Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Thromb Res ; 233: 119-126, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38039724

RESUMEN

BACKGROUND: Microvesicles (MVs) produced by platelets upon activation possess high procoagulant activity and represent a possible thrombotic risk marker. However, direct experimental evaluation of the adhesive properties of MVs and their potential role in thrombus growth is lacking. OBJECTIVES: We investigated integrin αIIbß3 status and adhesive properties of plasma-circulating and platelet-derived MVs from healthy individuals. METHODS: MVs were isolated from whole blood or produced from activated platelets. Flow cytometry was used for quantification of fluorescently labeled PAC-1 and fibrinogen binding to MVs. Confocal microscopy was used for evaluation of MVs adhesion to fibrinogen and for estimation of their involvement in whole blood thrombus formation in a parallel-plate flow chambers under arterial shear conditions. RESULTS AND CONCLUSIONS: Neither circulating plasma MVs, nor platelet-activation-produced MVs bound PAC-1. However, both types of MVs specifically and weakly bound fibrinogen (about 400 molecules of bound fibrinogen per MV versus >100,000 per non-procoagulant activated platelet). Still, the MVs did not adhere stably to the immobilized fibrinogen. Both types of MVs were weakly incorporated into a thrombus and did not affect thrombus formation: average thrombus height in the recalcified whole blood in the presence of platelet-activation-produced MVs was 4.19 ± 1.38 µm versus 4.87 ± 1.72 µm (n = 6, p > 0.05) in the control experiments. This suggests that MVs present in plasma of healthy individuals are not likely to be directly involved in thrombus formation under arterial flow conditions.


Asunto(s)
Plaquetas , Trombosis , Humanos , Plaquetas/metabolismo , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Fibrinógeno/metabolismo
2.
Dokl Biochem Biophys ; 499(1): 242-246, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34426920

RESUMEN

Using the methods of dynamic and elastic light scattering and confocal laser scanning microscopy, the damage in the spatial fibrin structure during peroxide- and hypochlorite-induced oxidation of fibrinogen was studied. Peroxide had a weak effect on the structural organization of fibrin, whereas hypochlorite caused the formation of abnormal fibrin with reduced individual fiber diameter and decreased porosity. Measurements of the size distributions of the native and oxidized fibrinogen revealed a decrease in the hydrodynamic size of the oxidized fibrinogen molecule with an increase in the concentration of oxidizers. These results indicate that the hydrophobicity of fibrinogen surface increased and its colloidal stability decreased. The possible role of oxidative sites in the assembly of structurally abnormal fibrin is analyzed.


Asunto(s)
Fibrina/química , Fibrinógeno/metabolismo , Ácido Hipocloroso/farmacología , Peróxidos/farmacología , Fibrina/metabolismo , Oxidación-Reducción/efectos de los fármacos
3.
Hamostaseologie ; 41(2): 146-153, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33860522

RESUMEN

During the past decades, it has been increasingly recognized that the major function of accelerating membrane-dependent reactions of blood coagulation is predominantly implemented by a subset of activated platelets. These procoagulant platelets (also called collagen- and thrombin-activated or COAT, coated, necrotic, although there could be subtle differences between these definitions) are uniquely characterized by both procoagulant activity and, at the same time, inactivated integrins and profibrinolytic properties. The mechanisms of their generation both in vitro and in situ have been increasingly becoming clear, suggesting unique and multidirectional roles in hemostasis and thrombosis. In this mini-review, we shall highlight the existing concepts and challenges in this field.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Humanos
4.
Bull Math Biol ; 83(5): 50, 2021 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-33772645

RESUMEN

Blood coagulation represents one of the most studied processes in biomedical modelling. However, clinical applications of this modelling remain limited because of the complexity of this process and because of large inter-patient variation of the concentrations of blood factors, kinetic constants and physiological conditions. Determination of some of these patients-specific parameters is experimentally possible, but it would be related to excessive time and material costs impossible in clinical practice. We propose in this work a methodological approach to patient-specific modelling of blood coagulation. It begins with conventional thrombin generation tests allowing the determination of parameters of a reduced kinetic model. Next, this model is used to study spatial distributions of blood factors and blood coagulation in flow, and to evaluate the results of medical treatment of blood coagulation disorders.


Asunto(s)
Coagulación Sanguínea , Modelos Biológicos , Modelación Específica para el Paciente , Trastornos de la Coagulación Sanguínea/diagnóstico , Trastornos de la Coagulación Sanguínea/patología , Humanos
5.
Biochemistry (Mosc) ; 85(10): 1267-1276, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33202211

RESUMEN

Programmed cell death of non-nucleated blood cells - platelets - could be associated with pathophysiology of oncologic and oncohematologic diseases. It contributes to both bleedings (caused by the thrombocytopenia, which is induced by elimination of the platelets) and thrombosis (caused by the processes of blood coagulation on the surface of phosphatidylserine exposing platelets). Here we characterized functional responses of platelets from the patients with various oncological disorders undergoing chemotherapy and compared them to the platelets from the healthy donors and platelets pre-incubated with apoptosis inducer ABT-737. Some patients exhibited diminished capability of platelets to aggregate. Immunophenotyping of these platelets revealed their pre-activation in comparison to the platelets from the healthy donors. Calcium signaling analysis revealed that in the patient-derived platelets, as well as in the apoptotic platelets, intracellular calcium levels were increased in resting cells. However, moderate level of this increase together with weak expression of phosphatidylserine allows us to assume that apoptotic processes in the circulating platelets from the patients are limited.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Neoplasias Hematológicas , Adolescente , Compuestos de Bifenilo/farmacología , Coagulación Sanguínea/efectos de los fármacos , Calcio/metabolismo , Niño , Preescolar , Femenino , Neoplasias Hematológicas/sangre , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Masculino , Nitrofenoles/farmacología , Fosfatidilserinas/sangre , Piperazinas/farmacología , Sulfonamidas/farmacología
6.
Biomed Khim ; 64(5): 387-396, 2018 Sep.
Artículo en Ruso | MEDLINE | ID: mdl-30378555

RESUMEN

Platelet activating receptor CLEC-2 has been identified on platelet surface a decade ago. The only confirmed endogenous CLEC-2 agonist is podoplanin. Podoplanin is a transmembrane protein expressed by lymphatic endothelial cells, reticular fibroblastic cells in lymph nodes, kidney podocytes and by cells of certain tumors. CLEC-2 and podoplanin are involved in the processes of embryonic development (blood-lymph vessel separation and angiogenesis), maintaining of vascular integrity of small vessels during inflammation and prevention of blood-lymphatic mixing in high endothelial venules. However, CLEC-2 and podoplanin are contributing to tumor methastasis progression, Salmonella sepsis, deep-vein thrombosis. CLEC-2 signalling cascade includes tyrosine-kinases (Syk, SFK, Btk) as well as adapter LAT and phospholipase Cg2, which induces calcium signalling. CLEC-2, podoplanin and proteins, participating in CLEC-2 signalling cascade, are perspective targets for antithrombotic therapy.


Asunto(s)
Plaquetas , Activación Plaquetaria , Lectinas Tipo C , Glicoproteínas de Membrana , Transducción de Señal
8.
Phys Life Rev ; 26-27: 57-95, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29550179

RESUMEN

Hemostasis is a complex physiological mechanism that functions to maintain vascular integrity under any conditions. Its primary components are blood platelets and a coagulation network that interact to form the hemostatic plug, a combination of cell aggregate and gelatinous fibrin clot that stops bleeding upon vascular injury. Disorders of hemostasis result in bleeding or thrombosis, and are the major immediate cause of mortality and morbidity in the world. Regulation of hemostasis and thrombosis is immensely complex, as it depends on blood cell adhesion and mechanics, hydrodynamics and mass transport of various species, huge signal transduction networks in platelets, as well as spatiotemporal regulation of the blood coagulation network. Mathematical and computational modeling has been increasingly used to gain insight into this complexity over the last 30 years, but the limitations of the existing models remain profound. Here we review state-of-the-art-methods for computational modeling of thrombosis with the specific focus on the analysis of unresolved challenges. They include: a) fundamental issues related to physics of platelet aggregates and fibrin gels; b) computational challenges and limitations for solution of the models that combine cell adhesion, hydrodynamics and chemistry; c) biological mysteries and unknown parameters of processes; d) biophysical complexities of the spatiotemporal networks' regulation. Both relatively classical approaches and innovative computational techniques for their solution are considered; the subjects discussed with relation to thrombosis modeling include coarse-graining, continuum versus particle-based modeling, multiscale models, hybrid models, parameter estimation and others. Fundamental understanding gained from theoretical models are highlighted and a description of future prospects in the field and the nearest possible aims are given.


Asunto(s)
Simulación por Computador , Modelos Biológicos , Trombosis , Coagulación Sanguínea , Hemostasis , Humanos , Cinética , Adhesividad Plaquetaria , Agregación Plaquetaria , Trombosis/sangre
9.
J Thromb Haemost ; 14(10): 2045-2057, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27513817

RESUMEN

Essentials Roles of the two thrombin receptors in platelet signaling are poorly understood. Computational systems biology modeling was used together with continuous flow cytometry. Dual-receptor system has wide-range sensitivity to thrombin and optimal response dynamics. Procoagulant platelet formation is determined by donor-specific activities of the two receptors. SUMMARY: Background Activation of human platelets with thrombin proceeds via two protease-activated receptors (PARs), PAR1 and PAR4, that have identical main intracellular signaling responses. Although there is evidence that they have different cleavage/inactivation kinetics (and some secondary variations in signaling), the reason for such redundancy is not clear. Methods We developed a multicompartmental stochastic computational systems biology model of dual-receptor thrombin signaling in platelets to gain insight into the mechanisms and roles of PAR1 and PAR4 functioning. Experiments employing continuous flow cytometry of washed human platelets were used to validate the model and test its predictions. Activity of PAR receptors in donors was evaluated by mRNA measurement and by polymorphism sequencing. Results Although PAR1 activation produced rapid and short-lived response, signaling via PAR4 developed slowly and propagated in time. Response of the dual-receptor system was both rapid and prolonged in time. Inclusion of PAR1/PAR4 heterodimer formation promoted PAR4 signaling in the medium range of thrombin concentration (about 10 nm), with little contribution at high and low thrombin. Different dynamics and dose-dependence of procoagulant platelet formation in healthy donors was associated with individual variations in PAR1 and PAR4 activities and particularly by the Ala120Thr polymorphism in the F2RL3 gene encoding PAR4. Conclusions The dual-receptor combination is critical to produce a response combining three critical advantages: sensitivity to thrombin concentration, rapid onset and steady propagation; specific features of the protease-activated receptors do not allow combination of all three in a single receptor.


Asunto(s)
Plaquetas/metabolismo , Activación Plaquetaria , Biología de Sistemas , Trombina/metabolismo , Adolescente , Adulto , Animales , Plaquetas/citología , Simulación por Computador , Dimerización , Femenino , Humanos , Cinética , Masculino , Agregación Plaquetaria , Polimorfismo Genético , Receptor PAR-1/sangre , Receptores de Trombina/sangre , Receptores de Trombina/genética , Transducción de Señal , Adulto Joven
10.
J Thromb Haemost ; 14(9): 1867-81, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27343487

RESUMEN

UNLABELLED: Essentials The sequence and logic of events leading to platelet procoagulant activity are poorly understood. Confocal time-lapse microscopy was used to investigate activation of single adherent platelets. Platelet transition to the procoagulant state followed cytosolic calcium oscillations. Mitochondria did not collapse simultaneously and membrane potential loss could be reversible. SUMMARY: Background Activated platelets form two subpopulations, one of which is able to efficiently aggregate, and another that externalizes phosphatidylserine (PS) and thus accelerates membrane-dependent reactions of blood coagulation. The latter, procoagulant subpopulation is characterized by a high cytosolic calcium level and the loss of inner mitochondrial membrane potential, and there are conflicting opinions on their roles in its formation. Methods We used confocal microscopy to investigate the dynamics of subpopulation formation by imaging single, fibrinogen-bound platelets with individual mitochondria in them upon loading with calcium-sensitive and mitochondrial potential-sensitive dyes. Stimulation was performed with thrombin or the protease-activated receptor (PAR) 1 agonist SFLLRN. Stochastic simulations with a computational systems biology model of PAR1 calcium signaling were employed for analysis. Results Platelet activation resulted in a series of cytosolic calcium spikes and mitochondrial calcium uptake in all platelets. The frequency of spikes decreased with time for SFLLRN stimulation, but remained high for a long period of time for thrombin. In some platelets, uptake of calcium by mitochondria led to the mitochondrial permeability transition pore opening and inner mitochondrial membrane potential loss, which could be either reversible or irreversible. The latter resulted in an increase in the cytosolic calcium level and PS exposure. These platelets had higher cytosolic calcium levels before activation, and their mitochondria collapsed not simultaneously but one after another. Conclusions These results support a model of procoagulant subpopulation development following a series of stochastic cytosolic calcium spikes that are accumulated by mitochondria, leading to a collapse, and suggest important roles of individual platelet reactivity and signal exchange between different mitochondria of a platelet.


Asunto(s)
Señalización del Calcio , Calcio/química , Mitocondrias/metabolismo , Fosfatidilserinas/química , Coagulación Sanguínea , Plaquetas/metabolismo , Simulación por Computador , Citosol/metabolismo , Humanos , Potencial de la Membrana Mitocondrial , Microscopía Confocal , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Fragmentos de Péptidos/química , Activación Plaquetaria , Adhesividad Plaquetaria , Receptor PAR-1/antagonistas & inhibidores , Transducción de Señal , Procesos Estocásticos , Trombina/metabolismo
11.
Vox Sang ; 110(3): 244-52, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26646605

RESUMEN

BACKGROUND AND OBJECTIVES: Pathogen reduction technologies may affect platelet quality during storage. We studied functional characteristics and clinical effectiveness of platelet concentrates (PCs) treated with Mirasol in plasma and in platelet-additive solution SSP+. MATERIALS AND METHODS: Mirasol-treated, gamma-irradiated and untreated apheresis PCs were examined on days 0, 1, 3 and 5 of storage. Phosphatidylserine, P-selectin and active glycoprotein IIb/IIIa were analysed using flow cytometry before and after platelet stimulation. Platelet count increments, the numbers of inefficient transfusions and post-transfusion reactions were analysed to estimate clinical effectiveness. RESULTS: A significant increase in all platelet activation markers occurred during storage in all PC groups. Activation markers in Mirasol-treated samples were already significantly higher compared with the control ones on the day of harvesting, and continued to grow during the storage. Mirasol treatment increased the number of platelets with a mitochondrial membrane potential loss. On the 3rd day of storage, 50% of Mirasol-treated platelets did not respond to activation; on the 5th day, none did. This agreed well with a decrease (approximately twofold) in the effectiveness of Mirasol-treated PC transfusions. Transfusions of PCs stored in SSP+ were accompanied by fewer inefficient transfusions and post-transfusion reactions than of PCs stored in plasma. CONCLUSION: Treatment with Mirasol decreased platelet function, particularly profoundly on the 5th day of storage, and led to a decrease in the effectiveness of transfusions. SSP+ did not affect laboratory parameters significantly compared with plasma, but decreased the percentage of transfusion complications.


Asunto(s)
Plaquetas/citología , Plaquetas/efectos de los fármacos , Riboflavina/farmacología , Rayos Ultravioleta , Plaquetas/efectos de la radiación , Conservación de la Sangre , Citometría de Flujo , Humanos , Potencial de la Membrana Mitocondrial , Selectina-P/sangre , Fosfatidilserinas/sangre , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/efectos de la radiación , Recuento de Plaquetas , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/análisis , Transfusión de Plaquetas , Plaquetoferesis
12.
J Math Biol ; 72(3): 649-81, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26001742

RESUMEN

The paper is devoted to mathematical modelling of clot growth in blood flow. Great complexity of the hemostatic system dictates the need of usage of the mathematical models to understand its functioning in the normal and especially in pathological situations. In this work we investigate the interaction of blood flow, platelet aggregation and plasma coagulation. We develop a hybrid DPD-PDE model where dissipative particle dynamics (DPD) is used to model plasma flow and platelets, while the regulatory network of plasma coagulation is described by a system of partial differential equations. Modelling results confirm the potency of the scenario of clot growth where at the first stage of clot formation platelets form an aggregate due to weak inter-platelet connections and then due to their activation. This enables the formation of the fibrin net in the centre of the platelet aggregate where the flow velocity is significantly reduced. The fibrin net reinforces the clot and allows its further growth. When the clot becomes sufficiently large, it stops growing due to the narrowed vessel and the increase of flow shear rate at the surface of the clot. Its outer part is detached by the flow revealing the inner part covered by fibrin. This fibrin cap does not allow new platelets to attach at the high shear rate, and the clot stops growing. Dependence of the final clot size on wall shear rate and on other parameters is studied.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/fisiología , Fibrina/fisiología , Modelos Biológicos , Animales , Biología Computacional , Hemorreología , Hemostasis/fisiología , Humanos , Conceptos Matemáticos , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología
13.
Biochim Biophys Acta ; 1850(12): 2518-29, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26391841

RESUMEN

BACKGROUND: Two major soluble blood platelet activators are thrombin and ADP. Of these two, only thrombin can induce mitochondrial collapse and programmed cell death leading to phosphatidylserine (PS) exposure required for blood clotting reactions acceleration. Thrombin can also greatly potentiate collagen-induced PS exposure. However, ADP acting through the P2Y12 receptor was shown to increase the PS-exposing (PS+) platelets fraction produced by thrombin or thrombin-plus-collagen via an unknown mechanism. METHODS: We developed a comprehensive multicompartmental computational model of platelet PAR1-and-P2Y12 calcium signal transduction that included cytoplasmic signaling, dense tubular system and mitochondria. To test model predictions, flow cytometry experiments with washed, annexin V-labeled platelets were performed. RESULTS: Stimulation of thrombin receptor PAR1 in the model induced cytoplasmic calcium oscillations, calcium uptake by mitochondria, opening of the permeability transition pore and collapse of the mitochondrial membrane potential. ADP stimulation of P2Y12 led to cAMP decrease that, in turn, caused changes in phospholipase C phosphorylation by protein kinase A, increase in cytoplasmic calcium level and, consequently, PS+ platelet formation. ADP addition before stimulation of PAR1 produced much greater increase of the PS+ fraction because cAMP concentration had time to go down prior to calcium oscillations; this prediction was also tested and confirmed experimentally. CONCLUSION: These results suggest a mechanism of ADP-dependent PS exposure regulation and show a likely mode of action that could be important for the PS exposure regulation in thrombi, where ADP is released before thrombin formation.


Asunto(s)
Plaquetas/citología , Receptor PAR-1/metabolismo , Receptores Purinérgicos P2Y12/fisiología , Transducción de Señal , Adenosina Difosfato/metabolismo , Calcio/metabolismo , Señalización del Calcio , Citosol/metabolismo , Humanos
14.
Usp Fiziol Nauk ; 46(4): 3-14, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-27183780

RESUMEN

All major coagulation reactions do not occurs in blood plasma itself, these processes are actually two-dimensional reactions localized to thephospholipid membranes. Almost all blood cells, lipoproteins, and microparticles provide assembly of protein complexes. A central role among them are played by platelets and platelet-derived microparticles. On their membranes occurs the most important coagulation reactions such as activation of prothrombin by prothrombin complex, activation of factor X by complexes intrinsic and extrinsic tenase. This reactions are important for processes activation of the contact path coagulation, activation factor XI by thrombin, appearance of enzymatic activity of factor VIIa etc. This review is focused on the membrane-dependent reactions, here are discussed mechanisms and regulation these reactions and the possible prospects of the study.


Asunto(s)
Coagulación Sanguínea , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Animales , Factores de Coagulación Sanguínea/metabolismo , Plaquetas/metabolismo , Humanos
15.
Biomed Khim ; 60(2): 182-200, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-24837309

RESUMEN

Platelet participation in hemostatic plug formation requires transition into an activated state (or, rather, variety of states) upon action of agonists like ADP, thromboxane A , collagen, thrombin, and others. The mechanisms of action for different agonists, their receptors and signaling pathways associated with them, as well as the mechanisms of platelet response inhibition are the subject of the present review. Collagen exposed upon vessel wall damage induced initial platelet attachment and start of thrombus formation, which involves numerous processes such as aggregation, activation of integrins, granule secretion and increase of intracellular Ca2+. Thrombin, ADP, thromboxane A , and ATP activated platelets that were not initially in contact with the wall and induce additional secretion of activating substances. Vascular endothelium and secretory organs also affect platelet activation, producing both positive (adrenaline) an d negative (prostacyclin, nitric oxide) regulators, thereby determining the relation of activation and inhibition signals, which plays a significant role in the formation of platelet aggregate under normal and pathological conditions. The pathways of platelet signaling are still incompletely understood, and their exploration presents an important objective both for basic cell biology and for the development of new drugs, the methods of diagnostics and of treatment of hemostasis disorders.


Asunto(s)
Plaquetas/fisiología , Activación Plaquetaria/fisiología , Transducción de Señal/fisiología , Adenosina Difosfato/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Colágeno/metabolismo , Humanos , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombina/metabolismo , Tromboxano A2/metabolismo
16.
J Theor Biol ; 337: 30-41, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23916879

RESUMEN

Hemostatic plug covering the injury site (or a thrombus in the pathological case) is formed due to the complex interaction of aggregating platelets with biochemical reactions in plasma that participate in blood coagulation. The mechanisms that control clot growth and which lead to growth arrest are not yet completely understood. We model them with numerical simulations based on a hybrid DPD-PDE model. Dissipative particle dynamics (DPD) is used to model plasma flow with platelets while fibrin concentration is described by a simplified reaction-diffusion-advection equation. The model takes into account consecutive stages of clot growth. First, a platelet is weakly connected to the clot and after some time this connection becomes stronger due to other surface receptors involved in platelet adhesion. At the same time, the fibrin mesh is formed inside the clot. This becomes possible because flow does not penetrate the clot and cannot wash out the reactants participating in blood coagulation. Platelets covered by the fibrin mesh cannot attach new platelets. Modelling shows that the growth of a hemostatic plug can stop as a result of its exterior part being removed by the flow thus exposing its non-adhesive core to the flow.


Asunto(s)
Hemorreología , Modelos Biológicos , Trombosis/patología , Coagulación Sanguínea , Plaquetas/metabolismo , Simulación por Computador , Fibrina/metabolismo , Humanos , Trombosis/metabolismo
17.
Biophys J ; 103(10): 2233-40, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23200057

RESUMEN

Injury-induced bleeding is stopped by a hemostatic plug formation that is controlled by a complex nonlinear and spatially heterogeneous biochemical network of proteolytic enzymes called blood coagulation. We studied spatial dynamics of thrombin, the central enzyme of this network, by developing a fluorogenic substrate-based method for time- and space-resolved imaging of thrombin enzymatic activity. Clotting stimulation by immobilized tissue factor induced localized thrombin activity impulse that propagated in space and possessed all characteristic traits of a traveling excitation wave: constant spatial velocity, constant amplitude, and insensitivity to the initial stimulation once it exceeded activation threshold. The parameters of this traveling wave were controlled by the availability of phospholipids or platelets, and the wave did not form in plasmas from hemophilia A or C patients who lack factors VIII and XI, which are mediators of the two principal positive feedbacks of coagulation. Stimulation of the negative feedback of the protein C pathway with thrombomodulin produced nonstationary patterns of wave formation followed by deceleration and annihilation. This indicates that blood can function as an excitable medium that conducts traveling waves of coagulation.


Asunto(s)
Coagulación Sanguínea , Trombina/metabolismo , Animales , Fenómenos Biomecánicos , Retroalimentación Fisiológica , Fibrina/metabolismo , Hemostasis , Humanos , Proteína C/metabolismo , Conejos , Trombomodulina/metabolismo
18.
Biophys J ; 101(8): 1816-24, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22004734

RESUMEN

Blood coagulation is triggered not only by surface tissue factor (TF) density but also by surface TF distribution. We investigated recognition of surface TF distribution patterns during blood coagulation and identified the underlying molecular mechanisms. For these investigations, we employed 1), an in vitro reaction-diffusion experimental model of coagulation; and 2), numerical simulations using a mathematical model of coagulation in a three-dimensional space. When TF was uniformly immobilized over the activating surface, the clotting initiation time in normal plasma increased from 4 min to >120 min, with a decrease in TF density from 100 to 0.7 pmol/m(2). In contrast, surface-immobilized fibroblasts initiated clotting within 3-7 min, independently of fibroblast quantity and despite a change in average surface TF density from 0.5 to 130 pmol/m(2). Experiments using factor V-, VII-, and VIII-deficient plasma and computer simulations demonstrated that different responses to these two TF distributions are caused by two positive feedback loops in the blood coagulation network: activation of the TF-VII complex by factor Xa, and activation of factor V by thrombin. This finding suggests a new role for these reactions: to supply sensitivity to local TF density during blood coagulation.


Asunto(s)
Coagulación Sanguínea , Factor VII/metabolismo , Factor V/metabolismo , Retroalimentación Fisiológica , Modelos Biológicos , Difusión , Factor Xa/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Cinética , Lípidos/sangre , Trombina/metabolismo
19.
J Thromb Haemost ; 9(9): 1825-34, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21696535

RESUMEN

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is a major regulator of clotting initiation and a promising target for pro- and anticoagulation therapy. The aptamer BAX499 (formerly ARC19499) is a high-affinity specific TFPI antagonist designed to improve hemostasis. However, it is not clear how stimulation of coagulation onset by inactivating TFPI will affect spatial and temporal clot propagation. OBJECTIVE: To examine the BAX499 effect on clotting in a spatial, reaction-diffusion experimental system in comparison with that of recombinant activated factor VII (rVIIa). METHODS: Clotting in plasma activated by immobilized tissue factor (TF) was monitored by videomicroscopy. RESULTS: BAX499 dose-dependently improved coagulation in normal and hemophilia A plasma activated with TF at 2 pmole m(-2) by shortening lag time and increasing clot size by up to ~2-fold. The effect was TFPI specific as confirmed by experiments in TFPI-depleted plasma with or without TFPI supplementation. Clotting improvement was half-maximal at 0.7 nm of BAX499 and reached a plateau at 10 nm, remaining there at concentrations up to 1000 nm. The BAX499 effect decreased with TF surface density increase. RVIIa improved clotting in hemophilia A plasma activated with TF at 2 or 20 pmole m(-2) , both by shortening lag time and increasing spatial velocity of clot propagation; its effects were strongly concentration dependent. CONCLUSIONS: BAX499 significantly improves spatial coagulation by inhibiting TFPI in a spatially localized manner that is different to that observed with rVIIa.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Coagulación Sanguínea/efectos de los fármacos , Fibrina/biosíntesis , Lipoproteínas/antagonistas & inhibidores , Aptámeros de Nucleótidos/administración & dosificación , Coagulación Sanguínea/fisiología , Simulación por Computador , Factor VIIa/administración & dosificación , Factor VIIa/farmacología , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Hemostasis/efectos de los fármacos , Hemostasis/fisiología , Humanos , Técnicas In Vitro , Lipoproteínas/deficiencia , Lipoproteínas/fisiología , Masculino , Microscopía por Video , Modelos Biológicos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología
20.
Biochemistry (Mosc) ; 75(6): 734-43, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20636265

RESUMEN

A method for transmembrane protein thromboplastin (tissue factor) immobilization on polystyrene surface is described. Tissue factor is the main activating factor launching the blood coagulation process. It is a cofactor of factor VIIa, the first protease in the cascade of coagulation reactions. The proposed method preserves kinetic characteristics specific for native tissue factor on the fibroblast surface. The kinetics of binding to factor VIIa and enzymic activity of the formed complex follow Michaelis-Menten kinetics, which is also characteristic of native complex. A small difference is that dissociation constant for tissue factor immobilized on polystyrene surface exceeds 2.7-fold that for native factor. The proposed technique of immobilization provides for protein density on the activating surface corresponding to the tissue factor density on the fibroblast surface. The immobilized tissue factor can be used to activate blood coagulation in methods simulating spatial dynamics of in vitro clot growth. Investigation in this direction will make it possible to register both hypo- and hypercoagulation states of the system. This approach is advantageous over traditional methods of estimation of the coagulation system conditions, which mainly register only hypocoagulation. Investigation of the storage time has shown that activators containing immobilized tissue factor can be stored and used during for at least 100 days in the method studying spatial dynamics of fibrin clot formation.


Asunto(s)
Coagulación Sanguínea , Poliestirenos/química , Tromboplastina/metabolismo , Factores de Coagulación Sanguínea/metabolismo , Factor VIIa/metabolismo , Fibrina/metabolismo , Fibroblastos/metabolismo , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Tromboplastina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA