Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(29): e2401136121, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38985762

RESUMEN

Hemostasis relies on a reaction network of serine proteases and their cofactors to form a blood clot. Coagulation factor IXa (protease) plays an essential role in hemostasis as evident from the bleeding disease associated with its absence. RNA aptamers specifically targeting individual coagulation factors have potential as anticoagulants and as probes of the relationship between structure and function. Here, we report X-ray structures of human factor IXa without a ligand bound to the active site either in the apo-form or in complex with an inhibitory aptamer specific for factor IXa. The aptamer binds to an exosite in the catalytic domain and allosterically distorts the active site. Our studies reveal a conformational ensemble of IXa states, wherein large movements of Trp215 near the active site drive functional transitions between the closed (aptamer-bound), latent (apo), and open (substrate-bound) states. The latent state of the apo-enzyme may bear on the uniquely poor catalytic activity of IXa compared to other coagulation proteases. The exosite, to which the aptamer binds, has been implicated in binding VIIIa and heparin, both of which regulate IXa function. Our findings reveal the importance of exosite-driven allosteric modulation of IXa function and new strategies to rebalance hemostasis for therapeutic gain.


Asunto(s)
Aptámeros de Nucleótidos , Factor IXa , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/metabolismo , Factor IXa/metabolismo , Factor IXa/química , Factor IXa/antagonistas & inhibidores , Humanos , Regulación Alostérica , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Unión Proteica , Anticoagulantes/química , Anticoagulantes/metabolismo , Anticoagulantes/farmacología
2.
Biochem Biophys Res Commun ; 720: 150099, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38749192

RESUMEN

Binding of activated factor IX (fIXa) to the phosphatidylserine-expressing procoagulant platelets is a critical step in blood coagulation, which is necessary for the membrane-dependent intrinsic tenase complex assembly and factor X activation. However, the nature and parameters of the fIXa binding sites on the procoagulant platelet surface remain unclear. We used flow cytometry to elucidate the quantitative details of the fluorescently labeled fIXa binding to gel-filtered activated platelets. FIXa bound to the procoagulant platelet subpopulation only, with the parameters (maximal number of binding sites at 58900 ± 3400, Kd at 1000 ± 170 nM) similar to binding observed with phospholipid vesicles. No specific high-affinity binding sites for fIXa were detected, and binding proceeded similarly for different methods of procoagulant platelet production (thrombin, thrombin receptor activation peptide, collagen-related peptide, their combinations, or calcium ionophore A23187). Factor VIII, known to form a high affinity complex with fIXa, enhanced fIXa binding to platelets. In contrast, only competition effects were observed for factor X, which binds fIXa with much lower affinity. Unexpectedly, fIXa itself, fIX, and prothrombin also dose-dependently enhance fIXa binding at concentrations below 1000 nM, suggesting the formation of membrane-bound fIXa dimers and fIXa-prothrombin complexes on platelets. These findings provide a novel perspective on the fIXa binding site on procoagulant platelets, which does not have any major differences from pure phospholipid-based model membranes, exhibits inherently low affinity (3-5 orders of magnitude below the physiologically relevant fIXa concentration) but is significantly enhanced by its cofactor VIII, and regulated by previously unknown membrane interactions.


Asunto(s)
Plaquetas , Factor IXa , Unión Proteica , Humanos , Plaquetas/metabolismo , Factor IXa/metabolismo , Sitios de Unión , Coagulación Sanguínea , Trombina/metabolismo , Factor X/metabolismo , Citometría de Flujo , Fosfatidilserinas/metabolismo , Proteínas Portadoras , Péptidos
3.
J Theor Biol ; 582: 111757, 2024 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-38336240

RESUMEN

BACKGROUND: Factor X activation by the phospholipid-bound intrinsic tenase complex is a critical membrane-dependent reaction of blood coagulation. Its regulation mechanisms are unclear, and a number of questions regarding diffusional limitation, pathways of assembly and substrate delivery remain open. METHODS: We develop and analyze here a detailed mechanism-driven computer model of intrinsic tenase on phospholipid surfaces. Three-dimensional reaction-diffusion-advection and stochastic simulations were used where appropriate. RESULTS: Dynamics of the system was predominantly non-stationary under physiological conditions. In order to describe experimental data, we had to assume both membrane-dependent and solution-dependent delivery of the substrate. The former pathway dominated at low cofactor concentration, while the latter became important at low phospholipid concentration. Factor VIIIa-factor X complex formation was the major pathway of the complex assembly, and the model predicted high affinity for their lipid-dependent interaction. Although the model predicted formation of the diffusion-limited layer of substrate for some conditions, the effects of this limitation on the fXa production were small. Flow accelerated fXa production in a flow reactor model by bringing in fIXa and fVIIIa rather than fX. CONCLUSIONS: This analysis suggests a concept of intrinsic tenase that is non-stationary, employs several pathways of substrate delivery depending on the conditions, and is not particularly limited by diffusion of the substrate.


Asunto(s)
Factor X , Proteínas de Neoplasias , Fosfolípidos , Factor X/metabolismo , Fosfolípidos/metabolismo , Factor IXa/metabolismo , Cisteína Endopeptidasas/metabolismo , Cinética
4.
Bioorg Chem ; 148: 107463, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38776649

RESUMEN

Thrombosis leads to elevated mortality rates and substantial medical expenses worldwide. Human factor IXa (HFIXa) protease is pivotal in tissue factor (TF)-mediated thrombin generation, and represents a promising target for anticoagulant therapy. We herein isolated novel DNA aptamers that specifically bind to HFIXa through systematic evolution of ligands by exponential enrichment (SELEX) method. We identified two distinct aptamers, seq 5 and seq 11, which demonstrated high binding affinity to HFIXa (Kd = 74.07 ± 2.53 nM, and 4.93 ± 0.15 nM, respectively). Computer software was used for conformational simulation and kinetic analysis of DNA aptamers and HFIXa binding. These aptamers dose-dependently prolonged activated partial thromboplastin time (aPTT) in plasma. We further rationally optimized the aptamers by truncation and site-directed mutation, and generated the truncated forms (Seq 5-1t, Seq 11-1t) and truncated-mutated forms (Seq 5-2tm, Seq 11-2tm). They also showed good anticoagulant effects. The rationally and structurally designed antidotes (seq 5-2b and seq 11-2b) were competitively bound to the DNA aptamers and effectively reversed the anticoagulant effect. This strategy provides DNA aptamer drug-antidote pair with effective anticoagulation and rapid reversal, developing advanced therapies by safe, regulatable aptamer drug-antidote pair.


Asunto(s)
Antídotos , Aptámeros de Nucleótidos , Factor IXa , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Humanos , Factor IXa/antagonistas & inhibidores , Factor IXa/metabolismo , Antídotos/farmacología , Antídotos/química , Antídotos/síntesis química , Relación Dosis-Respuesta a Droga , Anticoagulantes/farmacología , Anticoagulantes/química , Relación Estructura-Actividad , Estructura Molecular , Técnica SELEX de Producción de Aptámeros
5.
Molecules ; 28(19)2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37836752

RESUMEN

Thromboembolic disorders, arising from abnormal coagulation, pose a significant risk to human life in the modern world. The FDA has recently approved several anticoagulant drugs targeting factor Xa (FXa) to manage these disorders. However, these drugs have potential side effects, leading to bleeding complications in patients. To mitigate these risks, coagulation factor IXa (FIXa) has emerged as a promising target due to its selective regulation of the intrinsic pathway. Due to the high structural and functional similarities of these coagulation factors and their inhibitor binding modes, designing a selective inhibitor specifically targeting FIXa remains a challenging task. The dynamic behavior of protein-ligand interactions and their impact on selectivity were analyzed using molecular dynamics simulation, considering the availability of potent and selective compounds for both coagulation factors and the co-crystal structures of protein-ligand complexes. Throughout the simulations, we examined ligand movements in the binding site, as well as the contact frequencies and interaction fingerprints, to gain insights into selectivity. Interaction fingerprint (IFP) analysis clearly highlights the crucial role of strong H-bond formation between the ligand and D189 and A190 in the S1 subsite for FIXa selectivity, consistent with our previous study. This dynamic analysis also reveals additional FIXa-specific interactions. Additionally, the absence of polar interactions contributes to the selectivity for FXa, as observed from the dynamic profile of interactions. A contact frequency analysis of the protein-ligand complexes provides further confirmation of the selectivity criteria for FIXa and FXa, as well as criteria for binding and activity. Moreover, a ligand movement analysis reveals key interaction dynamics that highlight the tighter binding of selective ligands to the proteins compared to non-selective and inactive ligands.


Asunto(s)
Factor IXa , Factor Xa , Humanos , Factor Xa/química , Factor IXa/metabolismo , Simulación de Dinámica Molecular , Ligandos , Factores de Coagulación Sanguínea
6.
J Membr Biol ; 255(6): 733-737, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36098799

RESUMEN

Blood coagulation is an intricate process, and it requires precise control of the activities of pro- and anticoagulant factors and sensitive signaling systems to monitor and respond to blood vessel insults. These requirements are fulfilled by phosphatidylserine, a relatively miniscule-sized lipid molecule amid the myriad of large coagulation proteins. This review limelight the role of platelet membrane phosphatidylserine (PS) in regulating a key enzymatic reaction of blood coagulation; conversion of factor X to factor Xa by the enzyme factor IXa and its cofactor factor VIIIa. PS is normally located on the inner leaflet of the resting platelet membrane but appears on the outer leaflet surface of the membrane surface after an injury happens. Human platelet activation leads to exposure of buried PS molecules on the surface of the platelet-derived membranes and the exposed PS binds to discrete and specific sites on factors IXa and VIIIa. PS binding to these sites allosterically regulates both factors IXa and VIIIa. The exposure of PS and its binding to factors IXa/VIIIa is a vital step during clotting. Insufficient exposure or a defective binding of PS to these clotting proteins is responsible for various hematologic diseases which are discussed in this review.


Asunto(s)
Factor IXa , Factor VIIIa , Humanos , Factor VIIIa/química , Factor VIIIa/metabolismo , Factor IXa/química , Factor IXa/metabolismo , Fosfatidilserinas/química , Factor X/metabolismo , Factor Xa/metabolismo , Cinética , Sitios de Unión
7.
Blood ; 136(23): 2703-2714, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-32678887

RESUMEN

The assembly of the enzyme-activated factor IX (FIXa) with its cofactor, activated factor VIII (FVIIIa) is a crucial event in the coagulation cascade. The absence or dysfunction of either enzyme or cofactor severely compromises hemostasis and causes hemophilia. FIXa is a notoriously inefficient enzyme that needs FVIIIa to drive its hemostatic potential, by a mechanism that has remained largely elusive to date. In this study, we employed hydrogen-deuterium exchange-mass spectrometry (HDX-MS) to investigate how FIXa responds to assembly with FVIIIa in the presence of phospholipids. This revealed a complex pattern of changes that partially overlaps with those changes that occur upon occupation of the substrate-binding site by an active site-directed inhibitor. Among the changes driven by both cofactor and substrate, HDX-MS highlighted several surface loops that have been implicated in allosteric networks in related coagulation enzymes. Inspection of FVIIIa-specific changes indicated that 3 helices are involved in FIXa-FVIIIa assembly. These are part of a basic interface that is also known as exosite II. Mutagenesis of basic residues herein, followed by functional studies, identified this interface as an extended FVIIIa-interactive patch. HDX-MS was also applied to recombinant FIXa variants that are associated with severe hemophilia B. This revealed that single amino acid substitutions can silence the extended network of FVIIIa-driven allosteric changes. We conclude that HDX-MS has the potential to visualize the functional impact of disease-associated mutations on enzyme-cofactor complexes in the hemostatic system.


Asunto(s)
Medición de Intercambio de Deuterio , Factor IXa/química , Factor VIII/química , Espectrometría de Masas , Mutación , Regulación Alostérica/genética , Factor IXa/genética , Factor IXa/metabolismo , Factor VIII/genética , Factor VIII/metabolismo , Hemofilia B/genética , Hemofilia B/metabolismo , Humanos , Conformación Proteica en Hélice alfa , Dominios Proteicos
8.
J Biol Chem ; 295(45): 15198-15207, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32859749

RESUMEN

Factor X activation by the intrinsic Xase complex, composed of factor IXa bound to factor VIIIa on membranes, is essential for the amplified blood coagulation response. The biological significance of this step is evident from bleeding arising from deficiencies in factors VIIIa or IXa in hemophilia. Here, we assess the mechanism(s) that enforce the distinctive specificity of intrinsic Xase for its biological substrate. Active-site function of IXa was assessed with a tripeptidyl substrate (PF-3688). The reversible S1 site binder, 4-aminobenzamidine (pAB), acted as a classical competitive inhibitor of PF-3688 cleavage by Xase. In contrast, pAB acted as a noncompetitive inhibitor of factor X activation. This disconnect between peptidyl substrate and protein substrate cleavage indicates a major role for interactions between factor X and extended sites on Xase in determining substrate affinity. Accordingly, an uncleavable factor X variant, not predicted to engage the active site of IXa within Xase, acted as a classical competitive inhibitor of factor X activation. Fluorescence studies confirmed the binding of factor X to Xase assembled with IXa with a covalently blocked active site. Our findings suggest that the recognition of factor X by the intrinsic Xase complex occurs through a multistep "dock-and-lock" pathway in which the initial interaction between factor X and intrinsic Xase occurs at exosites distant from the active site, followed by active-site docking and bond cleavage.


Asunto(s)
Factor IXa/metabolismo , Factor VIIIa/metabolismo , Factor X/metabolismo , Sitios de Unión , Humanos , Cinética , Proteínas Recombinantes/metabolismo
9.
Molecules ; 26(17)2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34500804

RESUMEN

Blood coagulation is an essential physiological process for hemostasis; however, abnormal coagulation can lead to various potentially fatal disorders, generally known as thromboembolic disorders, which are a major cause of mortality in the modern world. Recently, the FDA has approved several anticoagulant drugs for Factor Xa (FXa) which work via the common pathway of the coagulation cascade. A main side effect of these drugs is the potential risk for bleeding in patients. Coagulation Factor IXa (FIXa) has recently emerged as the strategic target to ease these risks as it selectively regulates the intrinsic pathway. These aforementioned coagulation factors are highly similar in structure, functional architecture, and inhibitor binding mode. Therefore, it remains a challenge to design a selective inhibitor which may affect only FIXa. With the availability of a number of X-ray co-crystal structures of these two coagulation factors as protein-ligand complexes, structural alignment, molecular docking, and pharmacophore modeling were employed to derive the relevant criteria for selective inhibition of FIXa over FXa. In this study, six ligands (three potent, two selective, and one inactive) were selected for FIXa inhibition and six potent ligands (four FDA approved drugs) were considered for FXa. The pharmacophore hypotheses provide the distribution patterns for the principal interactions that take place in the binding site. None of the pharmacophoric patterns of the FXa inhibitors matched with any of the patterns of FIXa inhibitors. Based on pharmacophore analysis, a selectivity of a ligand for FIXa over FXa may be defined quantitatively as a docking score of lower than -8.0 kcal/mol in the FIXa-grids and higher than -7.5 kcal/mol in the FXa-grids.


Asunto(s)
Anticoagulantes/farmacología , Factor IXa/antagonistas & inhibidores , Inhibidores del Factor Xa/farmacología , Factor Xa/metabolismo , Anticoagulantes/química , Cristalografía por Rayos X , Factor IXa/genética , Factor IXa/metabolismo , Factor Xa/genética , Inhibidores del Factor Xa/química , Humanos , Modelos Moleculares , Estructura Molecular
10.
Bioorg Med Chem Lett ; 30(15): 127279, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32527459

RESUMEN

The synthesis and structure activity relationship development of a pyrimidine series of heterocyclic Factor IXa inhibitors is described. Increased selectivity over Factor Xa inhibition was achieved through SAR expansion of the P1 element. Select compounds were evaluated in vivo to assess their plasma levels in rat.


Asunto(s)
Descubrimiento de Drogas , Factor IXa/antagonistas & inhibidores , Inhibidores del Factor Xa/farmacología , Pirimidinas/farmacología , Relación Dosis-Respuesta a Droga , Factor IXa/metabolismo , Inhibidores del Factor Xa/síntesis química , Inhibidores del Factor Xa/química , Humanos , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad
11.
Arterioscler Thromb Vasc Biol ; 38(1): 266-274, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29097362

RESUMEN

OBJECTIVE: Combined oral contraceptives induce a reversible hypercoagulable state with an enhanced risk of venous thromboembolism, but the underlying mechanism(s) remain unclear. Subjects on combined oral contraceptives also demonstrate a characteristic resistance to APC (activated protein C) in the thrombin generation assay. Here, we report the potential role of plasma factor IXa (FIXa) as a mechanism for hormone-induced systemic hypercoagulability. APPROACH AND RESULTS: A novel assay was used to determine FIXa activity in plasma samples from volunteer blood donors. Plasma from 36 premenopausal females on hormonal contraception and 35 not on hormonal contraception, 35 postmenopausal females, and 10 males were analyzed for FIXa activity, total PS (protein S), total tissue factor pathway inhibitor (TFPI), and TFPI-α antigen. Premenopausal females on hormonal contraception demonstrated significantly increased FIXa activity and decreased TFPI-α compared with the other groups. Remarkably, FIXa values were not normally distributed in the hormonal contraception group, but skewed toward the high end. Plasma FIXa activity inversely correlated with both TFPI-α and total PS antigen. Ex vivo determination of TF-dependent FIX activation in FV-deficient plasma demonstrated that inhibitory anti-TFPI antibodies enhanced FIXa generation by 2- to 3-fold, whereas addition of 75 nmol/L PS reduced FIXa generation by ≈2-fold. Further, increasing FIXa concentration enhanced APC resistance during TF-triggered plasma thrombin generation. CONCLUSIONS: Elevation of plasma FIXa activity in association with reductions in TFPI-α and PS is a potential mechanism for systemic hypercoagulability and resistance to APC in premenopausal females on hormonal contraception.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Anticonceptivos Orales Combinados/administración & dosificación , Factor IXa/metabolismo , Premenopausia/sangre , Resistencia a la Proteína C Activada/sangre , Resistencia a la Proteína C Activada/inducido químicamente , Adulto , Anciano , Biomarcadores/sangre , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Lipoproteínas/sangre , Masculino , Persona de Mediana Edad , Proteína S/metabolismo , Factores de Riesgo , Factores Sexuales , Trombofilia/sangre , Trombofilia/inducido químicamente , Regulación hacia Arriba , Adulto Joven
12.
Arterioscler Thromb Vasc Biol ; 38(4): 816-828, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29419409

RESUMEN

OBJECTIVE: PS (protein S) is a plasma protein that directly inhibits the coagulation FIXa (factor IXa) in vitro. Because elevated FIXa is associated with increased risk of venous thromboembolism, it is important to establish how PS inhibits FIXa function in vivo. The goal of this study is to confirm direct binding of PS with FIXa in vivo, identify FIXa amino acid residues required for binding PS in vivo, and use an enzymatically active FIXa mutant that is unable to bind PS to measure the significance of PS-FIXa interaction in hemostasis. APPROACH AND RESULTS: We demonstrate that PS inhibits FIXa in vivo by associating with the FIXa heparin-binding exosite. We used fluorescence tagging, immunohistochemistry, and protein-protein crosslinking to show in vivo interaction between FIXa and PS. Importantly, platelet colocalization required a direct interaction between the 2 proteins. FIXa and PS also coimmunoprecipitated from plasma, substantiating their interaction in a physiological milieu. PS binding to FIXa and PS inhibition of the intrinsic Xase complex required residues K132, K126, and R170 in the FIXa heparin-binding exosite. A double mutant, K132A/R170A, retained full activity but could not bind to PS. Crucially, Hemophilia B mice infused with FIXa K132A/R170A displayed an accelerated rate of fibrin clot formation compared with wild-type FIXa. CONCLUSIONS: Our findings establish PS as an important in vivo inhibitor of FIXa. Disruption of the interaction between PS and FIXa causes an increased rate of thrombus formation in mice. This newly discovered function of PS implies an unexploited target for antithrombotic therapeutics.


Asunto(s)
Plaquetas/metabolismo , Factor IXa/metabolismo , Hemofilia B/sangre , Hemostasis , Heparina/metabolismo , Proteína S/metabolismo , Trombosis de la Vena/prevención & control , Animales , Sitios de Unión , Unión Competitiva , Coagulantes/administración & dosificación , Modelos Animales de Enfermedad , Factor IX/genética , Factor IX/metabolismo , Factor IXa/administración & dosificación , Factor IXa/genética , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemostasis/efectos de los fármacos , Humanos , Infusiones Intravenosas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Trombosis de la Vena/sangre , Trombosis de la Vena/genética
13.
Niger J Clin Pract ; 22(3): 416-421, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30837433

RESUMEN

INTRODUCTION: Inhibitor formation is a major complication of hemophilia treatment because it interferes with the clinical response to factor replacement and causes significant morbidity. This cross-sectional study was conducted to assess the presence and frequency of inhibitors among registered person with hemophilia and to identify risk factors associated with inhibitor development. PATIENTS AND METHODS: A total of 143 hemophilics, 118 with hemophilia A (HA) and 25 with hemophilia B (HB), were enrolled for the study. Participant's clinical data were obtained through patient's medical records. Factor VIII and IX levels and the presence of inhibitors were assessed using a fully automated coagulometer. From the results of a Bethesda assay, patients were divided into those with high titers (≥5 BU) and those with low titers (<5 BU). RESULTS: The patient's age ranged from 1 to 67 years with median of 13.8 years. Inhibitors were detected in 18.6% and none of HA and HB patients, respectively. Of the 22 patients with HA and inhibitors, 18 (82%) had high titer inhibitors. The frequency of inhibitors was significantly higher among patients with severe hemophilia, a history of early exposure (≤3 months) to factor VIII concentrate, and family histories of autoimmune disease and immune system challenges (P < 0.05). The independent risk factors associated with inhibitor development were severe hemophilia (95% CIs = 1.02-55.6, OR = 7.5) and immune system challenges (95% CIs = 1.14-5.99, OR = 2.6). CONCLUSION: Inhibitors were common among HA patients, and both severe HA and immune system challenges (surgery and trauma) are independent risk factors for inhibitor development.


Asunto(s)
Factor IXa/antagonistas & inhibidores , Factor VIII/antagonistas & inhibidores , Hemofilia A/sangre , Hemofilia B/sangre , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios Transversales , Factor IXa/metabolismo , Factor IXa/uso terapéutico , Factor VIII/metabolismo , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Hemostáticos/efectos adversos , Hemostáticos/uso terapéutico , Humanos , Lactante , Irak , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
14.
Curr Opin Hematol ; 25(5): 382-388, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30015643

RESUMEN

PURPOSE OF REVIEW: Since the selection of the first thrombin-binding aptamer in 1992, the use of nucleic acid aptamers to target specific coagulation factors has emerged as a valuable approach for generating novel anticoagulant and procoagulant therapeutics. Herein, we highlight the most recent discoveries involving application of aptamers for those purposes. RECENT FINDINGS: Learning from the successes and pitfalls of the FIXa-targeting aptamer pegnivacogin in preclinical and clinical studies, the latest efforts to develop antidote-controllable anticoagulation strategies for cardiopulmonary bypass that avoid unfractionated heparin involve potentiation of the exosite-binding factor X (FX)a aptamer 11F7t by combination with either a small molecule FXa catalytic site inhibitor or a thrombin aptamer. Recent work has also focused on identifying aptamer inhibitors of contact pathway factors such as FXIa and kallikrein, which may prove to be well tolerated and effective antithrombotic agents in certain clinical settings. Finally, new approaches to develop procoagulant aptamers to control bleeding associated with hemophilia and other coagulopathies involve targeting activated protein C and tissue plasminogen activator. SUMMARY: Overall, these recent findings exemplify the versatility of aptamers to modulate a variety of procoagulant and anticoagulant factors, along with their capacity to be used complementarily with other aptamers or drugs for wide-ranging applications.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Factor IXa , Inhibidores del Factor Xa/uso terapéutico , Hemostasis , Animales , Antídotos/farmacocinética , Antídotos/uso terapéutico , Aptámeros de Nucleótidos/efectos adversos , Aptámeros de Nucleótidos/farmacocinética , Puente Cardiopulmonar , Dominio Catalítico , Factor IXa/antagonistas & inhibidores , Factor IXa/metabolismo , Inhibidores del Factor Xa/efectos adversos , Inhibidores del Factor Xa/farmacocinética , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Calicreínas/metabolismo
16.
Biochem J ; 473(15): 2395-411, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27208168

RESUMEN

Coagulation Factor IX is positioned at the merging point of the intrinsic and extrinsic blood coagulation cascades. Factor IXa (activated Factor IX) serves as the trigger for amplification of coagulation through formation of the so-called Xase complex, which is a ternary complex of Factor IXa, its substrate Factor X and the cofactor Factor VIIIa on the surface of activated platelets. Within the Xase complex the substrate turnover by Factor IXa is enhanced 200000-fold; however, the mechanistic and structural basis for this dramatic enhancement remains only partly understood. A multifaceted approach using enzymatic, biophysical and crystallographic methods to evaluate a key set of activity-enhanced Factor IXa variants has demonstrated a delicately balanced bidirectional network. Essential molecular interactions across multiple regions of the Factor IXa molecule co-operate in the maturation of the active site. This maturation is specifically facilitated by long-range communication through the Ile(212)-Ile(213) motif unique to Factor IXa and a flexibility of the 170-loop that is further dependent on the conformation in the Cys(168)-Cys(182) disulfide bond. Ultimately, the network consists of compensatory brakes (Val(16) and Ile(213)) and accelerators (Tyr(99) and Phe(174)) that together allow for a subtle fine-tuning of enzymatic activity.


Asunto(s)
Factor IXa/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Biocatálisis , Activación Enzimática , Factor IXa/química , Humanos , Modelos Moleculares , Plásmidos , Conformación Proteica , Pliegue de Proteína , Proteolisis , Homología de Secuencia de Aminoácido , Espectrometría de Fluorescencia
17.
Biochim Biophys Acta ; 1854(10 Pt A): 1351-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26012870

RESUMEN

Alterations in coagulation factor X (FX) activation, mediated by the extrinsic VIIa/tissue factor (FVIIa/TF) or the intrinsic factor IXa/factor VIIIa (FIXa/FVIIIa) complexes, can result in hemorrhagic/prothrombotic tendencies. However, the molecular determinants involved in substrate recognition by these enzymes are poorly defined. Here, we investigated the role of arginine 386 (chymotrypsin numbering c202), a surface-exposed residue on the FX catalytic domain. The naturally occurring FX386Cys mutant and FX386Ala variant were characterized. Despite the unpaired cysteine, recombinant (r)FX386Cys was efficiently secreted (88.6±21.3% of rFXwt) and possessed normal clearance in mice. rFX386Cys was also normally activated by FVIIa/TF and displayed intact amidolytic activity. In contrast, rFX386Cys activation by the FIXa/FVIIIa complex was 4.5-fold reduced, which was driven by a decrease in the kcat (1.6∗10(-4) s(-1) vs 5.8∗10(-4) s(-1), rFXwt). The virtually unaltered Km (70.6 nM vs 55.6nM, rFXwt) suggested no major alterations in the FX substrate exosite. Functional assays in plasma supplemented with rFX386Cys indicated a remarkable reduction in the thrombin generation rate and thus in coagulation efficiency. Consistently, the rFX386Ala variant displayed similar biochemical features suggesting that global changes at position 386 impact the intrinsic pathway activation. These data indicate that the FXArg386 is involved in FIXa/FVIIIa-mediated FX activation and help in elucidating the bleeding tendency associated with the FX386Cys in a rare FX deficiency case. Taking advantage of the unpaired cysteine, the rFX386Cys mutant may be efficiently targeted by thiol-specific ligands and represent a valuable tool to study FX structure-function relationships both in vitro and in vivo.


Asunto(s)
Coagulación Sanguínea/genética , Factor X/metabolismo , Factor Xa/metabolismo , Mutación , Animales , Pruebas de Coagulación Sanguínea , Dominio Catalítico , Factor IXa/genética , Factor IXa/metabolismo , Factor VIIIa/genética , Factor VIIIa/metabolismo , Factor X/química , Factor X/genética , Factor Xa/química , Factor Xa/genética , Células HEK293 , Humanos , Cinética , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombina/genética , Trombina/metabolismo
18.
Haemophilia ; 22(5): 780-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27038076

RESUMEN

INTRODUCTION: The thrombin generation test (TGT) is used both as a global haemostasis assay, and to compare activities of coagulation factor concentrates that have been spiked into patient plasma. However, TGT has not been systematically optimized to evaluate factor VIII (FVIII) product potency. AIMS: To improve the sensitivity of TGT to FVIII and allow a comparative analysis of the thrombin generating capacities of FVIII concentrates against reference preparations with known FVIII activity. METHODS: Concentrations of TGT components (analytical variables) were assessed to maximize the linearity and range of responses to the concentration of FVIII. RESULTS: We optimized the range and sensitivity of the TGT assay with respect to FVIII through the addition of FXIa to the assay. Other parameters that were adjusted, i.e. tissue factor (TF), procoagulant lipids and plasma concentrations, did not improve the ability of the assay to measure both high and very low levels of FVIII. In the optimized TF/FXIa-activated TGT assay, all thrombin generation curve parameters were suitable for FVIII quantification, but thrombin peak height and maximal velocity demonstrated better linearity in the desired FVIII range. We found that the optimized TF/FXIa-activated TGT has a wider range of sensitivity to FVIII than a commercially available TGT. Additionally, we demonstrated that the TF/FXIa-activated assay performs adequately by comparing potency measurements of five commercially available FVIII products using TGT and traditional chromogenic and one-stage clotting assays. CONCLUSIONS: The optimized TGT assay can be used to quantify and compare the thrombin generating capacities of FVIII concentrates.


Asunto(s)
Pruebas de Coagulación Sanguínea , Factor VIII/análisis , Trombina/metabolismo , Automatización , Compuestos Cromogénicos/química , Factor IXa/química , Factor IXa/metabolismo , Factor XIa/química , Factor XIa/metabolismo , Heparina/química , Humanos , Juego de Reactivos para Diagnóstico , Especificidad por Sustrato , Trombomodulina/química , Tromboplastina/química
19.
Vox Sang ; 111(1): 33-42, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26927329

RESUMEN

BACKGROUND AND OBJECTIVES: In this study, differences in levels of proteins involved in coagulation and fibrinolysis were compared between fresh frozen (quarantine plasma) and Omniplasma. Furthermore, thawing conditions and plasma stability after thawing were studied. MATERIALS AND METHODS: 10 Omniplasma and 10 quarantine plasma units were used to study different procoagulation, anticoagulation and fibrinolytic parameters. Analysis took place at different time-points during plasma storage at 2-6°C. RESULTS: At baseline, significant reduced levels of factor V, free protein S, α2-antiplasmin and tPA-induced ROTEM lysis time were observed in Omniplasma as compared to quarantine plasma. Moreover, thrombin generation, IXa-AT complex levels and factor XIa were significantly increased in Omniplasma. The majority of the parameters studied remained stable in Omniplasma 48 h after thawing, with the exception of factor VIII (decrease) and IXa-AT (increase). CONCLUSION: Our results suggest an increased coagulation potential, presumingly as a result of contact activation during the production process and also, an increased fibrinolytic potential in Omniplasma. The stability of Omniplasma, based upon the different parameters studied, is comparable to Q-plasma. A maximum post-thawing time of 48 hfor Omniplasma can be suggested.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Detergentes/farmacología , Plasma/química , Solventes/química , Detergentes/química , Factor IXa/metabolismo , Factor XIa/metabolismo , Humanos , Activador de Tejido Plasminógeno/metabolismo , alfa 2-Antiplasmina/metabolismo
20.
J Biol Chem ; 289(20): 14020-9, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24692542

RESUMEN

The factor VIII (FVIII) crystal structure suggests a possible bonding interaction of His(281) (A1 domain) with Ser(524) (A2 domain), although the resolution of the structure (∼4 Å) does not firmly establish this bonding. To establish that side chains of these residues participate in an interdomain bond, we prepared and examined the functional properties of a residue swap variant (H281S/S524H) where His(281) and Ser(524) residues were exchanged with one another and a disulfide-bridged variant (H281C/S524C) where the two residues were replaced with Cys. The latter variant showed efficient disulfide bonding of the A1 and A2 domains. The swap variant showed WT-like FVIII and FVIIIa stability, which were markedly reduced for H281A and S524A variants in an earlier study. The disulfide-bridged variant showed ∼20% increased FVIII stability, and FVIIIa did not decay during the time course measured. This variant also yielded 35% increased thrombin peak values compared with WT in a plasma-based thrombin generation assay. Binding analyses of H281S-A1/A3C1C2 dimer with S524H-A2 subunit yielded a near WT-like affinity value, whereas combining the variant dimer or A2 subunit with the WT complement yielded ∼5- and ∼10-fold reductions, respectively, in affinity. Other functional properties including thrombin generation potential, FIXa binding affinity, Km for FX of FXase complexes, thrombin activation efficiency, and down-regulation by activated protein C showed similar results for the two variants compared with WT FVIII. These results indicate that the side chains of His(281) and Ser(524) are in close proximity and contribute to a bonding interaction in FVIII that is retained in FVIIIa.


Asunto(s)
Coagulación Sanguínea , Factor VIII/química , Factor VIII/metabolismo , Factor VIIIa/metabolismo , Histidina , Serina , Factor IXa/metabolismo , Factor VIII/genética , Humanos , Modelos Moleculares , Mutagénesis , Mutación , Estabilidad Proteica , Estructura Terciaria de Proteína , Trombina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA