Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biophys J ; 122(4): 697-712, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36635963

RESUMEN

During clotting under flow, thrombin rapidly generates fibrin, whereas fibrin potently sequesters thrombin. This co-regulation was studied using microfluidic whole blood clotting on collagen/tissue factor, followed by buffer wash, and a start/stop cycling flow assay using the thrombin fluorogenic substrate, Boc-Val-Pro-Arg-AMC. After 3 min of clotting (100 s-1) and 5 min of buffer wash, non-elutable thrombin activity was easily detected during cycles of flow cessation. Non-elutable thrombin was similarly detected in plasma clots or arterial whole blood clots (1000 s-1). This thrombin activity was ablated by Phe-Pro-Arg-chloromethylketone (PPACK), apixaban, or Gly-Pro-Arg-Pro to inhibit fibrin. Reaction-diffusion simulations predicted 108 nM thrombin within the clot. Heparin addition to the start/stop assay had little effect on fibrin-bound thrombin, whereas addition of heparin-antithrombin (AT) required over 6 min to inhibit the thrombin, indicating a substantial diffusion limitation. In contrast, heparin-AT rapidly inhibited thrombin within microfluidic plasma clots, indicating marked differences in fibrin structure and functionality between plasma clots and whole blood clots. Addition of GPVI-Fab to blood before venous or arterial clotting (200 or 1000 s-1) markedly reduced fibrin-bound thrombin, whereas GPVI-Fab addition after 90 s of clotting had no effect. Perfusion of AF647-fibrinogen over washed fluorescein isothiocyanate (FITC)-fibrin clots resulted in an intense red layer around, but not within, the original FITC-fibrin. Similarly, introduction of plasma/AF647-fibrinogen generated substantial red fibrin masses that did not penetrate the original green clots, demonstrating that fibrin cannot be re-clotted with fibrinogen. Overall, thrombin within fibrin is non-elutable, easily accessed by peptides, slowly accessed by average-sized proteins (heparin/AT), and not accessible to fresh fibrinogen.


Asunto(s)
Fibrina , Trombina , Trombosis , Humanos , Fibrina/química , Fibrinógeno/metabolismo , Fluoresceína-5-Isotiocianato , Heparina , Microfluídica/métodos , Trombina/química , Trombosis/metabolismo , Glicoproteínas de Membrana Plaquetaria/química
2.
PLoS Comput Biol ; 18(1): e1009850, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35089923

RESUMEN

Modeling thrombus growth in pathological flows allows evaluation of risk under patient-specific pharmacological, hematological, and hemodynamical conditions. We have developed a 3D multiscale framework for the prediction of thrombus growth under flow on a spatially resolved surface presenting collagen and tissue factor (TF). The multiscale framework is composed of four coupled modules: a Neural Network (NN) that accounts for platelet signaling, a Lattice Kinetic Monte Carlo (LKMC) simulation for tracking platelet positions, a Finite Volume Method (FVM) simulator for solving convection-diffusion-reaction equations describing agonist release and transport, and a Lattice Boltzmann (LB) flow solver for computing the blood flow field over the growing thrombus. A reduced model of the coagulation cascade was embedded into the framework to account for TF-driven thrombin production. The 3D model was first tested against in vitro microfluidics experiments of whole blood perfusion with various antiplatelet agents targeting COX-1, P2Y1, or the IP receptor. The model was able to accurately capture the evolution and morphology of the growing thrombus. Certain problems of 2D models for thrombus growth (artifactual dendritic growth) were naturally avoided with realistic trajectories of platelets in 3D flow. The generalizability of the 3D multiscale solver enabled simulations of important clinical situations, such as cylindrical blood vessels and acute flow narrowing (stenosis). Enhanced platelet-platelet bonding at pathologically high shear rates (e.g., von Willebrand factor unfolding) was required for accurately describing thrombus growth in stenotic flows. Overall, the approach allows consideration of patient-specific platelet signaling and vascular geometry for the prediction of thrombotic episodes.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas , Modelos Biológicos , Trombosis/metabolismo , Animales , Plaquetas/citología , Plaquetas/fisiología , Biología Computacional , Ratones , Agregación Plaquetaria/fisiología , RNA-Seq , Análisis de la Célula Individual
3.
Biophys J ; 120(11): 2102-2111, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33838138

RESUMEN

Many biologically important cell binding processes, such as the rolling of leukocytes in the vasculature, are multivalent, being mediated by large numbers of weak binding ligands. Quantitative agreement between experiments and models of rolling has been elusive and often limited by the poor understanding of the binding and unbinding kinetics of the ligands involved. Here, we present a cell-free experimental model for such rolling, consisting of polymer microspheres whose adhesion to a glass surface is mediated by ligands with well-understood force-dependent binding free energy-short complementary DNA strands. We observe robust rolling activity for certain values of the shear rate and the grafted DNA strands' binding free energy and force sensitivity. The simulation framework developed to model leukocyte rolling, adhesive dynamics, quantitatively captures the mean rolling velocity and lateral diffusivity of the experimental particles using known values of the experimental parameters. Moreover, our model captures the velocity variations seen within the trajectories of single particles. Particle-to-particle variations can be attributed to small, plausible differences in particle characteristics. Overall, our findings confirm that state-of-the-art adhesive dynamics simulations are able to capture the complex physics of particle rolling, boding well for their extension to modeling more complex systems of rolling cells.


Asunto(s)
Adhesivos , Rodamiento de Leucocito , Adhesión Celular , ADN , Leucocitos , Microesferas
4.
Br J Haematol ; 193(5): 976-987, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33973229

RESUMEN

The development of anti-drug antibodies (ADAs) is a serious outcome of treatment strategies involving biological medicines. Coagulation factor VIII (FVIII) is used to treat haemophilia A patients, but its immunogenicity precludes a third of severe haemophiliac patients from receiving this treatment. The availability of patient-derived anti-drug antibodies can help us better understand drug immunogenicity and identify ways to overcome it. Thus, there were two aims to this work: (i) to develop and characterise a panel of recombinant, patient-derived, monoclonal antibodies covering a range of FVIII epitopes with varying potencies, kinetics and mechanism of action, and (ii) to demonstrate their applicability to assay development, evaluation of FVIII molecules and basic research. For the first objective we used recombinant antibodies to develop a rapid, sensitive, flexible and reproducible ex vivo assay that recapitulates inhibitor patient blood using blood from healthy volunteers. We also demonstrate how the panel can provide important information about the efficacy of FVIII products and reagents without the need for patient or animal material. These materials can be used as experimental exemplars or controls, as well as tools for rational, hypothesis-driven research and assay development in relation to FVIII immunogenicity and FVIII-related products.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Inhibidores de Factor de Coagulación Sanguínea/química , Factor VIII/química , Hemofilia A/sangre , Anticuerpos Monoclonales/sangre , Anticuerpos Neutralizantes/sangre , Inhibidores de Factor de Coagulación Sanguínea/sangre , Humanos , Proteínas Recombinantes/química
5.
PLoS Comput Biol ; 15(8): e1007266, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31381558

RESUMEN

During thrombosis, thrombin generates fibrin, however fibrin reversibly binds thrombin with low affinity E-domain sites (KD = 2.8 µM) and high affinity γ'-fibrin sites (KD = 0.1 µM). For blood clotting on collagen/tissue factor (1 TF-molecule/µm2) at 200 s-1 wall shear rate, high µM-levels of intraclot thrombin suggest robust prothrombin penetration into clots. Setting intraclot zymogen concentrations to plasma levels (and neglecting cofactor rate limitations) allowed the linearization of 7 Michaelis-Menton reactions between 6 species to simulate intraclot generation of: Factors FXa (via TF/VIIa or FIXa), FIXa (via TF/FVIIa or FXIa), thrombin, fibrin, and FXIa. This reduced model [7 rates, 2 KD's, enzyme half-lives~1 min] predicted the measured clot elution rate of thrombin-antithrombin (TAT) and fragment F1.2 in the presence and absence of the fibrin inhibitor Gly-Pro-Arg-Pro. To predict intraclot fibrin reaching 30 mg/mL by 15 min, the model required fibrinogen penetration into the clot to be strongly diffusion-limited (actual rate/ideal rate = 0.05). The model required free thrombin in the clot (~100 nM) to have an elution half-life of ~2 sec, consistent with measured albumin elution, with most thrombin (>99%) being fibrin-bound. Thrombin-feedback activation of FXIa became prominent and reached 5 pM FXIa at >500 sec in the simulation, consistent with anti-FXIa experiments. In predicting intrathrombus thrombin and fibrin during 15-min microfluidic experiments, the model revealed "cascade amplification" from 30 pM levels of intrinsic tenase to 15 nM prothrombinase to 15 µM thrombin to 90 µM fibrin. Especially useful for multiscale simulation, this reduced model predicts thrombin and fibrin co-regulation during thrombosis under flow.


Asunto(s)
Coagulación Sanguínea/fisiología , Modelos Biológicos , Trombosis/sangre , Plaquetas/metabolismo , Colágeno/sangre , Biología Computacional , Simulación por Computador , Cisteína Endopeptidasas/sangre , Factor XIa/metabolismo , Fibrina/metabolismo , Humanos , Técnicas In Vitro , Cinética , Proteínas de Neoplasias/sangre , Flujo Sanguíneo Regional/fisiología , Trombina/metabolismo , Tromboplastina/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 317(1): H73-H86, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30978134

RESUMEN

Quantifying the relationship between vascular injury and the dynamic bleeding rate requires a multiscale model that accounts for changing and coupled hemodynamics between the global and microvascular levels. A lumped, global hemodynamic model of the human cardiovascular system with baroreflex control was coupled to a local 24-level bifurcating vascular network that spanned diameters from the muscular artery scale (0.1-1.3 mm) to capillaries (5-10 µm) via conservation of momentum and conservation of mass boundary conditions. For defined injuries of severing all vessels at each nth-level, the changing pressures and flowrates were calculated using prescribed shear-dependent hemostatic clot growth rates (normal or coagulopathic). Key results were as follows: 1) the upstream vascular network rapidly depressurizes to reduce blood loss; 2) wall shear rates at the hemorrhaging wound exit are sufficiently high (~10,000 s-1) to drive von Willebrand factor unfolding; 3) full coagulopathy results in >2-liter blood loss in 2 h for severing all vessels of 0.13- to 0.005-mm diameter within the bifurcating network, whereas full hemostasis limits blood loss to <100 ml within 2 min; and 4) hemodilution from transcapillary refill increases blood loss and could be implicated in trauma-induced coagulopathy. A sensitivity analysis on length-to-diameter ratio and branching exponent demonstrated that bleeding was strongly dependent on these tissue-dependent network parameters. This is the first bleeding model that prescribes the geometry of the injury to calculate the rate of pressure-driven blood loss and local wall shear rate in the presence or absence of coagulopathic blood. NEW & NOTEWORTHY We developed a multiscale model that couples a lumped, global hemodynamic model of a patient to resolved, single-vessel wounds ranging from the small artery to capillary scale. The model is able to quantify wall shear rates, seal rates, and blood loss rates in the presence and absence of baroreflex control and hemodilution.


Asunto(s)
Coagulación Sanguínea , Sistema Cardiovascular/fisiopatología , Simulación por Computador , Hemodinámica , Hemorragia/sangre , Hemorragia/fisiopatología , Microcirculación , Modelos Cardiovasculares , Barorreflejo , Humanos
7.
Arterioscler Thromb Vasc Biol ; 38(7): 1528-1536, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29724819

RESUMEN

OBJECTIVE: We investigated the coregulation of thrombin and fibrin as blood flows over a procoagulant surface. APPROACH AND RESULTS: Using microfluidic perfusion of factor XIIa-inhibited human whole blood (200 s-1 wall shear rate) over a 250-µm long patch of collagen/TF (tissue factor; ≈1 molecule per µm2) and immunoassays of the effluent for F1.2 (prothrombin fragment 1.2), TAT (thrombin-antithrombin complex), and D-dimer (post-end point plasmin digest), we sought to establish the transient mass balance for clotting under venous flow. F1.2 (but almost no free thrombin detected via TAT assay) continually eluted from clots when fibrin was allowed to form. Low-dose fluorescein-Phe-Pro-Arg-chloromethylketone stained fibrin-bound thrombin-a staining ablated by anti-γ'-fibrinogen or the fibrin inhibitor glypro-arg-pro but highly resistant to 7-minute buffer rinse, demonstrating tight binding of thrombin to γ'-fibrin. With fibrin polymerizing for 500 seconds, 92 000 thrombin molecules and 203 000 clot-associated fibrin monomer equivalents were generated per TF molecule (or per µm2). Fibrin reached 15 mg/mL in the pore space (porosity ≈0.5) of a 15-µm-thick thrombus core by 500 seconds and 30 mg/mL by 800 seconds. For a known rate of ≈60 FPA (fibrinopeptide-A) per thrombin per second, each thrombin molecule generated only 3 fibrin monomer equivalents during 500 seconds, indicating an intraclot thrombin half-life of ≈70 ms, much shorter than its diffusional escape time (≈10 seconds). By 800 seconds, gly-pro-arg-pro allowed 4-fold more F1.2 generation, consistent with gly-pro-arg-pro ablating fibrin's antithrombin-I activity and facilitating thrombin-mediated FXIa activation. CONCLUSIONS: Under flow, fibrinogen continually penetrates the clot, and γ'-fibrin regulates thrombin.


Asunto(s)
Coagulación Sanguínea , Fibrina/metabolismo , Hemodinámica , Trombina/metabolismo , Tromboplastina/metabolismo , Trombosis/sangre , Antitrombina III , Velocidad del Flujo Sanguíneo , Factor XIIa/metabolismo , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Fibrinógenos Anormales/metabolismo , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Fragmentos de Péptidos/sangre , Péptido Hidrolasas/sangre , Porosidad , Protrombina , Trombosis/fisiopatología
8.
J Biomech Eng ; 141(3)2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30516240

RESUMEN

The efficacy of reduced order modeling for transstenotic pressure drop in the coronary arteries is presented. Coronary artery disease is a leading cause of death worldwide and the computation of pressure drop in the coronary arteries has become a standard for evaluating the functional significance of a coronary stenosis. Comprehensive models typically employ three-dimensional (3D) computational fluid dynamics (CFD) to simulate coronary blood flow in order to compute transstenotic pressure drop at the arterial stenosis. In this study, we evaluate the capability of different hydrodynamic models to compute transstenotic pressure drop. Models range from algebraic formulae to one-dimensional (1D), two-dimensional (2D), and 3D time-dependent CFD simulations. Although several algebraic pressure-drop formulae have been proposed in the literature, these models were found to exhibit wide variation in predictions. Nonetheless, we demonstrate an algebraic formula that provides consistent predictions with 3D CFD results for various changes in stenosis severity, morphology, location, and flow rate. The accounting of viscous dissipation and flow separation were found to be significant contributions to accurate reduce order modeling of transstenotic coronary hemodynamics.

9.
Biophys J ; 114(4): 978-991, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29490257

RESUMEN

The structure and growth of a blood clot depend on the localization of tissue factor (TF), which can trigger clotting during the hemostatic process or promote thrombosis when exposed to blood under pathological conditions. We sought to understand how the growth, structure, and mechanical properties of clots under flow are shaped by the simultaneously varying TF surface density and its exposure area. We used an eight-channel microfluidic device equipped with a 20- or 100-µm-long collagen surface patterned with lipidated TF of surface densities ∼0.1 and ∼2 molecules/µm2. Human whole blood was perfused at venous shear, and clot growth was continually measured. Using our recently developed computational model of clot formation, we performed simulations to gain insights into the clot's structure and its resistance to blood flow. An increase in TF exposure area resulted not only in accelerated bulk platelet, thrombin, and fibrin accumulation, but also in increased height of the platelet mass and increased clot resistance to flow. Moreover, increasing the TF surface density or exposure area enhanced platelet deposition by approximately twofold, and thrombin and fibrin generation by greater than threefold, thereby increasing both clot size and its viscous resistance. Finally, TF effects on blood flow occlusion were more pronounced for the longer thrombogenic surface than for the shorter one. Our results suggest that TF surface density and its exposure area can independently enhance both the clot's occlusivity and its resistance to blood flow. These findings provide, to our knowledge, new insights into how TF affects thrombus growth in time and space under flow.


Asunto(s)
Plaquetas/metabolismo , Estrés Mecánico , Tromboplastina/metabolismo , Trombosis/fisiopatología , Venas/fisiopatología , Coagulación Sanguínea , Simulación por Computador , Fibrina/metabolismo , Humanos , Dispositivos Laboratorio en un Chip , Modelos Teóricos , Resistencia al Corte , Trombina/metabolismo
10.
Circulation ; 136(7): 646-660, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28487393

RESUMEN

BACKGROUND: Platelet cross-linking during arterial thrombosis involves von Willebrand Factor (VWF) multimers. Therefore, proteolysis of VWF appears promising to disaggregate platelet-rich thrombi and restore vessel patency in acute thrombotic disorders such as ischemic stroke, acute coronary syndrome, or acute limb ischemia. N-Acetylcysteine (NAC, a clinically approved mucolytic drug) can reduce intrachain disulfide bonds in large polymeric proteins. In the present study, we postulated that NAC might cleave the VWF multimers inside occlusive thrombi, thereby leading to their dissolution and arterial recanalization. METHODS: Experimental models of thrombotic stroke induced by either intra-arterial thrombin injection or ferric chloride application followed by measurement of cerebral blood flow using a combination of laser Doppler flowmetry and MRI were performed to uncover the effects of NAC on arterial thrombi. To investigate the effect of NAC on larger vessels, we also performed ferric chloride-induced carotid artery thrombosis. In vitro experiments were performed to study the molecular bases of NAC thrombolytic effect, including platelet aggregometry, platelet-rich thrombi lysis assays, thromboelastography (ROTEM), and high-shear VWF string formation using microfluidic devices. We also investigated the putative prohemorrhagic effect of NAC in a mouse model of intracranial hemorrhage induced by in situ collagenase type VII injection. RESULTS: We demonstrated that intravenous NAC administration promotes lysis of arterial thrombi that are resistant to conventional approaches such as recombinant tissue-type plasminogen activator, direct thrombin inhibitors, and antiplatelet treatments. Through in vitro and in vivo experiments, we provide evidence that the molecular target underlying the thrombolytic effects of NAC is principally the VWF that cross-link platelets in arterial thrombi. Coadministration of NAC and a nonpeptidic GpIIb/IIIa inhibitor further improved its thrombolytic efficacy, essentially by accelerating thrombus dissolution and preventing rethrombosis. Thus, in a new large-vessel thromboembolic stroke model in mice, this cotreatment significantly improved ischemic lesion size and neurological outcome. It is important to note that NAC did not worsen hemorrhagic stroke outcome, suggesting that it exerts thrombolytic effects without significantly impairing normal hemostasis. CONCLUSIONS: We provide evidence that NAC is an effective and safe alternative to currently available antithrombotic agents to restore vessel patency after arterial occlusion.


Asunto(s)
Acetilcisteína/uso terapéutico , Fibrinolíticos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Tromboembolia/tratamiento farmacológico , Acetilcisteína/farmacología , Animales , Plaquetas/citología , Plaquetas/metabolismo , Cloruros/toxicidad , Modelos Animales de Enfermedad , Compuestos Férricos/toxicidad , Fibrinolíticos/farmacología , Infarto de la Arteria Cerebral Media/etiología , Masculino , Ratones , Agregación Plaquetaria/efectos de los fármacos , Ristocetina/farmacología , Tromboembolia/inducido químicamente , Trombosis/prevención & control , Activador de Tejido Plasminógeno/uso terapéutico , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
11.
Blood ; 127(12): 1598-605, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26738537

RESUMEN

Previous studies have shown that hemostatic thrombi formed in response to penetrating injuries have a core of densely packed, fibrin-associated platelets overlaid by a shell of less-activated, loosely packed platelets. Here we asked, first, how the diverse elements of this structure combine to stem the loss of plasma-borne molecules and, second, whether antiplatelet agents and anticoagulants that perturb thrombus structure affect the re-establishment of a tight vascular seal. The studies combined high-resolution intravital microscopy with a photo-activatable fluorescent albumin marker to simultaneously track thrombus formation and protein transport following injuries to mouse cremaster muscle venules. The results show that protein loss persists after red cell loss has ceased. Blocking platelet deposition with an αIIbß3antagonist delays vessel sealing and increases extravascular protein accumulation, as does either inhibiting adenosine 5'-diphosphate (ADP) P2Y12receptors or reducing integrin-dependent signaling and retraction. In contrast, sealing was unaffected by introducing hirudin to block fibrin accumulation or a Gi2α gain-of-function mutation to expand the thrombus shell. Collectively, these observations describe a novel approach for studying vessel sealing after injury in real time in vivo and show that (1) the core/shell architecture previously observed in arterioles also occurs in venules, (2) plasma leakage persists well beyond red cell escape and mature thrombus formation, (3) the most critical events for limiting plasma extravasation are the stable accumulation of platelets, ADP-dependent signaling, and the emergence of a densely packed core, not the accumulation of fibrin, and (4) drugs that affect platelet accumulation and packing can delay vessel sealing, permitting protein escape to continue.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Hemostasis , Microvasos/lesiones , Microvasos/patología , Trombosis/patología , Adenosina Difosfato/metabolismo , Animales , Proteínas Sanguíneas/análisis , Fibrina/análisis , Fibrina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/metabolismo , Activación Plaquetaria , Recuento de Plaquetas , Trombosis/sangre , Trombosis/metabolismo
12.
Circ Res ; 118(9): 1348-62, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27126646

RESUMEN

The systems analysis of thrombosis seeks to quantitatively predict blood function in a given vascular wall and hemodynamic context. Relevant to both venous and arterial thrombosis, a Blood Systems Biology approach should provide metrics for rate and molecular mechanisms of clot growth, thrombotic risk, pharmacological response, and utility of new therapeutic targets. As a rapidly created multicellular aggregate with a polymerized fibrin matrix, blood clots result from hundreds of unique reactions within and around platelets propagating in space and time under hemodynamic conditions. Coronary artery thrombosis is dominated by atherosclerotic plaque rupture, complex pulsatile flows through stenotic regions producing high wall shear stresses, and plaque-derived tissue factor driving thrombin production. In contrast, venous thrombosis is dominated by stasis or depressed flows, endothelial inflammation, white blood cell-derived tissue factor, and ample red blood cell incorporation. By imaging vessels, patient-specific assessment using computational fluid dynamics provides an estimate of local hemodynamics and fractional flow reserve. High-dimensional ex vivo phenotyping of platelet and coagulation can now power multiscale computer simulations at the subcellular to cellular to whole vessel scale of heart attacks or strokes. In addition, an integrated systems biology approach can rank safety and efficacy metrics of various pharmacological interventions or clinical trial designs.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Simulación por Computador , Trombosis/sangre , Animales , Plaquetas/patología , Hemodinámica , Humanos , Microfluídica , Biología de Sistemas
13.
J Biol Chem ; 291(44): 23027-23035, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27605669

RESUMEN

Coagulation kinetics are well established for purified blood proteases or human plasma clotting isotropically. However, less is known about thrombin generation kinetics and transport within blood clots formed under hemodynamic flow. Using microfluidic perfusion (wall shear rate, 200 s-1) of corn trypsin inhibitor-treated whole blood over a 250-µm long patch of type I fibrillar collagen/lipidated tissue factor (TF; ∼1 TF molecule/µm2), we measured thrombin released from clots using thrombin-antithrombin immunoassay. The majority (>85%) of generated thrombin was captured by intrathrombus fibrin as thrombin-antithrombin was largely undetectable in the effluent unless Gly-Pro-Arg-Pro (GPRP) was added to block fibrin polymerization. With GPRP present, the flux of thrombin increased to ∼0.5 × 10-12 nmol/µm2-s over the first 500 s of perfusion and then further increased by ∼2-3-fold over the next 300 s. The increased thrombin flux after 500 s was blocked by anti-FXIa antibody (O1A6), consistent with thrombin-feedback activation of FXI. Over the first 500 s, ∼92,000 molecules of thrombin were generated per surface TF molecule for the 250-µm-long coating. A single layer of platelets (obtained with αIIbß3 antagonism preventing continued platelet deposition) was largely sufficient for thrombin production. Also, the overall thrombin-generating potential of a 1000-µm-long coating became less efficient on a per µm2 basis, likely due to distal boundary layer depletion of platelets. Overall, thrombin is robustly generated within clots by the extrinsic pathway followed by late-stage FXIa contributions, with fibrin localizing thrombin via its antithrombin-I activity as a potentially self-limiting hemostatic mechanism.


Asunto(s)
Colágeno/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo , Trombosis/enzimología , Coagulación Sanguínea , Plaquetas/química , Plaquetas/metabolismo , Colágeno/química , Colágeno/genética , Fibrina/química , Fibrina/metabolismo , Humanos , Cinética , Oligopéptidos/química , Oligopéptidos/metabolismo , Trombina/química , Trombina/genética , Tromboplastina/química , Tromboplastina/genética , Trombosis/genética , Trombosis/metabolismo
14.
Blood ; 126(12): 1494-502, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26136249

RESUMEN

Factor XIIa (FXIIa) and factor XIa (FXIa) contribute to thrombosis in animal models, whereas platelet-derived polyphosphate (polyP) may potentiate contact or thrombin-feedback pathways. The significance of these mediators in human blood under thrombotic flow conditions on tissue factor (TF) -bearing surfaces remains inadequately resolved. Human blood (corn trypsin inhibitor treated [4 µg/mL]) was tested by microfluidic assay for clotting on collagen/TF at TF surface concentration ([TF]wall) from ∼0.1 to 2 molecules per µm(2). Anti-FXI antibodies (14E11 and O1A6) or polyP-binding protein (PPXbd) were used to block FXIIa-dependent FXI activation, FXIa-dependent factor IX (FIX) activation, or platelet-derived polyP, respectively. Fibrin formation was sensitive to 14E11 at 0 to 0.1 molecules per µm(2) and sensitive to O1A6 at 0 to 0.2 molecules per µm(2). However, neither antibody reduced fibrin generation at ∼2 molecules per µm(2) when the extrinsic pathway became dominant. Interestingly, PPXbd reduced fibrin generation at low [TF]wall (0.1 molecules per µm(2)) but not at zero or high [TF]wall, suggesting a role for polyP distinct from FXIIa activation and requiring low extrinsic pathway participation. Regardless of [TF]wall, PPXbd enhanced fibrin sensitivity to tissue plasminogen activator and promoted clot retraction during fibrinolysis concomitant with an observed PPXbd-mediated reduction of fibrin fiber diameter. This is the first detection of endogenous polyP function in human blood under thrombotic flow conditions. When triggered by low [TF]wall, thrombosis may be druggable by contact pathway inhibition, although thrombolytic susceptibility may benefit from polyP antagonism regardless of [TF]wall.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Factor XIa/metabolismo , Polifosfatos/metabolismo , Tromboplastina/metabolismo , Anticuerpos/farmacología , Pruebas de Coagulación Sanguínea , Plaquetas/citología , Colágeno/metabolismo , Humanos , Terapia Molecular Dirigida , Transducción de Señal , Trombina/metabolismo , Trombosis/tratamiento farmacológico , Trombosis/metabolismo
15.
Platelets ; 28(5): 457-462, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28102731

RESUMEN

Platelets present a number of intracellular and transmembrane targets subject to pharmacological modulation, either for cardiovascular disease reduction or as an unintended drug response. Microfluidic devices allow human blood to clot on a defined surface under controlled hemodynamic and pharmacological conditions. The potencies of a number of antiplatelet and anticancer drugs have been tested with respect to platelet deposition on collagen under flow. Inhibitors of cyclooxygenase-1 (COX-1) reduce platelet deposition, either when added ex vivo to blood or ingested orally by patients prior to testing. Some individuals display a functional "aspirin-insensitivity" in microfluidic assay. When certain nonsteroidal anti-inflammatory drugs (NSAIDs) are taken orally, they block COX-1 acetylation by aspirin with concomitant reduction of aspirin efficacy against platelets in microfluidic assay. Both P2Y1 and P2Y12 inhibitors reduce platelet deposition under flow, as do NO donors and iloprost that target the guanylate cyclase and the prostacyclin receptor, respectively. In a microfluidic assay of 37 kinase inhibitors, dasatinib had potent antiplatelet activity, while bosutinib was less potent. Dasatinib and bosutinib have known profiles against numerous kinases, revealing overlapping and nonoverlapping activities relevant to their unique actions against platelets. Also, dasatinib caused a marked and specific inhibition of GPVI signaling induced by convulxin, consistent with a dasatinib-associated bleeding risk. Microfluidic devices facilitate drug library screening, dose-response testing, and drug-drug interaction studies. Kinase inhibitors developed as anticancer agents may present antiplatelet activities that are detectable by microfluidic assay and potentially linked to bleeding risks.


Asunto(s)
Plaquetas/metabolismo , Evaluación Preclínica de Medicamentos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/métodos , Preparaciones Farmacéuticas , Animales , Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Humanos
16.
Proc Natl Acad Sci U S A ; 111(47): 16830-5, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385584

RESUMEN

The cardiovascular safety of nonsteroidal antiinflammatory drugs (NSAIDs) may be influenced by interactions with antiplatelet doses of aspirin. We sought to quantitate precisely the propensity of commonly consumed NSAIDs­ibuprofen, naproxen, and celecoxib­to cause a drug-drug interaction with aspirin in vivo by measuring the target engagement of aspirin directly by MS. We developed a novel assay of cyclooxygenase-1 (COX-1) acetylation in platelets isolated from volunteers who were administered aspirin and used conventional and microfluidic assays to evaluate platelet function. Although ibuprofen, naproxen, and celecoxib all had the potential to compete with the access of aspirin to the substrate binding channel of COX-1 in vitro, exposure of volunteers to a single therapeutic dose of each NSAID followed by 325 mg aspirin revealed a potent drug-drug interaction between ibuprofen and aspirin and between naproxen and aspirin but not between celecoxib and aspirin. The imprecision of estimates of aspirin consumption and the differential impact on the ability of aspirin to inactivate platelet COX-1 will confound head-to-head comparisons of distinct NSAIDs in ongoing clinical studies designed to measure their cardiovascular risk.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Plaquetas/efectos de los fármacos , Ciclooxigenasa 1/metabolismo , Acetilación , Plaquetas/enzimología , Humanos , Microfluídica
17.
Blood ; 124(11): 1808-15, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24951424

RESUMEN

Hemostatic thrombi develop a characteristic architecture in which a core of highly activated platelets is covered by a shell of less-activated platelets. Here we have used a systems biology approach to examine the interrelationship of this architecture with transport rates and agonist distribution in the gaps between platelets. Studies were performed in mice using probes for platelet accumulation, packing density, and activation plus recently developed transport and thrombin activity probes. The results show that intrathrombus transport within the core is much slower than within the shell. The region of slowest transport coincides with the region of greatest packing density and thrombin activity, and appears prior to full platelet activation. Deleting the contact-dependent signaling molecule, Sema4D, delays platelet activation, but not the emergence of the low transport region. Collectively, these results suggest a timeline in which initial platelet accumulation and the narrowing gaps between platelets create a region of reduced transport that facilitates local thrombin accumulation and greater platelet activation, whereas faster transport rates within the shell help to limit thrombin accumulation and growth of the core. Thus, from a systems perspective, platelet accumulation produces an altered microenvironment that shapes thrombus architecture, which in turn affects agonist distribution and subsequent thrombus growth.


Asunto(s)
Coagulación Sanguínea , Modelos Cardiovasculares , Activación Plaquetaria , Trombina/metabolismo , Animales , Humanos , Ratones , Transporte de Proteínas
18.
Blood ; 124(11): 1816-23, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24951425

RESUMEN

Hemostatic thrombi formed after a penetrating injury have a heterogeneous architecture in which a core of highly activated, densely packed platelets is covered by a shell of less-activated, loosely packed platelets. In the first manuscript in this series, we show that regional differences in intrathrombus protein transport rates emerge early in the hemostatic response and are preserved as the thrombus develops. Here, we use a theoretical approach to investigate this process and its impact on agonist distribution. The results suggest that hindered diffusion, rather than convection, is the dominant mechanism responsible for molecular movement within the thrombus. The analysis also suggests that the thrombus core, as compared with the shell, provides an environment for retaining soluble agonists such as thrombin, affecting the extent of platelet activation by establishing agonist-specific concentration gradients radiating from the site of injury. This analysis accounts for the observed weaker activation and relative instability of platelets in the shell and predicts that a failure to form a tightly packed thrombus core will limit thrombin accumulation, a prediction tested by analysis of data from mice with a defect in clot retraction.


Asunto(s)
Coagulación Sanguínea , Simulación por Computador , Modelos Cardiovasculares , Activación Plaquetaria , Trombina/metabolismo , Animales , Humanos , Ratones , Transporte de Proteínas
19.
Blood ; 124(11): 1824-31, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24951426

RESUMEN

Hemostatic thrombi formed after a penetrating injury have a distinctive structure in which a core of highly activated, closely packed platelets is covered by a shell of less-activated, loosely packed platelets. We have shown that differences in intrathrombus molecular transport emerge in parallel with regional differences in platelet packing density and predicted that these differences affect thrombus growth and stability. Here we test that prediction in a mouse vascular injury model. The studies use a novel method for measuring thrombus contraction in vivo and a previously characterized mouse line with a defect in integrin αIIbß3 outside-in signaling that affects clot retraction ex vivo. The results show that the mutant mice have a defect in thrombus consolidation following vascular injury, resulting in an increase in intrathrombus transport rates and, as predicted by computational modeling, a decrease in thrombin activity and platelet activation in the thrombus core. Collectively, these data (1) demonstrate that in addition to the activation state of individual platelets, the physical properties of the accumulated mass of adherent platelets is critical in determining intrathrombus agonist distribution and platelet activation and (2) define a novel role for integrin signaling in the regulation of intrathrombus transport rates and localization of thrombin activity.


Asunto(s)
Coagulación Sanguínea , Modelos Cardiovasculares , Activación Plaquetaria , Trombina/metabolismo , Animales , Humanos , Ratones , Transporte de Proteínas
20.
PLoS Comput Biol ; 11(2): e1004118, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25723389

RESUMEN

Since platelet intracellular calcium mobilization [Ca(t)]i controls granule release, cyclooxygenase-1 and integrin activation, and phosphatidylserine exposure, blood clotting simulations require prediction of platelet [Ca(t)]i in response to combinatorial agonists. Pairwise Agonist Scanning (PAS) deployed all single and pairwise combinations of six agonists (ADP, convulxin, thrombin, U46619, iloprost and GSNO used at 0.1, 1, and 10xEC50; 154 conditions including a null condition) to stimulate platelet P2Y1/P2Y12 GPVI, PAR1/PAR4, TP, IP receptors, and guanylate cyclase, respectively, in Factor Xa-inhibited (250 nM apixaban), diluted platelet rich plasma that had been loaded with the calcium dye Fluo-4 NW. PAS of 10 healthy donors provided [Ca(t)]i data for training 10 neural networks (NN, 2-layer/12-nodes) per donor. Trinary stimulations were then conducted at all 0.1x and 1xEC50 doses (160 conditions) as was a sampling of 45 higher ordered combinations (four to six agonists). The NN-ensemble average was a calcium calculator that accurately predicted [Ca (t)]i beyond the single and binary training set for trinary stimulations (R = 0.924). The 160 trinary synergy scores, a normalized metric of signaling crosstalk, were also well predicted (R = 0.850) as were the calcium dynamics (R = 0.871) and high-dimensional synergy scores (R = 0.695) for the 45 higher ordered conditions. The calculator even predicted sequential addition experiments (n = 54 conditions, R = 0.921). NN-ensemble is a fast calcium calculator, ideal for multiscale clotting simulations that include spatiotemporal concentrations of ADP, collagen, thrombin, thromboxane, prostacyclin, and nitric oxide.


Asunto(s)
Factores de Coagulación Sanguínea/agonistas , Plaquetas/metabolismo , Calcio/análisis , Calcio/metabolismo , Activación Plaquetaria/efectos de los fármacos , Procesamiento de Señales Asistido por Computador , Biología Computacional , Humanos , Redes Neurales de la Computación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA