cyclotide engineering, particularly for accessing libraries of sufficient chemical diversity to design potent and selective cyclotide variants. Here, we report a strategy using mRNA display enabling us to select potent cyclotide-based FXIIa inhibitors from a library comprising more than 1012 members based on the cyclotide scaffold of Momordica cochinchinensis trypsin inhibitor-II (MCoTI-II). The most potent and selective inhibitor, cMCoFx1, has a pM inhibitory constant toward FXIIa with greater than three orders of magnitude selectivity over related serine proteases, realizing specific inhibition of the intrinsic coagulation pathway. The cocrystal structure of cMCoFx1 and FXIIa revealed interactions at several positions across the contact interface that conveyed high affinity binding, highlighting that such cyclotides are attractive cystine knot scaffolds for therapeutic development.


Asunto(s)
Proteínas Sanguíneas/farmacología , Ciclotidas/farmacología , Factor XIIa/metabolismo , Proteínas Sanguíneas/química , Ciclotidas/química , Factor XIIa/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos
6.
Blood Adv ; 5(22): 4741-4751, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34597365

RESUMEN

Extracellular vesicles (EV) have been implicated in diverse biological processes, including intracellular communication, transport of nucleic acids, and regulation of vascular function. Levels of EVs are elevated in cancer, and studies suggest that EV may stimulate thrombosis in patients with cancer through expression of tissue factor. However, limited data also implicate EV in the activation of the contact pathway of coagulation through activation of factor XII (FXII) to FXIIa. To better define the ability of EV to initiate contact activation, we compared the ability of EV derived from different cancer cell lines to activate FXII. EV from all cell lines activated FXII, with those derived from pancreatic and lung cancer cell lines demonstrating the most potent activity. Concordant with the activation of FXII, EV induced the cleavage of high molecular weight kininogen (HK) to cleaved kininogen. We also observed that EVs from patients with cancer stimulated FXII activation and HK cleavage. To define the mechanisms of FXII activation by EV, EV were treated with calf intestinal alkaline phosphatase or Escherichia coli exopolyphosphatase to degrade polyphosphate; this treatment blocked binding of FXII to EVs and the ability of EV to mediate FXII activation. In vivo, EV induced pulmonary thrombosis in wild-type mice, with protection conferred by a deficiency in FXII, HK, or prekallikrein. Moreover, pretreatment of EVs with calf intestinal alkaline phosphatase inhibited their prothrombotic effect. These results indicate that polyphosphate mediates the binding of contact factors to EV and that EV-associated polyphosphate may contribute to the prothrombotic effects of EV in cancer.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Animales , Factor XII , Factor XIIa , Humanos , Ratones , Polifosfatos , Precalicreína
7.
Nat Commun ; 12(1): 5596, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34552086

RESUMEN

Contact activation refers to the process of surface-induced activation of factor XII (FXII), which initiates blood coagulation and is captured by the activated partial thromboplastin time (aPTT) assay. Here, we show the mechanism and diagnostic implications of FXII contact activation. Screening of recombinant FXII mutants identified a continuous stretch of residues Gln317-Ser339 that was essential for FXII surface binding and activation, thrombin generation and coagulation. Peptides spanning these 23 residues competed with surface-induced FXII activation. Although FXII mutants lacking residues Gln317-Ser339 were susceptible to activation by plasmin and plasma kallikrein, they were ineffective in supporting arterial and venous thrombus formation in mice. Antibodies raised against the Gln317-Ser339 region induced FXII activation and triggered controllable contact activation in solution leading to thrombin generation by the intrinsic pathway of coagulation. The antibody-activated aPTT allows for standardization of particulate aPTT reagents and for sensitive monitoring of coagulation factors VIII, IX, XI.


Asunto(s)
Coagulación Sanguínea , Factor XII/química , Factor XII/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/farmacología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Factor XII/genética , Factor XII/inmunología , Factor XIIa/metabolismo , Ratones , Mutación , Tiempo de Tromboplastina Parcial/normas , Péptidos/química , Péptidos/genética , Péptidos/inmunología , Péptidos/metabolismo , Trombosis/diagnóstico , Trombosis/genética , Trombosis/metabolismo
8.
Food Funct ; 12(20): 10136-10146, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34528647

RESUMEN

A novel anticoagulant peptide (IEELEEELEAER) derived from oyster (Crassostrea gigas) was discovered by combining the emerging bioinformatics with the classical enzymolysis approach. The anticoagulant peptide drastically reduced the extrinsic clotting activity (49% residual PT activity) and impaired the intrinsic clotting activity (77% residual PT activity). Consistent with the clotting data, the thrombin peak height reduced to 88.7 from 123.4 nM, and the thrombin generation time delayed to 5.32 from 4.42 min when an extrinsic trigger was applied. The inhibitory kinetics of FXIa, FIXa, FXa, FIIa, and APC in a purified component system rationally explained the reduction of the extrinsic clotting activity and impairment of thrombin generation. Besides the inhibition of FXa and FIIa activity, the activation processes of FX and FII by an intrinsic/extrinsic tenase complex and prothrombinase were also damaged. The anticoagulant activity in the plasma system was the result of comprehensive inhibition of various factors. The research provided a frame for anticoagulant evaluation and inhibitory mechanism of bioactive peptides from food products.


Asunto(s)
Anticoagulantes/farmacología , Biología Computacional/métodos , Crassostrea/química , Péptidos/farmacología , Animales , Anticoagulantes/química , Coagulación Sanguínea/efectos de los fármacos , Factor XIIa/metabolismo , Factor XIa/metabolismo , Factor Xa/metabolismo , Humanos , Cinética , Péptidos/química , Trombina/metabolismo , Tiempo de Trombina/métodos , Tromboplastina/metabolismo
9.
J Thromb Haemost ; 19(9): 2235-2247, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34060720

RESUMEN

BACKGROUND: Factor XII (FXII) is a serine protease that participates in the intrinsic coagulation pathway. Several studies have shown that plasma FXII exerts a deleterious role in cerebral ischemia and traumatic brain injury by promoting thrombo-inflammation. Nevertheless, the impact of FXII on neuronal cell fate remains unknown. OBJECTIVES: We investigated the role of FXII and FXIIa in neuronal injury and apoptotic cell death. METHODS: We tested the neuroprotective roles of FXII and FXIIa in an experimental model of neuronal injury induced by stereotaxic intracerebral injection of N-methyl-D-aspartic acid (NMDA) in vivo and in a model of apoptotic death of murine primary neuronal cultures through serum deprivation in vitro. RESULTS: Here, we found that exogenous FXII and FXIIa reduce brain lesions induced by NMDA injection in vivo. Furthermore, FXII protects cultured neurons from apoptosis through a growth factor--like effect. This mechanism was triggered by direct interaction with epidermal growth factor (EGF) receptor and subsequent activation of this receptor. Interestingly, the "proteolytically" active and two-chain form of FXII, FXIIa, exerts its protective effects by an alternative signaling pathway. FXIIa activates the pro-form of hepatocyte growth factor (HGF) into HGF, which in turn activated the HGF receptor (HGFR) pathway. CONCLUSION: This study describes two novel mechanisms of action of FXII and identifies neurons as target cells for the protective effects of single and two-chain forms of FXII. Therefore, inhibition of FXII in neurological disorders may have deleterious effects by preventing its beneficial effects on neuronal survival.


Asunto(s)
Factor XII , Proteínas Proto-Oncogénicas c-met , Animales , Apoptosis , Coagulación Sanguínea , Factor XIIa , Ratones , Neuronas
10.
J Med Chem ; 63(21): 13159-13186, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33089691

RESUMEN

We herein report the conventional and microscale parallel synthesis of selective inhibitors of human blood coagulation factor XIIa and thrombin exhibiting a 1,2,4-triazol-5-amine scaffold. Structural variations of this scaffold allowed identifying derivative 21i, a potent 29 nM inhibitor of FXIIa, with improved selectivity over other tested serine proteases and also finding compound 21m with 27 nM inhibitory activity toward thrombin. For the first time, acylated 1,2,4-triazol-5-amines were proved to have anticoagulant properties and the ability to affect thrombin- and cancer-cell-induced platelet aggregation. Performed mass spectrometric analysis and molecular modeling allowed us to discover previously unknown interactions between the synthesized inhibitors and the active site of FXIIa, which uncovered the mechanistic details of FXIIa inhibition. Synthesized compounds represent a promising starting point for the development of novel antithrombotic drugs or chemical tools for studying the role of FXIIa and thrombin in physiological and pathological processes.


Asunto(s)
Aminas/química , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor XIIa/metabolismo , Trombina/metabolismo , Aminas/síntesis química , Aminas/metabolismo , Anticoagulantes/síntesis química , Anticoagulantes/metabolismo , Sitios de Unión , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Factor XIIa/antagonistas & inhibidores , Humanos , Concentración 50 Inhibidora , Simulación de Dinámica Molecular , Agregación Plaquetaria/efectos de los fármacos , Relación Estructura-Actividad , Trombina/antagonistas & inhibidores , Triazoles/química
11.
Cardiovasc Eng Technol ; 11(4): 448-455, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32607901

RESUMEN

PURPOSE: Crosslinked poly(vinyl alcohol) (PVA) is a biomaterial that can be used for multiple cardiovascular applications. The success of implanted biomaterials is contingent on the properties of the material. A crucial consideration for blood-contacting devices is their potential to incite thrombus formation, which is dependent on the material surface properties. The goal of this study was to quantify the effect of different crosslinking methods of PVA hydrogels on in vitro thrombogenicity. METHODS: PVA was manufactured using three different crosslinking methods: 30% sodium trimetaphosphate (STMP), three 24 h freeze-thaw cycles (FT), and 2% glutaraldehyde-crosslinked (GA) to produce STMP-PVA, FT-PVA and GA-PVA, respectively. Expanded polytetrafluoroethylene (ePTFE) was used as a clinical control. As markers of thrombus formation, the degree of coagulation factor (F) XII activation, fibrin formation, and platelet adhesion were measured. RESULTS: The GA-PVA material increased FXII activation in the presence of cofactors compared to vehicle and increase platelet adhesion compared to other PVA surfaces. The STMP-PVA and FT-PVA materials had equivalent degrees of FXII activation, fibrin formation and platelet adhesion. CONCLUSION: This work supports crosslinker dependent thrombogenicity of PVA hydrogels and advances our understanding of how the manufacturing of a PVA hydrogel affects its hemocompatibility.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Congelación , Glutaral/química , Polifosfatos/química , Alcohol Polivinílico/química , Trombosis/prevención & control , Materiales Biocompatibles , Coagulación Sanguínea , Prótesis Vascular , Reactivos de Enlaces Cruzados/toxicidad , Factor XIIa/metabolismo , Fibrinólisis , Congelación/efectos adversos , Glutaral/toxicidad , Oclusión de Injerto Vascular/sangre , Oclusión de Injerto Vascular/etiología , Oclusión de Injerto Vascular/prevención & control , Humanos , Hidrogeles , Ensayo de Materiales , Adhesividad Plaquetaria , Polifosfatos/toxicidad , Alcohol Polivinílico/toxicidad , Diseño de Prótesis , Propiedades de Superficie , Trombosis/sangre , Trombosis/etiología
12.
Circ Res ; 127(5): 651-663, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32450779

RESUMEN

RATIONALE: Epidemiological studies have identified an associate between iron deficiency (ID) and the use of oral contraceptives (CC) and ischemic stroke (IS). To date, however, the underlying mechanism remains poorly understood. Both ID and CC have been demonstrated to upregulate the level and iron-binding ability of Tf (transferrin), with our recent study showing that this upregulation can induce hypercoagulability by potentiating FXIIa/thrombin and blocking antithrombin-coagulation proteases interactions. OBJECTIVE: To investigate whether Tf mediates IS associated with ID or CC and the underlying mechanisms. METHODS AND RESULTS: Tf levels were assayed in the plasma of IS patients with a history of ID anemia, ID anemia patients, venous thromboembolism patients using CC, and ID mice, and in the cerebrospinal fluid of some IS patients. Effects of ID and estrogen administration on Tf expression and coagulability and the underlying mechanisms were studied in vivo and in vitro. High levels of Tf and Tf-thrombin/FXIIa complexes were found in patients and ID mice. Both ID and estrogen upregulated Tf through hypoxia and estrogen response elements located in the Tf gene enhancer and promoter regions, respectively. In addition, ID, administration of exogenous Tf or estrogen, and Tf overexpression promoted platelet-based thrombin generation and hypercoagulability and thus aggravated IS. In contrast, anti-Tf antibodies, Tf knockdown, and peptide inhibitors of Tf-thrombin/FXIIa interaction exerted anti-IS effects in vivo. CONCLUSIONS: Our findings revealed that certain factors (ie, ID and CC) upregulating Tf are risk factors of thromboembolic diseases decipher a previously unrecognized mechanistic association among ID, CC, and IS and provide a novel strategy for the development of anti-IS medicine by interfering with Tf-thrombin/FXIIa interactions.


Asunto(s)
Anemia Ferropénica/complicaciones , Coagulación Sanguínea , Anticonceptivos Hormonales Orales/efectos adversos , Estrógenos/toxicidad , Accidente Cerebrovascular Isquémico/etiología , Trombofilia/etiología , Transferrina/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia Ferropénica/sangre , Anemia Ferropénica/diagnóstico , Animales , Biomarcadores/sangre , Estudios de Casos y Controles , Línea Celular , Modelos Animales de Enfermedad , Factor XIIa/metabolismo , Femenino , Humanos , Accidente Cerebrovascular Isquémico/sangre , Accidente Cerebrovascular Isquémico/diagnóstico , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo , Trombina/metabolismo , Trombofilia/sangre , Trombofilia/diagnóstico , Regulación hacia Arriba , Adulto Joven
13.
Thromb Haemost ; 120(3): 400-411, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31940673

RESUMEN

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.


Asunto(s)
Precursores Enzimáticos/química , Factor XII/química , Fibrinolisina/química , Fibronectinas/química , Calicreínas/química , Animales , Sitios de Unión , Coagulación Sanguínea , Bradiquinina/química , Dominio Catalítico , Bovinos , Factor XIIa/química , Fibronectinas/sangre , Células HEK293 , Humanos , Calicreínas/sangre , Caolín/química , Polifosfatos/química , Unión Proteica , Dominios Proteicos
14.
J Am Coll Cardiol ; 74(17): 2178-2189, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31648711

RESUMEN

BACKGROUND: Exposure of blood to polyanionic artificial surfaces, for example, during cardiopulmonary bypass (CPB), induces a highly procoagulant condition requiring strong anticoagulation. Unfractionated heparin (UFH) is currently used during CPB but can lead to serious bleeding complications or development of a hypercoagulable state culminating in life-threatening thrombosis, highlighting the need for safer antithrombotics. Ixodes ricinus contact phase inhibitor (Ir-CPI) is a protein expressed by I. ricinus ticks, which specifically inhibits both factors XIIa and XIa, 2 factors contributing to thrombotic disease while playing a limited role in hemostasis. OBJECTIVES: This study assessed the antithrombotic activity of Ir-CPI in animal contact phase-initiated thrombosis models, including CPB. The safety of Ir-CPI also was evaluated. METHODS: The authors evaluated the antithrombotic activity of Ir-CPI by using in vitro catheter-induced clotting assays and rabbit experimental models of catheter occlusion and arteriovenous shunt. During CPB with cardiac surgery in sheep, the clinical applicability of Ir-CPI was investigated and its efficacy compared to that of UFH using an uncoated system suitable for adult therapy. Taking advantage of the similar hemostatic properties of pigs and humans, the authors performed pig liver bleeding assays to evaluate the safety of Ir-CPI. RESULTS: Ir-CPI prevented clotting in catheter and arteriovenous shunt rabbit models. During CPB, Ir-CPI was as efficient as UFH in preventing clot formation within the extracorporeal circuit and maintained physiological parameters during and post-surgery. Unlike UFH, Ir-CPI did not promote bleeding. CONCLUSIONS: Preclinical animal models used in this study showed that Ir-CPI is an effective and safe antithrombotic agent that provides a clinically relevant approach to thrombosis prevention in bypass systems, including highly thrombogenic CPB.


Asunto(s)
Anticoagulantes/uso terapéutico , Puente Cardiopulmonar/métodos , Factor XIIa/antagonistas & inhibidores , Factor XIa/antagonistas & inhibidores , Animales , Coagulación Sanguínea , Proteínas Sanguíneas/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Fibrinolíticos , Hemorragia/tratamiento farmacológico , Hemostasis , Heparina/uso terapéutico , Humanos , Ixodes , Conejos , Ovinos , Porcinos , Trombosis/prevención & control , Garrapatas
15.
J Mol Graph Model ; 89: 225-233, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30921556

RESUMEN

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.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Factor XIIa/antagonistas & inhibidores , Factor XIIa/química , Péptidos/química , Péptidos/farmacología , Porphyra/química , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Conformación Molecular , Extractos Vegetales , Relación Estructura-Actividad
16.
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
17.
Arterioscler Thromb Vasc Biol ; 38(8): 1748-1760, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30354195

RESUMEN

Objective- Terminal complications of bacterial sepsis include development of disseminated intravascular consumptive coagulopathy. Bacterial constituents, including long-chain polyphosphates (polyP), have been shown to activate the contact pathway of coagulation in plasma. Recent work shows that activation of the contact pathway in flowing whole blood promotes thrombin generation and platelet activation and consumption distal to thrombus formation ex vivo and in vivo. Here, we sought to determine whether presence of long-chain polyP or bacteria in the bloodstream promotes platelet activation and consumption in a coagulation factor (F)XII-dependent manner. Approach and Results- Long-chain polyP promoted platelet P-selectin expression, microaggregate formation, and platelet consumption in flowing whole blood in a contact activation pathway-dependent manner. Moreover, long-chain polyP promoted local fibrin formation on collagen under shear flow in a FXI-dependent manner. Distal to the site of thrombus formation, platelet consumption was dramatically enhanced in the presence of long-chain polyP in the blood flow in a FXI- and FXII-dependent manner. In a murine model, long-chain polyP promoted platelet deposition and fibrin generation in lungs in a FXII-dependent manner. In a nonhuman primate model of bacterial sepsis, pre-treatment of animals with an antibody blocking FXI activation by FXIIa reduced lethal dose100 Staphylococcus aureus-induced platelet and fibrinogen consumption. Conclusions- This study demonstrates that bacterial-type long-chain polyP promotes platelet activation in a FXII-dependent manner in flowing blood, which may contribute to sepsis-associated thrombotic processes, consumptive coagulopathy, and thrombocytopenia.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Factor XII/metabolismo , Factor XIIa/metabolismo , Activación Plaquetaria/efectos de los fármacos , Polifosfatos/toxicidad , Trombosis/inducido químicamente , Animales , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Factor XII/genética , Factor XIIa/genética , Femenino , Fibrina/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Papio ursinus , Precalicreína/genética , Precalicreína/metabolismo , Embolia Pulmonar/sangre , Embolia Pulmonar/inducido químicamente , Embolia Pulmonar/genética , Sepsis/sangre , Sepsis/microbiología , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/microbiología , Trombosis/sangre , Trombosis/genética , Calicreínas de Tejido/genética , Calicreínas de Tejido/metabolismo
18.
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
19.
Chembiochem ; 19(18): 1934-1938, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-29944773

RESUMEN

A broadly applicable one-pot methodology for the facile transformation of linear peptides into tetracyclic peptides through a chemoenzymatic peptide synthesis/chemical ligation of peptides onto scaffolds/copper(I)-catalyzed reaction (CEPS/CLIPS/CuAAC; "triple-C") locking methodology is reported. Linear peptides with varying lengths (≥14 amino acids), comprising two cysteines and two azidohomoalanines (Aha), were efficiently cyclized head-to-tail by using the peptiligase variant omniligase-1 (CEPS). Subsequent ligation-cyclization with tetravalent (T41/2 ) scaffolds containing two bromomethyl groups (CLIPS) and two alkyne functionalities (CuAAC) yielded isomerically pure tetracyclic peptides. Sixteen different functional tetracycles, derived from bicyclic inhibitors against urokinase plasminogen activator (uPA) and coagulation factor XIIa (FXIIa), were successfully synthesized and their bioactivities evaluated. Two of these (FF-T41/2 ) exhibited increased inhibitory activity against FXIIa, compared with a bicyclic control peptide. The corresponding hetero-bifunctional variants (UF/FU-T41/2 ), with a single copy of each inhibitory sequence, exhibited micromolar activities against both uPA and FXIIa; thus illustrating the potential of the "bifunctional tetracyclic peptide" inhibitor concept.


Asunto(s)
Péptidos Cíclicos/síntesis química , Péptidos/química , Alanina/análogos & derivados , Alanina/química , Secuencia de Aminoácidos , Técnicas Químicas Combinatorias , Ciclización , Cisteína/química , Factor XIIa/antagonistas & inhibidores , Humanos , Modelos Moleculares , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
20.
J Thromb Haemost ; 16(9): 1674-1685, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29920929

RESUMEN

The plasma contact system contributes to thrombosis in experimental models. Even though our standard blood coagulation tests are prolonged when plasma lacks contact factors, this enzyme system appears to have a minor (if any) role in hemostasis. In this review, we explore the clinical phenotype of C1 esterase inhibitor (C1-INH) deficiency. C1-INH is the key plasma inhibitor of the contact system enzymes, and its deficiency causes hereditary angioedema (HAE). This inflammatory disorder is characterized by recurrent aggressive attacks of tissue swelling that occur at unpredictable locations throughout the body. Bradykinin, which is considered to be a byproduct of the plasma contact system during in vitro coagulation, is the main disease mediator in HAE. Surprisingly, there is little evidence for thrombotic events in HAE patients, suggesting mechanistic uncoupling from the intrinsic pathway of coagulation. In addition, it is questionable whether a surface is responsible for contact system activation in HAE. In this review, we discuss the clinical phenotype, disease modifiers and diagnostic challenges of HAE. We subsequently describe the underlying biochemical mechanisms and contributing disease mediators. Furthermore, we review three types of HAE that are not caused by C1-INH inhibitor deficiency. Finally, we propose a central enzymatic axis that we hypothesize to be responsible for bradykinin production in health and disease.


Asunto(s)
Angioedemas Hereditarios/sangre , Coagulación Sanguínea/fisiología , Bradiquinina/fisiología , Edad de Inicio , Angioedemas Hereditarios/enzimología , Angioedemas Hereditarios/etiología , Angioedemas Hereditarios/fisiopatología , Bradiquinina/biosíntesis , Permeabilidad Capilar , Activación de Complemento , Proteína Inhibidora del Complemento C1/fisiología , Factor XIIa/fisiología , Femenino , Angioedema Hereditario Tipos I y II/sangre , Angioedema Hereditario Tipos I y II/enzimología , Angioedema Hereditario Tipos I y II/fisiopatología , Humanos , Inflamación , Calidina/metabolismo , Calicreínas/fisiología , Quininógeno de Alto Peso Molecular/metabolismo , Masculino , Modelos Biológicos , Fenotipo , Polifosfatos/metabolismo , Inhibidores de Serina Proteinasa/deficiencia , Inhibidores de Serina Proteinasa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA